transforming-growth-factor-beta and Neoplasms

transforming-growth-factor-beta has been researched along with Neoplasms* in 1063 studies

Reviews

581 review(s) available for transforming-growth-factor-beta and Neoplasms

ArticleYear
TGFβ control of immune responses in cancer: a holistic immuno-oncology perspective.
    Nature reviews. Immunology, 2023, Volume: 23, Issue:6

    The immune system responds to cancer in two main ways. First, there are prewired responses involving myeloid cells, innate lymphocytes and innate-like adaptive lymphocytes that either reside in premalignant tissues or migrate directly to tumours, and second, there are antigen priming-dependent responses, in which adaptive lymphocytes are primed in secondary lymphoid organs before homing to tumours. Transforming growth factor-β (TGFβ) - one of the most potent and pleiotropic regulatory cytokines - controls almost every stage of the tumour-elicited immune response, from leukocyte development in primary lymphoid organs to their priming in secondary lymphoid organs and their effector functions in the tumour itself. The complexity of TGFβ-regulated immune cell circuitries, as well as the contextual roles of TGFβ signalling in cancer cells and tumour stromal cells, necessitates the use of rigorous experimental systems that closely recapitulate human cancer, such as autochthonous tumour models, to uncover the underlying immunobiology. The diverse functions of TGFβ in healthy tissues further complicate the search for effective and safe cancer therapeutics targeting the TGFβ pathway. Here we discuss the contextual complexity of TGFβ signalling in tumour-elicited immune responses and explain how understanding this may guide the development of mechanism-based cancer immunotherapy.

    Topics: Cytokines; Humans; Immunity; Immunotherapy; Neoplasms; Transforming Growth Factor beta

2023
Bone morphogenetic protein signaling is a possible therapeutic target in gynecologic cancer.
    Cancer science, 2023, Volume: 114, Issue:3

    Bone morphogenetic proteins (BMPs) belong to the transforming growth factor β (TGFβ) superfamily. BMPs play crucial roles in embryogenesis and bone remodeling. Recently, BMP signaling has been found to have diverse effects on different types of tumors. In this review, we summarized the effects of BMP signaling on gynecologic cancer. BMP signaling has tumor-promoting effects on ovarian cancer (OC) and endometrial cancer (EC), whereas it has tumor-suppressing effects on uterine cervical cancer (UCC). Interestingly, EC has frequent gain-of-function mutations in ACVR1, encoding one of the type I BMP receptors, which are also observed in fibrodysplasia ossificans progressiva and diffuse intrinsic pontine glioma. Little is known about the relationship between BMP signaling and other gynecologic cancers. Tumor-promoting effects of BMP signaling in OC and EC are dependent on the promotion of cancer stemness and epithelial-mesenchymal transition (EMT). In accordance, BMP receptor kinase inhibitors suppress the cell growth and migration of OC and EC. Since both cancer stemness and EMT are associated with chemoresistance, BMP signaling activation might also be an important mechanism by which OC and EC patients acquire chemoresistance. Therefore, BMP inhibitors are promising for OC and EC patients even if they become resistant to standard chemotherapy. In contrast, BMP signaling inhibits UCC growth in vitro. However, the in vivo effects of BMP signaling have not been elucidated in UCC. In conclusion, BMP signaling has a variety of functions, depending on the types of gynecologic cancer. Therefore, targeting BMP signaling should improve the treatment of patients with gynecologic cancer.

    Topics: Bone Morphogenetic Proteins; Epithelial-Mesenchymal Transition; Female; Humans; Myositis Ossificans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2023
Transforming growth factor-β signalling in tumour resistance to the anti-PD-(L)1 therapy: Updated.
    Journal of cellular and molecular medicine, 2023, Volume: 27, Issue:3

    Low frequency of durable responses in patients treated with immune checkpoint inhibitors (ICIs) demands for taking complementary strategies in order to boost immune responses against cancer. Transforming growth factor-β (TGF-β) is a multi-tasking cytokine that is frequently expressed in tumours and acts as a critical promoter of tumour hallmarks. TGF-β promotes an immunosuppressive tumour microenvironment (TME) and defines a bypass mechanism to the ICI therapy. A number of cells within the stroma of tumour are influenced from TGF-β activity. There is also evidence of a relation between TGF-β with programmed death-ligand 1 (PD-L1) expression within TME, and it influences the efficacy of anti-programmed death-1 receptor (PD-1) or anti-PD-L1 therapy. Combination of TGF-β inhibitors with anti-PD(L)1 has come to the promising outcomes, and clinical trials are under way in order to use agents with bifunctional capacity and fusion proteins for bonding TGF-β traps with anti-PD-L1 antibodies aiming at reinvigorating immune responses and promoting persistent responses against advanced stage cancers, especially tumours with immunologically cold ecosystem.

    Topics: B7-H1 Antigen; Ecosystem; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factors; Tumor Microenvironment

2023
Efficacy of therapies targeting TGF-β in solid tumors: a systematic review and meta-analysis of clinical trials.
    Immunotherapy, 2023, Volume: 15, Issue:4

    Cancer is the second leading cause of death in the world after cardiovascular diseases. Metastasis has a vital role in mortality rate of cancer patients. TGF-β, which regulates cell proliferation and invasion, is a key regulator of this process, in which activation of TGF-β is related to poor prognosis in cancer patients. Although several studies have shown therapeutic effects of inhibition of TGF-β in animal models and human clinical trials, a comprehensive report of the clinical effects, patient responsiveness and safety of TGF-β inhibitors in cancer patients would be of note. This study aims to investigate and analyze reported clinical outcomes after administration of TGF-β inhibitors in various cancers. The results of this study will be helpful for the study of dosages and sequencing of therapies in future combinatorial immunotherapy.

    Topics: Humans; Immunotherapy; Neoplasms; Transforming Growth Factor beta

2023
TGF-β signaling pathway: Therapeutic targeting and potential for anti-cancer immunity.
    European journal of pharmacology, 2023, May-15, Volume: 947

    Transforming growth factor-β (TGFβ) is a pleiotropic secretory cytokine exhibiting both cancer-inhibitory and promoting properties. It transmits its signals via Suppressor of Mother against Decapentaplegic (SMAD) and non-SMAD pathways and regulates cell proliferation, differentiation, invasion, migration, and apoptosis. In non-cancer and early-stage cancer cells, TGFβ signaling suppresses cancer progression via inducing apoptosis, cell cycle arrest, or anti-proliferation, and promoting cell differentiation. On the other hand, TGFβ may also act as an oncogene in advanced stages of tumors, wherein it develops immune-suppressive tumor microenvironments and induces the proliferation of cancer cells, invasion, angiogenesis, tumorigenesis, and metastasis. Higher TGFβ expression leads to the instigation and development of cancer. Therefore, suppressing TGFβ signals may present a potential treatment option for inhibiting tumorigenesis and metastasis. Different inhibitory molecules, including ligand traps, anti-sense oligo-nucleotides, small molecule receptor-kinase inhibitors, small molecule inhibitors, and vaccines, have been developed and clinically trialed for blocking the TGFβ signaling pathway. These molecules are not pro-oncogenic response-specific but block all signaling effects induced by TGFβ. Nonetheless, targeting the activation of TGFβ signaling with maximized specificity and minimized toxicity can enhance the efficacy of therapeutic approaches against this signaling pathway. The molecules that are used to target TGFβ are non-cytotoxic to cancer cells but designed to curtail the over-activation of invasion and metastasis driving TGFβ signaling in stromal and cancer cells. Here, we discussed the critical role of TGFβ in tumorigenesis, and metastasis, as well as the outcome and the promising achievement of TGFβ inhibitory molecules in the treatment of cancer.

    Topics: Carcinogenesis; Cell Differentiation; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2023
Application of rhBMP in spinal fusion surgery: any correlation of cancer incidence? A systematic review and meta-analysis.
    European spine journal : official publication of the European Spine Society, the European Spinal Deformity Society, and the European Section of the Cervical Spine Research Society, 2023, Volume: 32, Issue:6

    Safety concerns regarding the application of bone morphogenetic proteins (BMPs) have been highlighted in recent years. It is noted that both BMP and their receptors being identified as a trigger for cancer growth. Here, we aimed to determine the safety and efficacy of BMP for spinal fusion surgery.. We conducted this systematic review on topics of spinal fusion surgery with rhBMP application from three database (PubMed, EuropePMC, and Clinicaltrials.gov) with MeSH phrases such as "rh-BMP," "rhBMP," "spine surgery," "spinal arthrodesis," and "spinal fusion" were searched (using the Boolean operators "and" and "or"). Our research includes all articles, as long as published in English language. In the face of disagreement between the two reviewers, we discussed it together until all authors reached a consensus. The primary key outcome of our study is the incidence of cancer following rhBMP implantation.. Our study included a total of 8 unique studies (n = 37,682). The mean follow-up varies among all studies, with the longest follow-up is 66 months. Our meta-analysis showed that exposure to rhBMP in spinal surgery did increase the risk of cancers (RR 1.85, 95%CI [1.05, 3.24], p = 0.03).. Our study found that rhBMP was not associated with the increased risk of cancer incidence within the rhBMP cohort. Still, we did face several limitations, in which further studies are needed to confirm the result of our meta-analysis.

    Topics: Bone Morphogenetic Protein 2; Humans; Incidence; Neoplasms; Neurosurgical Procedures; Recombinant Proteins; Spinal Fusion; Transforming Growth Factor beta

2023
Dynamics of transforming growth factor β signaling and therapeutic efficacy.
    Growth factors (Chur, Switzerland), 2023, Volume: 41, Issue:2

    Transforming growth factor β (TGFβ) is a multifunctional cytokine, and its signalling responses are exerted via integrated intracellular pathways and complex regulatory mechanisms. Due to its high potency, TGFβ signalling is tightly controlled under normal circumstances, while its dysregulation in cancer favours metastasis. The recognised potential of TGFβ as a therapeutic target led to emerging development of anti-TGFβ reagents with preclinical success, yet these therapeutics failed to recapitulate their efficacy in experimental settings. In this review, possible reasons for this inconsistency are discussed, addressing the knowledge gap between theoretical and actual behaviours of TGFβ signalling. Previous studies on oncogenic cells have demonstrated the spatiotemporal heterogeneity of TGFβ signalling intensity. Under feedback mechanisms and exosomal ligand recycling, cancer cells may achieve cyclic TGFβ signalling to facilitate dissemination and colonisation. This challenges the current presumption of persistently high TGFβ signalling in cancer, pointing to a new direction of research on TGFβ-targeted therapeutics.

    Topics: Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2023
Therapeutic targeting approach on epithelial-mesenchymal plasticity to combat cancer metastasis.
    Medical oncology (Northwood, London, England), 2023, May-29, Volume: 40, Issue:7

    Epithelial-mesenchymal plasticity (EMP) is a process in which epithelial cells lose their characteristics and acquire mesenchymal properties, leading to increased motility and invasiveness, which are key factors in cancer metastasis. Targeting EMP has emerged as a promising therapeutic approach to combat cancer metastasis. Various strategies have been developed to target EMP, including inhibition of key signaling pathways, such as TGF-β, Wnt/β-catenin, and Notch, that regulate EMP, as well as targeting specific transcription factors, such as Snail, Slug, and Twist, that promote EMP. Additionally, targeting the tumor microenvironment, which plays a critical role in promoting EMP, has also shown promise. Several preclinical and clinical studies have demonstrated the efficacy of EMP-targeting therapies in inhibiting cancer metastasis. However, further research is needed to optimize these strategies and improve their clinical efficacy. Overall, therapeutic targeting of EMP represents a promising approach for the development of novel cancer therapies that can effectively inhibit metastasis, a major cause of cancer-related mortality.

    Topics: Epithelial Cells; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2023
Small extracellular vesicle TGF-β in cancer progression and immune evasion.
    Cancer gene therapy, 2023, Volume: 30, Issue:10

    Transforming growth factor-β (TGF-β) is a well-known cytokine that controls various processes in normal physiology and disease context. Strong preclinical and clinical literature supports the crucial roles of the TGF-β in several aspects of cancer biology. Recently emerging evidence reveals that the release of TGF-β from tumor/immune/stromal cells in small extracellular vesicles (sEVs) plays an important part in tumor development and immune evasion. Hence, this review aims to address the packaging, release, and signaling pathways of TGF-β carried in sEVs (sEV-TGF-β) in cancer, and to explore its underpinning roles in tumor development, growth, progression, metastasis, etc. We also highlight key progresses in deciphering the roles of sEV-TGF-β in subverting anti-tumor immune responses. The paper ends with a focus on the clinical significance of TGF-β carried in sEVs and draws attention to its diagnostic, therapeutic, and prognostic importance.

    Topics: Cytokines; Humans; Immune Evasion; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2023
Dual anti-PD-(L)1/TGF-β inhibitors in cancer immunotherapy - Updated.
    International immunopharmacology, 2023, Volume: 122

    Topics: B7-H1 Antigen; Humans; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2023
Bone morphogenetic protein (BMP)9 in cancer development: mechanistic, diagnostic, and therapeutic approaches?
    Journal of drug targeting, 2023, Volume: 31, Issue:7

    Bone morphogenetic protein (BMP)-9 is considered a member of the transforming growth factor (TGF)β superfamily. It was first found as an inducer of bone and cartilage formation and then discovered that this factor mediates several physiologic functions and hemostasis. Besides physiological conditions, BMP9 has also been elucidated that it is involved in several pathological situations, especially cancer. In various cancers, dysregulation of BMP9 has raised the issue that BMP9 might play a conflicting role in tumour development. BMP9 binding to its receptors (BMPRs), including ALKs and BMPRII, induces canonical SMAD-dependent and non-canonical PI3K/AKT and MAPK signalling pathways in tumour cells. BMP9,

    Topics: Bone Morphogenetic Proteins; Growth Differentiation Factor 2; Humans; Neoplasms; Phosphatidylinositol 3-Kinases; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2023
Bispecific antibody targeting TGF-β and PD-L1 for synergistic cancer immunotherapy.
    Frontiers in immunology, 2023, Volume: 14

    The PD-1/PD-L1 signaling pathway plays a crucial role in cancer immune evasion, and the use of anti-PD-1/PD-L1 antibodies represents a significant milestone in cancer immunotherapy. However, the low response rate observed in unselected patients and the development of therapeutic resistance remain major obstacles to their clinical application. Accumulating studies showed that overexpressed TGF-β is another immunosuppressive factor apart from traditional immune checkpoints. Actually, the effects of PD-1 and TGF-β pathways are independent and interactive, which work together contributing to the immune evasion of cancer cell. It has been verified that blocking TGF-β and PD-L1 simultaneously could enhance the efficacy of PD-L1 monoclonal antibody and overcome its treatment resistance. Based on the bispecific antibody or fusion protein technology, multiple bispecific and bifunctional antibodies have been developed. In the preclinical and clinical studies, these updated antibodies exhibited potent anti-tumor activity, superior to anti-PD-1/PD-L1 monotherapies. In the review, we summarized the advances of bispecific antibodies targeting TGF-β and PD-L1 in cancer immunotherapy. We believe these next-generation immune checkpoint inhibitors would substantially alter the cancer treatment paradigm, especially in anti-PD-1/PD-L1-resistant patients.

    Topics: Antibodies, Bispecific; Antibodies, Monoclonal; B7-H1 Antigen; Humans; Immunotherapy; Neoplasms; Transforming Growth Factor beta

2023
Context-dependent TGFβ family signalling in cell fate regulation.
    Nature reviews. Molecular cell biology, 2023, Volume: 24, Issue:12

    The transforming growth factor-β (TGFβ) family are a large group of evolutionarily conserved cytokines whose signalling modulates cell fate decision-making across varying cellular contexts at different stages of life. Here we discuss new findings in early embryos that reveal how, in contrast to our original understanding of morphogen interpretation, robust cell fate specification can originate from a noisy combination of signalling inputs and a broad range of signalling levels. We compare this evidence with novel findings on the roles of TGFβ family signalling in tissue maintenance and homeostasis during juvenile and adult life, spanning the skeletal, haemopoietic and immune systems. From these comparisons, it emerges that in contrast to robust developing systems, relatively small perturbations in TGFβ family signalling have detrimental effects at later stages in life, leading to aberrant cell fate specification and disease, for example in cancer or congenital disorders. Finally, we highlight novel strategies to target and amend dysfunction in signalling and discuss how gleaning knowledge from different fields of biology can help in the development of therapeutics for aberrant TGFβ family signalling in disease.

    Topics: Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2023
Lactate acidosis and simultaneous recruitment of TGF-β leads to alter plasticity of hypoxic cancer cells in tumor microenvironment.
    Pharmacology & therapeutics, 2023, Volume: 250

    Lactate acidosis is often observed in the tumor microenvironment (TME) of solid tumors. This is because glucose breaks down quickly via glycolysis, causing lactate acidity. Lactate is harmful to healthy cells, but is a major oncometabolite for solid cancer cells that do not receive sufficient oxygen. As an oncometabolite, it helps tumor cells perform different functions, which helps solid hypoxic tumor cells spread to other parts of the body. Studies have shown that the acidic TME contains VEGF, Matrix metalloproteinases (MMPs), cathepsins, and transforming growth factor-β (TGF-β), all of which help spread in direct and indirect ways. Although each cytokine is important in its own manner in the TME, TGF-β has received much attention for its role in metastatic transformation. Several studies have shown that lactate acidosis can cause TGF-β expression in solid hypoxic cancers. TGF-β has also been reported to increase the production of fatty acids, making cells more resistant to treatment. TGF-β has also been shown to control the expression of VEGF and MMPs, which helps solid hypoxic tumors become more aggressive by helping them spread and create new blood vessels through an unknown process. The role of TGF-β under physiological conditions has been described previously. In this study, we examined the role of TGF-β, which is induced by lactate acidosis, in the spread of solid hypoxic cancer cells. We also found that TGF-β and lactate work together to boost fatty acid production, which helps angiogenesis and invasiveness.

    Topics: Acidosis; Humans; Hypoxia; Lactic Acid; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment; Vascular Endothelial Growth Factor A

2023
Role of long non-coding RNAs and TGF-β signaling in the regulation of breast cancer pathogenesis and therapeutic targets.
    Cytokine, 2023, Volume: 170

    The cytokine known as transforming growth factor (TGF) is essential for cell development, differentiation, and apoptosis in BC. TGF-β dysregulation can either promote or inhibit tumor development, and it is a key signaling pathway in BC spread. A recently identified family of ncRNAs known as lncRNAs has received a great deal of effort and is an important regulator of many cellular processes, including transcription of genes, chromatin remodeling, progression of the cell cycle, and posttranscriptional processing. Furthermore, both TGF-β signaling and lncRNAs serve as important early-stage biomarkers for BC diagnosis and prognosis and also play a significant role in BC drug resistance. According to recent studies, lncRNAs can regulate TGF-β by modulating its cofactors in BC. However, the particular functions of lncRNAs and the TGF-β pathway in controlling BC progression are not well understood yet. This review explores the lncRNAs' functional properties in BC as tumor suppressors or oncogenes in the regulation of genes, with a focus on dysregulated TGF-β signaling. Further, we emphasize the functional roles of lncRNAs and TGF-β pathway in the progression of BC to discover new treatment strategies and better comprehend the fundamental cellular pathways.

    Topics: Cell Differentiation; Cytokines; Neoplasms; RNA, Long Noncoding; Signal Transduction; Transforming Growth Factor beta

2023
Egress of resident memory T cells from tissue with neoadjuvant immunotherapy: Implications for systemic anti-tumor immunity.
    Oral oncology, 2023, Volume: 146

    Resident memory T (T. The presence of T. Reversing tumor sequestration of tumor-specific T cells prior to surgical removal or radiation of tumor may increase systemic antitumor immunity. This finding may underlie the improved recurrence free survival observed with neoadjuvant immunotherapy in clinical trials.

    Topics: Humans; Immunologic Memory; Immunotherapy; Memory T Cells; Neoadjuvant Therapy; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2023
Exploiting Canonical TGFβ Signaling in Cancer Treatment.
    Molecular cancer therapeutics, 2022, Volume: 21, Issue:1

    TGFβ is a pleiotropic cytokine that plays critical roles to define cancer cell phenotypes, construct the tumor microenvironment, and suppress antitumor immune responses. As such, TGFβ is a lynchpin for integrating cancer cell intrinsic pathways and communication among host cells in the tumor and beyond that together affect responses to genotoxic, targeted, and immune therapy. Despite decades of preclinical and clinical studies, evidence of clinical benefit from targeting TGFβ in cancer remains elusive. Here, we review the mechanisms by which TGFβ acts to oppose successful cancer therapy, the reported prognostic and predictive value of TGFβ biomarkers, and the potential impact of inhibiting TGFβ in precision oncology. Paradoxically, the diverse mechanisms by which TGFβ impedes therapeutic response are a principal barrier to implementing TGFβ inhibitors because it is unclear which TGFβ mechanism is functional in which patient. Companion diagnostic tools and specific biomarkers of TGFβ targeted biology will be the key to exploiting TGFβ biology for patient benefit.

    Topics: Humans; Neoplasms; Prognosis; Signal Transduction; Transforming Growth Factor beta

2022
Overcoming TGFβ-mediated immune evasion in cancer.
    Nature reviews. Cancer, 2022, Volume: 22, Issue:1

    Transforming growth factor-β (TGFβ) signalling controls multiple cell fate decisions during development and tissue homeostasis; hence, dysregulation of this pathway can drive several diseases, including cancer. Here we discuss the influence that TGFβ exerts on the composition and behaviour of different cell populations present in the tumour immune microenvironment, and the context-dependent functions of this cytokine in suppressing or promoting cancer. During homeostasis, TGFβ controls inflammatory responses triggered by exposure to the outside milieu in barrier tissues. Lack of TGFβ exacerbates inflammation, leading to tissue damage and cellular transformation. In contrast, as tumours progress, they leverage TGFβ to drive an unrestrained wound-healing programme in cancer-associated fibroblasts, as well as to suppress the adaptive immune system and the innate immune system. In consonance with this key role in reprogramming the tumour microenvironment, emerging data demonstrate that TGFβ-inhibitory therapies can restore cancer immunity. Indeed, this approach can synergize with other immunotherapies - including immune checkpoint blockade - to unleash robust antitumour immune responses in preclinical cancer models. Despite initial challenges in clinical translation, these findings have sparked the development of multiple therapeutic strategies that inhibit the TGFβ pathway, many of which are currently in clinical evaluation.

    Topics: Humans; Immune Evasion; Immunotherapy; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
TGF-β signaling and the interaction between platelets and T-cells in tumor microenvironment: Foes or friends?
    Cytokine, 2022, Volume: 150

    T-cells, as the main immune cells in fighting against cancer cells, are usually overwhelmed by many factors. Tumor microenvironment (TME) changes are one of the factors that can limit T-cells functions. On the other hand, platelets which are known as the main source of transforming growth factor-β (TGF-β) in TME, are seemingly insignificant immune cells that can affect T-cell functions. There is a hypothesis that platelets might prevent tumor growth by stimulating cellular immunity, especially T-cells in pre-cancer status while they can inhibit T-cells and stimulate tumor growth in the advanced stage of cancer. Therefore, platelets could act as a double-edged sword in the activation of T-cells under pre-cancer and advanced stages of cancer conditions. In this review, the interaction between platelets and T cells in pre-cancer and advanced stages of cancer and the role of TGF-β signaling in different stages of cancer will be discussed.

    Topics: Blood Platelets; Humans; Neoplasms; T-Lymphocytes; Transforming Growth Factor beta; Tumor Microenvironment

2022
Dual inhibition of TGF-β and PD-L1: a novel approach to cancer treatment.
    Molecular oncology, 2022, Volume: 16, Issue:11

    Transforming growth factor-β (TGF-β) and programmed death ligand 1 (PD-L1) initiate signaling pathways with complementary, nonredundant immunosuppressive functions in the tumor microenvironment (TME). In the TME, dysregulated TGF-β signaling suppresses antitumor immunity and promotes cancer fibrosis, epithelial-to-mesenchymal transition, and angiogenesis. Meanwhile, PD-L1 expression inactivates cytotoxic T cells and restricts immunosurveillance in the TME. Anti-PD-L1 therapies have been approved for the treatment of various cancers, but TGF-β signaling in the TME is associated with resistance to these therapies. In this review, we discuss the importance of the TGF-β and PD-L1 pathways in cancer, as well as clinical strategies using combination therapies that block these pathways separately or approaches with dual-targeting agents (bispecific and bifunctional immunotherapies) that may block them simultaneously. Currently, the furthest developed dual-targeting agent is bintrafusp alfa. This drug is a first-in-class bifunctional fusion protein that consists of the extracellular domain of the TGF-βRII receptor (a TGF-β 'trap') fused to a human immunoglobulin G1 (IgG1) monoclonal antibody blocking PD-L1. Given the immunosuppressive effects of the TGF-β and PD-L1 pathways within the TME, colocalized and simultaneous inhibition of these pathways may potentially improve clinical activity and reduce toxicity.

    Topics: Antibodies, Monoclonal; B7-H1 Antigen; Humans; Immunotherapy; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
Promyelocytic leukemia protein (PML) and stem cells: from cancer to pluripotency.
    The International journal of developmental biology, 2022, Volume: 66, Issue:1-2-3

    The promyelocytic leukemia protein (PML) is the core organizer of cognate nuclear bodies (PML-NBs). Through physical interaction or modification of diverse protein clients, PML-NBs regulate a multitude of - often antithetical- biological processes such as antiviral and stress response, inhibition of cell proliferation and autophagy, and promotion of apoptosis or senescence. Although PML was originally recognized as a tumor-suppressive factor, more recent studies have revealed a "double-faced" agent role for PML. Indeed, PML displayed tumor cell pro-survival and pro-migratory functions via inhibition of migration suppressing molecules or promotion of transforming growth factor beta (TGF-β) mediated Epithelial-Mesenchymal Transition (EMT) that may promote cancer cell dissemination. In this line, PML was found to correlate with poor patient prognosis in distinct tumor contexts. Furthermore, in the last decade, a number of publications have implicated PML in the physiology of normal or cancer stem cells (CSCs). Promyelocytic leukemia protein activates fatty acid oxidation (FAO), a metabolic mechanism required for the asymmetric divisions and maintenance of hematopoietic stem cells (HSCs). In embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), PML is required for maintenance of the naïve and acquisition of the induced pluripotency state, respectively. Correspondingly, PML ablation causes significant morphological gene expression and lineage choice changes. In this review, we focus on the mechanisms orchestrated by PML and PML-NBs in cancer and healthy stem cells, from cell physiology to the regulation of chromatin dynamics.

    Topics: Autophagy; Humans; Neoplasms; Promyelocytic Leukemia Protein; Stem Cells; Transcription Factors; Transforming Growth Factor beta

2022
TGF-β and Cancer Immunotherapy.
    Biological & pharmaceutical bulletin, 2022, Volume: 45, Issue:2

    The cytokine, transforming growth factor beta (TGF-β), has a history of more than 40 years. TGF-β is secreted by many tumor cells and is associated with tumor growth and cancer immunity. The canonical TGF-β signaling pathway, SMAD, controls both tumor metastasis and immune regulation, thereby regulating cancer immunity. TGF-β regulates multiple types of immune cells in tumor microenvironment, including T cells, natural killer (NK) cells, and macrophages. One of the main roles of TGF-β in the tumor microenvironment is the generation of regulatory T cells, which contribute to the suppression of anti-tumor immunity. Because cancer is one of the highest causes of death globally, the discovery of immune checkpoint inhibitors by Honjo and Allison in cancer immunotherapy earned a Nobel Prize in 2018. TGF-β also regulates the levels of immune checkpoints inhibitory receptors on immune cells. Immune checkpoints inhibitors are now being developed along with anti-TGF-β antibody and/or TGF-β inhibitors. More recently, chimeric antigen receptors (CARs) were applied to cancer immunity and tried to combine with TGF-β blockers.

    Topics: Gene Expression Regulation, Neoplastic; Humans; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2022
The radiobiology of TGFβ.
    Seminars in cancer biology, 2022, Volume: 86, Issue:Pt 3

    Ionizing radiation is a pillar of cancer therapy that is deployed in more than half of all malignancies. The therapeutic effect of radiation is attributed to induction of DNA damage that kills cancers cells, but radiation also affects signaling that alters the composition of the tumor microenvironment by activating transforming growth factor β (TGFβ). TGFβ is a ubiquitously expressed cytokine that acts as biological lynchpin to orchestrate phenotypes, the stroma, and immunity in normal tissue; these activities are subverted in cancer to promote malignancy, a permissive tumor microenvironment and immune evasion. The radiobiology of TGFβ unites targets at the forefront of oncology-the DNA damage response and immunotherapy. The cancer cell intrinsic and extrinsic network of TGFβ responses in the irradiated tumor form a barrier to both genotoxic treatments and immunotherapy response. Here, we focus on the mechanisms by which radiation induces TGFβ activation, how TGFβ regulates DNA repair, and the dynamic regulation of the tumor immune microenvironment that together oppose effective cancer therapy. Strategies to inhibit TGFβ exploit fundamental radiobiology that may be the missing link to deploying TGFβ inhibitors for optimal patient benefit from cancer treatment.

    Topics: DNA Damage; Humans; Neoplasms; Radiobiology; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2022
CD105: tumor diagnosis, prognostic marker and future tumor therapeutic target.
    Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 2022, Volume: 24, Issue:8

    Cancer is one of the diseases with the highest morbidity and mortality rates worldwide, and its therapeutic options are inadequate. The endothelial glycoprotein, also known as CD105, is a type I transmembrane glycoprotein located on the surface of the cell membranes and it is one of the transforming growth factor-β (TGF-β) receptor complexes. It regulates the responses associated with binding to transforming growth factor β1 egg (Activin-A), bone morphogenetic protein 2 (BMP-2), and bone morphogenetic protein 7 (BMP-7). Additionally, it is involved in the regulation of angiogenesis. This glycoprotein is indispensable in the treatment of tumor angiogenesis, and it also plays a leading role in tumor angiogenesis therapy. Therefore, CD105 is considered to be a novel therapeutic target. In this study, we explored the significance of CD105 in the diagnosis, treatment and prognosis of various tumors, and provided evidence for the effect and mechanism of CD105 on tumors.

    Topics: Antigens, CD; Endoglin; Humans; Neoplasms; Neovascularization, Pathologic; Prognosis; Receptors, Cell Surface; Transforming Growth Factor beta

2022
The functional multipotency of transforming growth factor β signaling at the intersection of senescence and cancer.
    Cellular and molecular life sciences : CMLS, 2022, Mar-19, Volume: 79, Issue:4

    The transforming growth factor β (TGF-β) family of cytokines comprises a group of proteins, their receptors, and effector molecules that, in a coordinated manner, modulate a plethora of physiological and pathophysiological processes. TGF-β1 is the best known and plausibly most active representative of this group. It acts as an immunosuppressant, contributes to extracellular matrix remodeling, and stimulates tissue fibrosis, differentiation, angiogenesis, and epithelial-mesenchymal transition. In recent years, this cytokine has been established as a vital regulator of organismal aging and cellular senescence. Finally, the role of TGF-β1 in cancer progression is no longer in question. Because this protein is involved in so many, often overlapping phenomena, the question arises whether it can be considered a molecular bridge linking some of these phenomena together and governing their reciprocal interactions. In this study, we reviewed the literature from the perspective of the role of various TGF-β family members as regulators of a complex mutual interplay between senescence and cancer. These aspects are then considered in a broader context of remaining TGF-β-related functions and coexisting processes. The main narrative axis in this work is centered around the interaction between the senescence of normal peritoneal cells and ovarian cancer cells. The discussion also includes examples of TGF-β activity at the interface of other normal and cancer cell types.

    Topics: Cellular Senescence; Humans; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2022
Targeting transforming growth factor-β signalling for cancer prevention and intervention: Recent advances in developing small molecules of natural origin.
    Clinical and translational medicine, 2022, Volume: 12, Issue:4

    Cancer is the world's second leading cause of death, but a significant advancement in cancer treatment has been achieved within the last few decades. However, major adverse effects and drug resistance associated with standard chemotherapy have led towards targeted treatment options.. Transforming growth factor-β (TGF-β) signaling plays a key role in cell proliferation, differentiation, morphogenesis, regeneration, and tissue homeostasis. The prime objective of this review is to decipher the role of TGF-β in oncogenesis and to evaluate the potential of various natural and synthetic agents to target this dysregulated pathway to confer cancer preventive and anticancer therapeutic effects.. Various authentic and scholarly databases were explored to search and obtain primary literature for this study. The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) criteria was followed for the review.. Here we provide a comprehensive and critical review of recent advances on our understanding of the effect of various bioactive natural molecules on the TGF-β signaling pathway to evaluate their full potential for cancer prevention and therapy.. Based on emerging evidence as presented in this work, TGF-β-targeting bioactive compounds from natural sources can serve as potential therapeutic agents for prevention and treatment of various human malignancies.

    Topics: Cell Proliferation; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2022
Targeting TGF-β signal transduction for fibrosis and cancer therapy.
    Molecular cancer, 2022, 04-23, Volume: 21, Issue:1

    Transforming growth factor β (TGF-β) has long been identified with its intensive involvement in early embryonic development and organogenesis, immune supervision, tissue repair, and adult homeostasis. The role of TGF-β in fibrosis and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, overexpressed TGF-β causes epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) deposition, cancer-associated fibroblast (CAF) formation, which leads to fibrotic disease, and cancer. Given the critical role of TGF-β and its downstream molecules in the progression of fibrosis and cancers, therapeutics targeting TGF-β signaling appears to be a promising strategy. However, due to potential systemic cytotoxicity, the development of TGF-β therapeutics has lagged. In this review, we summarized the biological process of TGF-β, with its dual role in fibrosis and tumorigenesis, and the clinical application of TGF-β-targeting therapies.

    Topics: Epithelial-Mesenchymal Transition; Extracellular Matrix; Fibrosis; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2022
Preclinical and clinical studies of bintrafusp alfa, a novel bifunctional anti-PD-L1/TGFβRII agent: Current status.
    Experimental biology and medicine (Maywood, N.J.), 2022, Volume: 247, Issue:13

    Bintrafusp alfa (anti-PD-L1/TGFβRII) is a first-in-class bifunctional agent designed to act both as a checkpoint inhibitor and as a "trap" for TGFβ in the tumor microenvironment (TME). This article is designed to review the preclinical studies interrogating the mode of action of bintrafusp alfa and to present a comprehensive overview of recent bintrafusp alfa clinical studies. Preclinical studies have demonstrated that bintrafusp alfa immune-mediating and antitumor activity can be enhanced by combining it with a human papillomavirus (HPV) therapeutic cancer vaccine, a tumor-targeting interleukin 12 (IL-12) immunocytokine and/or an IL-15 superagonist. The importance of TGFβ in HPV-associated malignancies is also reviewed. The clinical studies reviewed span extended phase I cohorts in patients with a spectrum of malignancies, two randomized phase II studies in lung and one in biliary tract cancers in which bintrafusp alfa did not demonstrate superiority over standard-of-care therapies, and provocative results in patients with HPV-associated malignancies, where as a monotherapy, bintrafusp alfa has shown response rates of 35%, compared to overall response rate (ORR) of 12-24% seen with other Food and Drug Administration (FDA)-approved or standard-of-care agents. This article also reviews preliminary phase II study results of patients with HPV

    Topics: B7-H1 Antigen; Clinical Trials, Phase II as Topic; Humans; Immunologic Factors; Immunotherapy; Interleukin-12; Neoplasms; Papillomavirus Infections; Randomized Controlled Trials as Topic; Receptor, Transforming Growth Factor-beta Type II; Transforming Growth Factor beta; Tumor Microenvironment

2022
NF-κB as a regulator of cancer metastasis and therapy response: A focus on epithelial-mesenchymal transition.
    Journal of cellular physiology, 2022, Volume: 237, Issue:7

    Metastasis of tumor cells is a complex challenge and significantly diminishes the overall survival and prognosis of cancer patients. The epithelial-to-mesenchymal transition (EMT) is a well-known mechanism responsible for the invasiveness of tumor cells. A number of molecular pathways can regulate the EMT mechanism in cancer cells and nuclear factor-kappaB (NF-κB) is one of them. The nuclear translocation of NF-κB p65 can induce the transcription of several genes involved in EMT induction. The present review describes NF-κB and EMT interaction in cancer cells and their association in cancer progression. Due to the oncogenic role NF-κB signaling, its activation enhances metastasis of tumor cells via EMT induction. This has been confirmed in various cancers including brain, breast, lung and gastric cancers, among others. The ZEB1/2, transforming growth factor-β, and Slug as inducers of EMT undergo upregulation by NF-κB to promote metastasis of tumor cells. After EMT induction driven by NF-κB, a significant decrease occurs in E-cadherin levels, while N-cadherin and vimentin levels undergo an increase. The noncoding RNAs can potentially also function as upstream mediators and modulate NF-κB/EMT axis in cancers. Moreover, NF-κB/EMT axis is involved in mediating drug resistance in tumor cells. Thus, suppressing NF-κB/EMT axis can also promote the sensitivity of cancer cells to chemotherapeutic agents.

    Topics: Animals; Cell Line, Tumor; Epithelial-Mesenchymal Transition; Humans; Neoplasms; NF-kappa B; Signal Transduction; Transforming Growth Factor beta

2022
The roles of TGF-β and VEGF pathways in the suppression of antitumor immunity in melanoma and other solid tumors.
    Pharmacology & therapeutics, 2022, Volume: 240

    Immune checkpoint blockade (ICB) has become well-known in cancer therapy, strengthening the body's antitumor immune response rather than directly targeting cancer cells. Therapies targeting immune inhibitory checkpoints, such as PD-1, PD-L1, and CTLA-4, have resulted in impressive clinical responses across different types of solid tumors. However, as with other types of cancer treatments, ICB-based immunotherapy is hampered by both innate and acquired drug resistance. We previously reported the enrichment of gene signatures associated with wound healing, epithelial-to-mesenchymal, and angiogenesis processes in the tumors of patients with innate resistance to PD-1 checkpoint antibody therapy; we termed these the Innate Anti-PD-1 Resistance Signatures (IPRES). The TGF-β and VEGFA pathways emerge as the dominant drivers of IPRES-associated processes. Here, we review these pathways' functions, their roles in immunosuppression, and the currently available therapies that target them. We also discuss recent developments in the targeting of TGF-β using a specific antibody class termed trap antibody. The application of trap antibodies opens the promise of localized targeting of the TGF-β and VEGFA pathways within the tumor microenvironment. Such specificity may offer an enhanced therapeutic window that enables suppression of the IPRES processes in the tumor microenvironment while sparing the normal homeostatic functions of TGF-β and VEGFA in healthy tissues.

    Topics: Antibodies; Humans; Immunotherapy; Melanoma; Neoplasms; Programmed Cell Death 1 Receptor; Transforming Growth Factor beta; Tumor Microenvironment; Vascular Endothelial Growth Factor A

2022
Serine-Arginine Protein Kinase 1 (SRPK1): a systematic review of its multimodal role in oncogenesis.
    Molecular and cellular biochemistry, 2022, Volume: 477, Issue:10

    Alternative splicing is implicated in each of the hallmarks of cancer, and is mechanised by various splicing factors. Serine-Arginine Protein Kinase 1 (SRPK1) is an enzyme which moderates the activity of splicing factors rich in serine/arginine domains. Here we review SRPK1's relationship with various cancers by performing a systematic review of all relevant published data. Elevated SRPK1 expression correlates with advanced disease stage and poor survival in many epithelial derived cancers. Numerous pre-clinical studies investigating a host of different tumour types; have found increased SRPK1 expression to be associated with proliferation, invasion, migration and apoptosis in vitro as well as tumour growth, tumourigenicity and metastasis in vivo. Aberrant SRPK1 expression is implicated in various signalling pathways associated with oncogenesis, a number of which, such as the PI3K/AKT, NF-КB and TGF-Beta pathway, are implicated in multiple different cancers. SRPK1-targeting micro RNAs have been identified in a number of studies and shown to have an important role in regulating SRPK1 activity. SRPK1 expression is also closely related to the response of various tumours to platinum-based chemotherapeutic agents. Future clinical applications will likely focus on the role of SRPK1 as a biomarker of treatment resistance and the potential role of its inhibition.

    Topics: Arginine; Arginine Kinase; Carcinogenesis; Cell Transformation, Neoplastic; Humans; Neoplasms; NF-kappa B; Phosphatidylinositol 3-Kinases; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-akt; RNA Splicing Factors; Serine; Transforming Growth Factor beta

2022
The Synergistic Cooperation between TGF-β and Hypoxia in Cancer and Fibrosis.
    Biomolecules, 2022, 04-25, Volume: 12, Issue:5

    Transforming growth factor β (TGF-β) is a multifunctional cytokine regulating homeostasis and immune responses in adult animals and humans. Aberrant and overactive TGF-β signaling promotes cancer initiation and fibrosis through epithelial-mesenchymal transition (EMT), as well as the invasion and metastatic growth of cancer cells. TGF-β is a key factor that is active during hypoxic conditions in cancer and is thereby capable of contributing to angiogenesis in various types of cancer. Another potent role of TGF-β is suppressing immune responses in cancer patients. The strong tumor-promoting effects of TGF-β and its profibrotic effects make it a focus for the development of novel therapeutic strategies against cancer and fibrosis as well as an attractive drug target in combination with immune regulatory checkpoint inhibitors. TGF-β belongs to a family of cytokines that exert their function through signaling via serine/threonine kinase transmembrane receptors to intracellular Smad proteins via the canonical pathway and in combination with co-regulators such as the adaptor protein and E3 ubiquitin ligases TNF receptor-associated factor 4 (TRAF4) and TNF receptor-associated factor 6 (TRAF6) to promote non-canonical pathways. Finally, the outcome of gene transcription initiated by TGF-β is context-dependent and controlled by signals exerted by other growth factors such as EGF and Wnt. Here, we discuss the synergistic cooperation between TGF-β and hypoxia in development, fibrosis and cancer.

    Topics: Animals; Fibrosis; Humans; Hypoxia; Neoplasms; Smad Proteins; TNF Receptor-Associated Factor 4; Transforming Growth Factor beta

2022
Oncofetal proteins and cancer stem cells.
    Essays in biochemistry, 2022, 09-16, Volume: 66, Issue:4

    Cancer stem cells (CSCs) are considered as a small population of cells with stem-like properties within the tumor bulk, and are largely responsible for tumor recurrence, metastasis, and therapy resistance. CSCs share critical features with embryonic stem cells (ESCs). The pluripotent transcription factors (TFs) and developmental signaling pathways of ESCs are invariably hijacked by CSCs termed 'oncofetal drivers' in many cancers, which are rarely detectable in adult tissues. The unique expression pattern makes oncofetal proteins ideal therapeutic targets in cancer treatment. Therefore, elucidation of oncofetal drivers in cancers is critical for the development of effective CSCs-directed therapy. In this review, we summarize the common pluripotent TFs such as OCT4, SOX2, NANOG, KLF4, MYC, SALL4, and FOXM1, as well as the development signaling including Wnt/β-catenin, Hedgehog (Hh), Hippo, Notch, and TGF-β pathways of ESCs and CSCs. We also describe the newly identified oncofetal proteins that drive the self-renewal, plasticity, and therapy-resistance of CSCs. Finally, we explore how the clinical implementation of targeting oncofetal drivers, including small-molecule inhibitors, vaccines, antibodies, and CAR-T (chimeric antigen receptor T cell) can facilitate the development of CSCs-directed therapy.

    Topics: Antigens, Neoplasm; beta Catenin; Hedgehog Proteins; Humans; Neoplasms; Neoplastic Stem Cells; Receptors, Chimeric Antigen; Transforming Growth Factor beta

2022
STAT3-EMT axis in tumors: Modulation of cancer metastasis, stemness and therapy response.
    Pharmacological research, 2022, Volume: 182

    Epithelial-to-mesenchymal transition (EMT) mechanism is responsible for metastasis of tumor cells and their spread to various organs and tissues of body, providing undesirable prognosis. In addition to migration, EMT increases stemness and mediates therapy resistance. Hence, pathways involved in EMT regulation should be highlighted. STAT3 is an oncogenic pathway that can elevate growth rate and migratory ability of cancer cells and induce drug resistance. The inhibition of STAT3 signaling impairs cancer progression and promotes chemotherapy-mediated cell death. Present review focuses on STAT3 and EMT interaction in modulating cancer migration. First of all, STAT3 is an upstream mediator of EMT and is able to induce EMT-mediated metastasis in brain tumors, thoracic cancers and gastrointestinal cancers. Therefore, STAT3 inhibition significantly suppresses cancer metastasis and improves prognosis of patients. EMT regulators such as ZEB1/2 proteins, TGF-β, Twist, Snail and Slug are affected by STAT3 signaling to stimulate cancer migration and invasion. Different molecular pathways such as miRNAs, lncRNAs and circRNAs modulate STAT3/EMT axis. Furthermore, we discuss how STAT3 and EMT interaction affects therapy response of cancer cells. Finally, we demonstrate targeting STAT3/EMT axis by anti-tumor agents and clinical application of this axis for improving patient prognosis.

    Topics: Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Neoplasm Metastasis; Neoplasms; Signal Transduction; STAT3 Transcription Factor; Transforming Growth Factor beta

2022
The Love-Hate Relationship Between TGF-β Signaling and the Immune System During Development and Tumorigenesis.
    Frontiers in immunology, 2022, Volume: 13

    Since TGF-β was recognized as an essential secreted cytokine in embryogenesis and adult tissue homeostasis a decade ago, our knowledge of the role of TGF-β in mammalian development and disease, particularly cancer, has constantly been updated. Mounting evidence has confirmed that TGF-β is the principal regulator of the immune system, as deprivation of TGF-β signaling completely abrogates adaptive immunity. However, enhancing TGF-β signaling constrains the immune response through multiple mechanisms, including boosting Treg cell differentiation and inducing CD8

    Topics: Animals; Carcinogenesis; CD8-Positive T-Lymphocytes; Cell Transformation, Neoplastic; Humans; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2022
Saga of monokines in shaping tumour-immune microenvironment: Origin to execution.
    Cytokine, 2022, Volume: 157

    Cellular communication mediated by cytokines is an important mechanism dictating immune responses, their cross talk and final immune output. Cytokines play a major role in dictating the immune outcome to cancer by regulating the events of development, differentiation and activation of innate immune cells. Cytokines are pleiotropic in nature, hence understanding their role individually or as member of network cytokines is critical to delineate their role in tumour immunity. Tumour systemically manipulates the immune system to evade and escape immune recognition for their uncontrollable growth and metastasis. The developing tumour comprise a large and diverse set of myeloid cells which are vulnerable to manipulation by the tumour-microenvironment. The innate immune cells of the monocytic lineage skew the fate of the adaptive immune cells and thus dictating cancer elimination or progression. Targeting cells at tumour cite is preposterous owing to their tight network, poor reach and abundance of immunosuppressive mechanisms. Monocytic lineage-derived cytokines (monokines) play crucial role in tumour regression or progression by either directly killing the tumour cells with TNFα or promoting its growth by TGFβ. In addition, the monokines like IL-12, IL-1β, IL-6, IL-10 and TGFβ direct the adaptive immune cells to secrete anti-tumour cytokines, TNFα, IFNγ, perforin and granzyme or pro-tumour cytokines, IL-10 and TGFβ. In this review, we elucidate the roles of monokines in dictating the fate of tumour by regulating responses at various stages of generation, differentiation and activation of immune cells along with the extensive cross talk. We have attempted to delineate the synergy and antagonism of major monokines among themselves or with tumour-derived or adaptive immune cytokines. The review provides an update on the possibilities of placing monokines to potential practical use as cytokine therapy against cancer.

    Topics: Cytokines; Humans; Interleukin-10; Monocytes; Monokines; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor-alpha

2022
Cancer Immunotherapy: Diverse Approaches and Obstacles.
    Current pharmaceutical design, 2022, Volume: 28, Issue:29

    Cancer immunotherapy approaches have progressed significantly during the last decade due to the significant improvement of our understanding of immunologic evasion of malignant cells. Depending on the type, stage, and grade of cancer, distinct immunotherapy approaches are being designed and recommended; each is different in efficacy and adverse effects. Malignant cells can adopt multiple strategies to alter the normal functioning of the immune system in recognizing and eliminating them. These strategies include secreting different immunosuppressive factors, polarizing tumor microenvironment cells to immunosuppressive ones, and interfering with the normal function of the antigen processing machinery (APM). In this context, careful evaluation of immune surveillance has led to a better understanding of the roles of cytokines, including IL-2, IL-12, IL-15, interferon-α (IFN-α), tumor necrosis factor-α (TNF-α), and transforming growth factor-β (TGF-β) in cancer formation and their potential application in cancer immunotherapy. Additionally, monoclonal antibodies (mAbs), adoptive cell therapy approaches, immune checkpoint blockade, and cancer vaccines also play significant roles in cancer immunotherapy. Moreover, the development of clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/CAS9) as an outstanding genome editing tool resolved many obstacles in cancer immunotherapy. In this regard, this review aimed to investigate the impacts of different immunotherapy approaches and their potential roles in the current and future roads of cancer treatment. Whatever the underlying solution for treating highly malignant cancers is, it seems that solving the question is nowhere near an achievement unless the precise cooperation of basic science knowledge with our translational experience.

    Topics: Antibodies, Monoclonal; Cancer Vaccines; CRISPR-Associated Protein 9; Humans; Immune Checkpoint Inhibitors; Immunotherapy; Interferon-alpha; Interleukin-12; Interleukin-15; Interleukin-2; Neoplasms; Transforming Growth Factor beta; Transforming Growth Factors; Tumor Microenvironment; Tumor Necrosis Factor-alpha

2022
Cancer-associated fibroblasts: Vital suppressors of the immune response in the tumor microenvironment.
    Cytokine & growth factor reviews, 2022, Volume: 67

    Since the "seed and soil" hypothesis was proposed, the biological functions of the tumor microenvironment (TME), especially its stromal components, have received increasing attention. Cancer-associated fibroblasts (CAFs) are the major components of the stromal region, providing material support for tumor cell proliferation, migration, and invasion. Furthermore, CAFs are important mediators of suppressing immune responses by attracting the accumulation of immunosuppressive cells through cytokine/chemokine secretion. In this review, we summarized the major cytokines, chemokines and metabolites, including transforming growth factor-β (TGF-β), interleukin-6 (IL-6), C-X-C chemokine ligand (CXCL)12, C-C chemokine ligand (CCL) 2, prostaglandin E2 (PGE2), and other factors, by which CAFs suppress the immune systems in a variety of cancers. More importantly, we highlight potential therapeutic strategies to alleviate the immunosuppression produced by CAFs, thereby inhibiting tumor progression.

    Topics: Cancer-Associated Fibroblasts; Cytokines; Fibroblasts; Humans; Immunity; Ligands; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
BETA PRIME: Phase I study of AdAPT-001 as monotherapy and combined with a checkpoint inhibitor in superficially accessible, treatment-refractory solid tumors.
    Future oncology (London, England), 2022, Volume: 18, Issue:29

    AdAPT-001 is an investigational therapy consisting of a replicative type 5 adenovirus armed with a TGF-β receptor-immunoglobulin Fc fusion trap, designed to neutralize isoforms 1 and 3 of the profibrotic and immunosuppressive cytokine, TGF-β. In preclinical studies with an immunocompetent mouse model, AdAPT-001 eradicated directly treated 'cold' tumors as well as distant untreated tumors, and, from its induction of systemic CD8. The purpose of this study is to find out more about the experimental oncolytic virus called AdAPT-001 that has been designed to selectively eliminate cancer cells. The virus is also designed to make a particular protein called a TGF-β trap, which neutralizes TGF-β, an overproduced chemical in cancer cells that puts the immune system into a comatose state. This article discusses a clinical trial called BETA PRIME for patients with no other standard treatment options. The trial will explore different doses of AdAPT-001 both alone and in combination with an approved checkpoint inhibitor or another immunotherapy, which blocks the ‘off' signal on immune cells, to determine the safest and best dose.

    Topics: Animals; Cell Line, Tumor; Clinical Trials, Phase I as Topic; Cytokines; Humans; Immunoglobulins; Immunotherapy; Mice; Neoplasms; Oncolytic Virotherapy; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

2022
TGF-β signaling in the tumor metabolic microenvironment and targeted therapies.
    Journal of hematology & oncology, 2022, 09-17, Volume: 15, Issue:1

    Transforming growth factor-β (TGF-β) signaling has a paradoxical role in cancer progression, and it acts as a tumor suppressor in the early stages but a tumor promoter in the late stages of cancer. Once cancer cells are generated, TGF-β signaling is responsible for the orchestration of the immunosuppressive tumor microenvironment (TME) and supports cancer growth, invasion, metastasis, recurrence, and therapy resistance. These progressive behaviors are driven by an "engine" of the metabolic reprogramming in cancer. Recent studies have revealed that TGF-β signaling regulates cancer metabolic reprogramming and is a metabolic driver in the tumor metabolic microenvironment (TMME). Intriguingly, TGF-β ligands act as an "endocrine" cytokine and influence host metabolism. Therefore, having insight into the role of TGF-β signaling in the TMME is instrumental for acknowledging its wide range of effects and designing new cancer treatment strategies. Herein, we try to illustrate the concise definition of TMME based on the published literature. Then, we review the metabolic reprogramming in the TMME and elaborate on the contribution of TGF-β to metabolic rewiring at the cellular (intracellular), tissular (intercellular), and organismal (cancer-host) levels. Furthermore, we propose three potential applications of targeting TGF-β-dependent mechanism reprogramming, paving the way for TGF-β-related antitumor therapy from the perspective of metabolism.

    Topics: Carcinogens; Humans; Ligands; Neoplasms; Transforming Growth Factor beta; Transforming Growth Factors; Tumor Microenvironment

2022
RLIP: A necessary transporter protein for translating oxidative stress into pro-obesity and pro-carcinogenic signaling.
    Biochimica et biophysica acta. Reviews on cancer, 2022, Volume: 1877, Issue:5

    Previously, we showed that knockout mice homozygous for deficiency of the mercapturic acid pathway (MAP) transporter protein, RLIP (RLIP

    Topics: Acetylcysteine; Adipokines; Animals; Carcinogenesis; Carcinogens; Carrier Proteins; Clathrin; Cytokines; Epidermal Growth Factor; GTPase-Activating Proteins; Humans; Infant; Insulins; Male; Mice; Neoplasms; Obesity; Oxidative Stress; Peptide Hormones; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2022
Tipping the scales: Immunotherapeutic strategies that disrupt immunosuppression and promote immune activation.
    Frontiers in immunology, 2022, Volume: 13

    Immunotherapy has emerged as an effective therapeutic approach for several cancer types. However, only a subset of patients exhibits a durable response due in part to immunosuppressive mechanisms that allow tumor cells to evade destruction by immune cells. One of the hallmarks of immune suppression is the paucity of tumor-infiltrating lymphocytes (TILs), characterized by low numbers of effector CD4+ and CD8+ T cells in the tumor microenvironment (TME). Additionally, the proper activation and function of lymphocytes that successfully infiltrate the tumor are hampered by the lack of co-stimulatory molecules and the increase in inhibitory factors. These contribute to the imbalance of effector functions by natural killer (NK) and T cells and the immunosuppressive functions by myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) in the TME, resulting in a dysfunctional anti-tumor immune response. Therefore, therapeutic regimens that elicit immune responses and reverse immune dysfunction are required to counter immune suppression in the TME and allow for the re-establishment of proper immune surveillance. Immuno-oncology (IO) agents, such as immune checkpoint blockade and TGF-β trapping molecules, have been developed to decrease or block suppressive factors to enable the activity of effector cells in the TME. Therapeutic agents that target immunosuppressive cells, either by direct lysis or altering their functions, have also been demonstrated to decrease the barrier to effective immune response. Other therapies, such as tumor antigen-specific vaccines and immunocytokines, have been shown to activate and improve the recruitment of CD4+ and CD8+ T cells to the tumor, resulting in improved T effector to Treg ratio. The preclinical data on these diverse IO agents have led to the development of ongoing phase I and II clinical trials. This review aims to provide an overview of select therapeutic strategies that tip the balance from immunosuppression to immune activity in the TME.

    Topics: Antigens, Neoplasm; Cancer Vaccines; Humans; Immune Checkpoint Inhibitors; Immunosuppression Therapy; Immunotherapy; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
TGF-β signaling networks in the tumor microenvironment.
    Cancer letters, 2022, 12-01, Volume: 550

    Transforming growth factor-β (TGF-β) signaling shows important roles in both physiology and pathology, especially in the progression of inflammatory diseases including cancer. Interestingly, TGF-β was first reported as a cancer suppressor, but increasing evidence confirmed its protumoral actions. Paradoxically, TGF-β can be produced by both cancer cells and stromal cells as a signaling network, which actively shapes the tumor microenvironment (TME). Surprisingly, disruption of TGF-β signaling results in both anti-cancer and pro-tumoral phenotypes in experimental cancer models, revealing the unexpected complexity of its downstream pathways for mediating cancer progression. Thus, a better understanding of the underlying mechanisms of TGF-β signaling at the molecular level can bring new insights for developing medications that can precisely separate the anti-cancer actions from the tumor-promoting outcomes. Here, we systematically summarized the latest discoveries of TGF-β signaling in cancer cells and the TME and discussed their translational implications for cancer.

    Topics: Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factors; Tumor Microenvironment

2022
TGF-β in developmental and fibrogenic EMTs.
    Seminars in cancer biology, 2022, Volume: 86, Issue:Pt 2

    TGF-β plays a prominent role as an inducer of epithelial-mesenchymal transitions (EMTs) during development and wound healing and in disease conditions such as fibrosis and cancer. During these processes EMT occurs together with changes in cell proliferation, differentiation, communication, and extracellular matrix remodeling that are orchestrated by multiple signaling inputs besides TGF-β. Chief among these inputs is RAS-MAPK signaling, which is frequently required for EMT induction by TGF-β. Recent work elucidated the molecular basis for the cooperation between the TGF-β-SMAD and RAS-MAPK pathways in the induction of EMT in embryonic, adult and carcinoma epithelial cells. These studies also provided direct mechanistic links between EMT and progenitor cell differentiation during gastrulation or intra-tumoral fibrosis during cancer metastasis. These insights illuminate the nature of TGF-β driven EMTs as part of broader processes during development, fibrogenesis and metastasis.

    Topics: Epithelial-Mesenchymal Transition; Fibrosis; Humans; Neoplasm Metastasis; Neoplasms; Transforming Growth Factor beta

2022
Integrin Alpha v Beta 6 (αvβ6) and Its Implications in Cancer Treatment.
    International journal of molecular sciences, 2022, Oct-15, Volume: 23, Issue:20

    Integrins are necessary for cell adhesion, migration, and positioning. Essential for inducing signalling events for cell survival, proliferation, and differentiation, they also trigger a variety of signal transduction pathways involved in mediating invasion, metastasis, and squamous-cell carcinoma. Several recent studies have demonstrated that the up- and down-regulation of the expression of αv and other integrins can be a potent marker of malignant diseases and patient prognosis. This review focuses on an arginine-glycine-aspartic acid (RGD)-dependent integrin αVβ6, its biology, and its role in healthy humans. We examine the implications of αVβ6 in cancer progression and the promotion of epithelial-mesenchymal transition (EMT) by contributing to the activation of transforming growth factor beta TGF-β. Although αvβ6 is crucial for proper function in healthy people, it has also been validated as a target for cancer treatment. This review briefly considers aspects of targeting αVβ6 in the clinic via different therapeutic modalities.

    Topics: Antigens, Neoplasm; Arginine; Aspartic Acid; Glycine; Humans; Integrin alphaV; Integrins; Neoplasms; Oligopeptides; Transforming Growth Factor beta

2022
Long non-coding RNA/epithelial-mesenchymal transition axis in human cancers: Tumorigenesis, chemoresistance, and radioresistance.
    Pharmacological research, 2022, Volume: 186

    Epithelial-to-mesenchymal transition (EMT) is a process that involves the transformation of polarized epithelial cells to attain a mesenchymal phenotype that presents an elevated migratory potential, invasiveness, and antiapoptotic properties. Many studies have demonstrated that EMT is a prominent event that is associated with embryogenesis, tumor progression, metastasis, and therapeutic resistance. The EMT process is driven by key transcription factors (such as Snail, Twist, ZEB, and TGF-β) and several long non-coding RNAs (lncRNAs) in many non-pathological as well as pathological conditions. In the present report, we have comprehensively discussed the oncogenic and tumor suppressor role of lncRNAs and their mechanism of action in the regulation of the EMT process in various cancers such as brain tumors, gastrointestinal tumors, and gynecological and urological tumors. We have also elaborated on the role of lncRNAs in the regulation of EMT-related transcription factors (such as Snail, Twist, ZEB, and TGF-β) and therapeutic response (chemoresistance and radioresistance). Lastly, we have emphasized the role of exosomal lncRNAs in the regulation of EMT, metastasis, and therapeutic response in the aforementioned cancers. Taken together, this review provides a detailed insight into the understanding of role of lncRNAs/exosomal lncRNAs in EMT, metastasis, and therapeutic response in human cancers.

    Topics: Carcinogenesis; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; RNA, Long Noncoding; Transcription Factors; Transforming Growth Factor beta

2022
Importance of TGFβ in Cancer and Nematode Infection and Their Interaction-Opinion.
    Biomolecules, 2022, 10-26, Volume: 12, Issue:11

    Historically, there has been little interaction between parasitologists and oncologists, although some helminth infections predispose to the development of tumours. In addition, both parasites and tumours need to survive immune attack. Recent research suggests that both tumours and parasites suppress the immune response to increase their chances of survival. They both co-opt the transforming growth factor beta (TGFβ) signalling pathway to modulate the immune response to their benefit. In particular, there is concern that suppression of the immune response by nematodes and their products could enhance susceptibility to tumours in both natural and artificial infections.

    Topics: Humans; Nematode Infections; Neoplasms; Transforming Growth Factor beta

2022
The difficulty in translating the preclinical success of combined TGFβ and immune checkpoint inhibition to clinical trial.
    EBioMedicine, 2022, Volume: 86

    Immune checkpoint inhibitors (ICIs) have transformed the treatment paradigm for solid tumors. However, even in cancers generally considered ICI-sensitive, responses can vary significantly. Thus, there is an ever-increasing interest in identifying novel means of improving therapeutic responses, both for cancers in which ICIs are indicated and those for which they have yet to show significant anti-tumor activity. To this end, Transforming Growth Factor β (TGFβ) signaling is emerging as an important barrier to the efficacy of ICIs. Accordingly, several preclinical studies now support the use of combined TGFβ and immune checkpoint blockade, with near-uniform positive results across a wide range of tumor types. However, as these approaches have started to emerge in clinical trials, the addition of TGFβ inhibitors has often failed to show a meaningful benefit beyond the current generation of ICIs alone. Here, we summarize landmark clinical studies exploring combined TGFβ and immune checkpoint blockade. These studies not only reinforce the difficulty in translating results from rodents to clinical trials in immune-oncology but also underscore the need to re-evaluate the design of trials exploring this approach, incorporating both mechanism-driven combination strategies and novel, predictive biomarkers to identify the patients most likely to derive clinical benefit.

    Topics: Clinical Trials as Topic; Humans; Immune Checkpoint Inhibitors; Immunotherapy; Neoplasms; Transforming Growth Factor beta

2022
New insight into GARP striking role in cancer progression: application for cancer therapy.
    Medical oncology (Northwood, London, England), 2022, Dec-02, Volume: 40, Issue:1

    T regulatory cells play a crucial role in antitumor immunity suppression. Glycoprotein-A repetitions predominant (GARP), transmembrane cell surface marker, is mostly expressed on Tregs and mediates intracellular organization of transforming growth factor-beta (TGF-β). The physiological role of GARP is immune system homeostasis, while it may cause tumor development by upregulating TGF-β secretion. Despite the vast application of anti- programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte Antigen-4 (CTLA-4) antibodies in immunotherapy, anti-GARP antibodies have the advantage of better response in patients who has resistance to anti-PD-1/PD-L1. Furthermore, simultaneous administration of anti-GARP antibody and anti-PD-1/PD-L1 antibody is much more effective than anti-PD-1/PD-L1 alone. It is worth mentioning that the GARP-mTGF-β complex is more potent than secretory TGF-β to induce T helper 17 cells differentiation in HIV + patients. On the other hand, TGF-β is an effective cytokine in cancer development, and some microRNAs could control its secretion by regulating GARP. In the present review, some information is provided about the undeniable role of GARP in cancer progression and its probable importance as a novel prognostic biomarker. Anti-GARP antibodies are also suggested for cancer immunotherapy.

    Topics: B7-H1 Antigen; Glycoproteins; Humans; Immunotherapy; Membrane Proteins; Neoplasms; Transforming Growth Factor beta

2022
Mechanism of Extracellular Vesicle Secretion Associated with TGF-β-Dependent Inflammatory Response in the Tumor Microenvironment.
    International journal of molecular sciences, 2022, Dec-05, Volume: 23, Issue:23

    Extracellular vesicles (EVs) serve as central mediators in communication between tumor and non-tumor cells. These interactions are largely dependent on the function of the endothelial barrier and the set of receptors present on its surface, as endothelial cells (ECs) are a plenteous source of EVs. The molecular basis for EV secretion and action in the tumor microenvironment (TME) has not been fully elucidated to date. Emerging evidence suggests a prominent role of inflammatory pathways in promoting tumor progression and metastasis. Although transforming growth factor β (TGF-β) is a cytokine with strong immunomodulatory and protective activity in benign and early-stage cancer cells, it plays a pro-tumorigenic role in advanced cancer cells, which is known as the "TGF-β paradox". Thus, the aim of this review is to describe the correlation between EV release, TGF-β-dependent inflammation, and dysregulation of downstream TGF-β signaling in the context of cancer development.

    Topics: Endothelial Cells; Extracellular Vesicles; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2022
TGF-β: A novel predictor and target for anti-PD-1/PD-L1 therapy.
    Frontiers in immunology, 2022, Volume: 13

    Transforming growth factor-β (TGF-β) signaling regulates multiple physiological processes, such as cell proliferation, differentiation, immune homeostasis, and wound healing. Besides, TGF-β plays a vital role in diseases, including cancer. Accumulating evidence indicates that TGF-β controls the composition and behavior of immune components in the tumor microenvironment (TME). Advanced cancers leverage TGF-β to reshape the TME and escape immune surveillance. TGF-β-mediated immune evasion is an unfavorable factor for cancer immunotherapy, especially immune checkpoint inhibitors (ICI). Numerous preclinical and clinical studies have demonstrated that hyperactive TGF-β signaling is closely associated with ICI resistance. It has been validated that TGF-β blockade synergizes with ICI and overcomes treatment resistance. TGF-β-targeted therapies, including trap and bispecific antibodies, have shown immense potential for cancer immunotherapy. In this review, we summarized the predictive value of TGF-β signaling and the prospects of TGF-β-targeted therapies for cancer immunotherapy.

    Topics: B7-H1 Antigen; Humans; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2022
TGF-β superfamily co-receptors in cancer.
    Developmental dynamics : an official publication of the American Association of Anatomists, 2022, Volume: 251, Issue:1

    Transforming growth factor-β (TGF-β) superfamily signaling via their cognate receptors is frequently modified by TGF-β superfamily co-receptors. Signaling through SMAD-mediated pathways may be enhanced or depressed depending on the specific co-receptor and cell context. This dynamic effect on signaling is further modified by the release of many of the co-receptors from the membrane to generate soluble forms that are often antagonistic to the membrane-bound receptors. The co-receptors discussed here include TβRIII (betaglycan), endoglin, BAMBI, CD109, SCUBE proteins, neuropilins, Cripto-1, MuSK, and RGMs. Dysregulation of these co-receptors can lead to altered TGF-β superfamily signaling that contributes to the pathophysiology of many cancers through regulation of growth, metastatic potential, and the tumor microenvironment. Here we describe the role of several TGF-β superfamily co-receptors on TGF-β superfamily signaling and the impact on cellular and physiological functions with a particular focus on cancer, including a discussion on recent pharmacological advances and potential clinical applications targeting these co-receptors.

    Topics: Humans; Neoplasms; Phosphorylation; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2022
The TGF-β/NADPH Oxidases Axis in the Regulation of Liver Cell Biology in Health and Disease.
    Cells, 2021, 09-03, Volume: 10, Issue:9

    The Transforming Growth Factor-beta (TGF-β) pathway plays essential roles in liver development and homeostasis and become a relevant factor involved in different liver pathologies, particularly fibrosis and cancer. The family of NADPH oxidases (NOXs) has emerged in recent years as targets of the TGF-β pathway mediating many of its effects on hepatocytes, stellate cells and macrophages. This review focuses on how the axis TGF-β/NOXs may regulate the biology of different liver cells and how this influences physiological situations, such as liver regeneration, and pathological circumstances, such as liver fibrosis and cancer. Finally, we discuss whether NOX inhibitors may be considered as potential therapeutic tools in liver diseases.

    Topics: Animals; Humans; Liver; Liver Cirrhosis; Liver Regeneration; NADPH Oxidases; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2021
TGF-beta signaling in cancer radiotherapy.
    Cytokine, 2021, Volume: 148

    Transforming growth factor beta (TGF-β) plays key roles in regulating cellular proliferation and maintaining tissue homeostasis. TGF-β exerts tumor-suppressive effects in the early stages of carcinogenesis, but it also plays tumor-promoting roles in established tumors. Additionally, it plays a critical role in cancer radiotherapy. TGF-β expression or activation increases in irradiated tissues, and studies have shown that TGF-β plays dual roles in cancer radiosensitivity and is involved in ionizing radiation-induced fibrosis in different tumor microenvironments (TMEs). Furthermore, TGF-β promotes radioresistance by inducing the epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs) and cancer-associated fibroblasts (CAFs), suppresses the immune system and facilitates cancer resistance. In particular, the links between TGF-β and the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) axis play a critical role in cancer therapeutic resistance. Growing evidence has shown that TGF-β acts as a radiation protection agent, leading to heightened interest in using TGF-β as a therapeutic target. The future of anti-TGF-β signaling therapy for numerous diseases appears bright, and the outlook for the use of TGF-β inhibitors in cancer radiotherapy as TME-targeting agents is promising.

    Topics: Drug Resistance, Neoplasm; Fibrosis; Humans; Models, Biological; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2021
The tumorigenic function of LINC00858 in cancer.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2021, Volume: 143

    Long non-coding RNA (lncRNA) plays an important regulatory role in the occurrence and development of human cancer. LINC00858 is a newly discovered lncRNA with a length of 2685 nucleotides. Existing studies have shown that LINC00858 has abnormally high expression levels in malignant tumors such as colorectal cancer, gastric cancer, hepatocellular carcinoma, lung cancer, non-small cell lung cancer, ovarian cancer, osteosarcoma, retinoblastoma, Wilms tumor, bladder cancer, and cervical cancer. By regulating a variety of microRNAs, LINC00858 can affect tumor cell proliferation, invasion, metastasis, and apoptosis. Related research also found that LINC00858 is related to nuclear transcription factor/protein kinase and gene methylation. The aberrant expression of LINC00858 is related to the prognosis and clinicopathological characteristics of a variety of tumors. Overexpressed LINC00858 is closely related to the clinical stage, lymph node metastasis, and distant metastasis of cancer, including colorectal cancer, gastric cancer, non-small cell lung cancer, ovarian cancer, and Wilms tumor. Also, it is summarized that LINC00858 can regulate MAPK and TGF-β signaling pathways. This review shows that LINC00858 as an important oncogene can promote tumorigenesis and cancer development.

    Topics: Animals; Cell Transformation, Neoplastic; Disease Progression; Gene Expression Regulation, Neoplastic; Gene Regulatory Networks; Humans; MicroRNAs; Mitogen-Activated Protein Kinases; Neoplasms; RNA, Long Noncoding; Signal Transduction; Transforming Growth Factor beta

2021
Correlation between Oxidative Stress and Transforming Growth Factor-Beta in Cancers.
    International journal of molecular sciences, 2021, Dec-07, Volume: 22, Issue:24

    The downregulation of reactive oxygen species (ROS) facilitates precancerous tumor development, even though increasing the level of ROS can promote metastasis. The transforming growth factor-beta (TGF-β) signaling pathway plays an anti-tumorigenic role in the initial stages of cancer development but a pro-tumorigenic role in later stages that fosters cancer metastasis. TGF-β can regulate the production of ROS unambiguously or downregulate antioxidant systems. ROS can influence TGF-β signaling by enhancing its expression and activation. Thus, TGF-β signaling and ROS might significantly coordinate cellular processes that cancer cells employ to expedite their malignancy. In cancer cells, interplay between oxidative stress and TGF-β is critical for tumorigenesis and cancer progression. Thus, both TGF-β and ROS can develop a robust relationship in cancer cells to augment their malignancy. This review focuses on the appropriate interpretation of this crosstalk between TGF-β and oxidative stress in cancer, exposing new potential approaches in cancer biology.

    Topics: Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Oxidative Stress; Reactive Oxygen Species; Signal Transduction; Transforming Growth Factor beta

2021
Duality of Interactions Between TGF-β and TNF-α During Tumor Formation.
    Frontiers in immunology, 2021, Volume: 12

    The tumor microenvironment is essential for the formation and development of tumors. Cytokines in the microenvironment may affect the growth, metastasis and prognosis of tumors, and play different roles in different stages of tumors, of which transforming growth factor β (TGF-β) and tumor necrosis factor α (TNF-α) are critical. The two have synergistic and antagonistic effect on tumor regulation. The inhibition of TGF-β can promote the formation rate of tumor, while TGF-β can promote the malignancy of tumor. TNF-α was initially determined to be a natural immune serum mediator that can induce tumor hemorrhagic necrosis, it has a wide range of biological activities and can be used clinically as a target to immune diseases as well as tumors. However, there are few reports on the interaction between the two in the tumor microenvironment. This paper combs the biological effect of the two in different aspects of different tumors. We summarized the changes and clinical medication rules of the two in different tissue cells, hoping to provide a new idea for the clinical application of the two cytokines.

    Topics: Animals; Apoptosis; Biomarkers; Biomarkers, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Disease Susceptibility; Epithelial-Mesenchymal Transition; Gene Expression Regulation; Genomic Instability; Humans; Neoplasms; Protein Binding; Signal Transduction; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2021
TGFβ biology in cancer progression and immunotherapy.
    Nature reviews. Clinical oncology, 2021, Volume: 18, Issue:1

    TGFβ signalling has key roles in cancer progression: most carcinoma cells have inactivated their epithelial antiproliferative response and benefit from increased TGFβ expression and autocrine TGFβ signalling through effects on gene expression, release of immunosuppressive cytokines and epithelial plasticity. As a result, TGFβ enables cancer cell invasion and dissemination, stem cell properties and therapeutic resistance. TGFβ released by cancer cells, stromal fibroblasts and other cells in the tumour microenvironment further promotes cancer progression by shaping the architecture of the tumour and by suppressing the antitumour activities of immune cells, thus generating an immunosuppressive environment that prevents or attenuates the efficacy of anticancer immunotherapies. The repression of TGFβ signalling is therefore considered a prerequisite and major avenue to enhance the efficacy of current and forthcoming immunotherapies, including in tumours comprising cancer cells that are not TGFβ responsive. Herein, we introduce the mechanisms underlying TGFβ signalling in tumours and their microenvironment and discuss approaches to inhibit these signalling mechanisms as well as the use of these approaches in cancer immunotherapies and their potential adverse effects.

    Topics: Animals; Disease Progression; Humans; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2021
TGFβ-Directed Therapeutics: 2020.
    Pharmacology & therapeutics, 2021, Volume: 217

    The transforming growth factor-beta (TGFβ) pathway is essential during embryo development and in maintaining normal homeostasis. During malignancy, the TGFβ pathway is co-opted by the tumor to increase fibrotic stroma, to promote epithelial to mesenchymal transition increasing metastasis and producing an immune-suppressed microenvironment which protects the tumor from recognition by the immune system. Compelling preclinical data demonstrate the therapeutic potential of blocking TGFβ function in cancer. However, the TGFβ pathway cannot be described as a driver of malignant disease. Two small molecule kinase inhibitors which block the serine-threonine kinase activity of TGFβRI on TGFβRII, a pan-TGFβ neutralizing antibody, a TGFβ trap, a TGFβ antisense agent, an antibody which stabilizes the latent complex of TGFβ and a fusion protein which neutralizes TGFβ and binds PD-L1 are in clinical development. The challenge is how to most effectively incorporate blocking TGFβ activity alone and in combination with other therapeutics to improve treatment outcome.

    Topics: Animals; B7-H1 Antigen; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Transforming Growth Factor beta; Tumor Microenvironment

2021
TGFβ Signaling in the Tumor Microenvironment.
    Advances in experimental medicine and biology, 2021, Volume: 1270

    Transforming growth factor beta (TGFβ) is a pleiotropic growth factor. Under normal physiological conditions, TGFβ maintains homeostasis in mammalian tissues by restraining the growth of cells and stimulating apoptosis. However, the role of TGFβ signaling in the carcinogenesis is complex. TGFβ acts as a tumor suppressor in the early stages of disease and as a tumor promoter in its later stages where cancer cells have been relieved from TGFβ growth controls. Overproduction of TGFβ by cancer cells lead to a local fibrotic and immune-suppressive microenvironment that fosters tumor growth and correlates with invasive and metastatic behavior of the cancer cells. Here, we present an overview of the complex biology of the TGFβ family, and we discuss the roles of TGFβ signaling in carcinogenesis and how this knowledge is being leveraged to develop TGFβ inhibition therapies against the tumor.

    Topics: Carcinogenesis; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2021
Exploring the extensive crosstalk between the antagonistic cytokines- TGF-β and TNF-α in regulating cancer pathogenesis.
    Cytokine, 2021, Volume: 138

    A plethora of cytokines are produced in the tumor microenvironment (TME) those play a vital role in cancer prognosis. Though it is completely contextual, cytokines produced from an inflammatory micro-environment can either modulate cancer progression at early stages of tumor development or in later stages cytokine derived cues can in turn control tumor cell invasion and metastasis. Therefore, understanding the crosstalk between the key cytokines regulating cancer prognosis is critical for the development of an effective therapy. In this regard, the role of transforming growth factor-beta (TGF-β) in cancer is controversially discussed in general inhibition of TGF-β promotes de novo tumorigenesis whereas paradoxically, TGF-β can promote malignancy in already established tumors. Another important cytokine, TNF-α have intense crosstalk with TGF-β from the fact that in a non-cancer context, TGF-β promotes fibrosis whereas TNF-α has anti-fibrotic activity. We have recently reported that TGF-β-induced differentiation of epithelial cells to mesenchymal type is suppressed by TNF-α through regulation of cellular homeostatic machinery- autophagy. Moreover, there are also rare reports of synergy between these two cytokines as well. The crosstalk between TGF-β and TNF-α is not only limited to regulating cancer cell differentiation and proliferation but also includes involvement in cell death. In this review, we hence summarize the molecular mechanisms by which these two important cytokines, TGF-β and TNF-α control cancer prognosis.

    Topics: Animals; Apoptosis; Autophagy; Cell Death; Cell Differentiation; Cell Proliferation; Cellular Senescence; Cytokines; Epithelial-Mesenchymal Transition; Homeostasis; Humans; Inflammation; Mice; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Prognosis; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2021
eIF3i regulation of protein synthesis, cell proliferation, cell cycle progression, and tumorigenesis.
    Cancer letters, 2021, 03-01, Volume: 500

    eIF3i, a 36-kDa protein, is a putative subunit of the eIF3 complex important for translation initiation of mRNAs. It is a WD40 domain-containing protein with seven WD40 repeats that forms a β-propeller structure with an important function in pre-initiation complex formation and mRNA translation initiation. In addition to participating in the eIF3 complex formation for global translational control, eIF3i may bind to specific mRNAs and regulate their translation individually. Furthermore, eIF3i has been shown to bind to TGF-β type II receptor and participate in TGF-β signaling. It may also participate in and regulate other signaling pathways including Wnt/β-catenin pathway via translational regulation of COX-2 synthesis. These multiple canonical and noncanonical functions of eIF3i in translational control and in regulating signal transduction pathways may be responsible for its role in cell differentiation, cell cycle regulation, proliferation, and tumorigenesis. In this review, we will critically evaluate recent progresses and assess future prospects in studying eIF3i.

    Topics: Carcinogenesis; Cell Cycle; Cell Proliferation; Eukaryotic Initiation Factor-3; Humans; Neoplasm Proteins; Neoplasms; Receptor, Transforming Growth Factor-beta Type II; Transforming Growth Factor beta; WD40 Repeats; Wnt Signaling Pathway

2021
Recent progress in TGF-β inhibitors for cancer therapy.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2021, Volume: 134

    Transforming growth factor-β (TGF-β) is a multifunctional cytokine that is involved in proliferation, metastasis, and many other important processes in malignancy. Inhibitors targeting TGF-β have been considered by pharmaceutical companies for cancer therapy, and some of them are in clinical trial now. Unfortunately, several of these programs have recently been relinquished, and most companies that remain in the contest are progressing slowly and cautiously. This review summarizes the TGF-β signal transduction pathway, its roles in oncogenesis and fibrotic diseases, and advancements in antibodies and small-molecule inhibitors of TGF-β.

    Topics: Animals; Antibodies, Neutralizing; Antineoplastic Agents; Aptamers, Peptide; Cancer Vaccines; Carcinogenesis; Clinical Trials as Topic; Endocytosis; Fibrosis; Humans; Membrane Microdomains; Mice; Neoplasms; Oligonucleotides, Antisense; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2021
Targeting transforming growth factor-β signaling for enhanced cancer chemotherapy.
    Theranostics, 2021, Volume: 11, Issue:3

    During the past decades, drugs targeting transforming growth factor-β (TGFβ) signaling have received tremendous attention for late-stage cancer treatment since TGFβ signaling has been recognized as a prime driver for tumor progression and metastasis. Nonetheless, in healthy and pre-malignant tissues, TGFβ functions as a potent tumor suppressor. Furthermore, TGFβ signaling plays a key role in normal development and homeostasis by regulating cell proliferation, differentiation, migration, apoptosis, and immune evasion, and by suppressing tumor-associated inflammation. Therefore, targeting TGFβ signaling for cancer therapy is challenging. Recently, we and others showed that blocking TGFβ signaling increased chemotherapy efficacy, particularly for nanomedicines. In this review, we briefly introduce the TGFβ signaling pathway, and the multifaceted functions of TGFβ signaling in cancer, including regulating the tumor microenvironment (TME) and the behavior of cancer cells. We also summarize TGFβ targeting agents. Then, we highlight TGFβ inhibition strategies to restore the extracellular matrix (ECM), regulate the tumor vasculature, reverse epithelial-mesenchymal transition (EMT), and impair the stemness of cancer stem-like cells (CSCs) to enhance cancer chemotherapy efficacy. Finally, the current challenges and future opportunities in targeting TGFβ signaling for cancer therapy are discussed.

    Topics: Animals; Antineoplastic Agents; Apoptosis; Cell Movement; Epithelial-Mesenchymal Transition; Extracellular Matrix; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2021
Therapeutic targeting of TGF-β in cancer: hacking a master switch of immune suppression.
    Clinical science (London, England : 1979), 2021, 01-15, Volume: 135, Issue:1

    Cancers may escape elimination by the host immune system by rewiring the tumour microenvironment towards an immune suppressive state. Transforming growth factor-β (TGF-β) is a secreted multifunctional cytokine that strongly regulates the activity of immune cells while, in parallel, can promote malignant features such as cancer cell invasion and migration, angiogenesis, and the emergence of cancer-associated fibroblasts. TGF-β is abundantly expressed in cancers and, most often, its abundance associated with poor clinical outcomes. Immunotherapeutic strategies, particularly T cell checkpoint blockade therapies, so far, only produce clinical benefit in a minority of cancer patients. The inhibition of TGF-β activity is a promising approach to increase the efficacy of T cell checkpoint blockade therapies. In this review, we briefly outline the immunoregulatory functions of TGF-β in physiological and malignant contexts. We then deliberate on how the therapeutic targeting of TGF-β may lead to a broadened applicability and success of state-of-the-art immunotherapies.

    Topics: Animals; Humans; Immune Checkpoint Inhibitors; Immunity, Innate; Immunotherapy; Integrins; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2021
Targeting TGFβ signal transduction for cancer therapy.
    Signal transduction and targeted therapy, 2021, 01-08, Volume: 6, Issue:1

    Transforming growth factor-β (TGFβ) family members are structurally and functionally related cytokines that have diverse effects on the regulation of cell fate during embryonic development and in the maintenance of adult tissue homeostasis. Dysregulation of TGFβ family signaling can lead to a plethora of developmental disorders and diseases, including cancer, immune dysfunction, and fibrosis. In this review, we focus on TGFβ, a well-characterized family member that has a dichotomous role in cancer progression, acting in early stages as a tumor suppressor and in late stages as a tumor promoter. The functions of TGFβ are not limited to the regulation of proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and metastasis of cancer cells. Recent reports have related TGFβ to effects on cells that are present in the tumor microenvironment through the stimulation of extracellular matrix deposition, promotion of angiogenesis, and suppression of the anti-tumor immune reaction. The pro-oncogenic roles of TGFβ have attracted considerable attention because their intervention provides a therapeutic approach for cancer patients. However, the critical function of TGFβ in maintaining tissue homeostasis makes targeting TGFβ a challenge. Here, we review the pleiotropic functions of TGFβ in cancer initiation and progression, summarize the recent clinical advancements regarding TGFβ signaling interventions for cancer treatment, and discuss the remaining challenges and opportunities related to targeting this pathway. We provide a perspective on synergistic therapies that combine anti-TGFβ therapy with cytotoxic chemotherapy, targeted therapy, radiotherapy, or immunotherapy.

    Topics: Carcinogenesis; Epithelial-Mesenchymal Transition; Humans; Neoplasm Proteins; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2021
E3 Ubiquitin Ligases: Key Regulators of TGFβ Signaling in Cancer Progression.
    International journal of molecular sciences, 2021, Jan-06, Volume: 22, Issue:2

    Transforming growth factor β (TGFβ) is a secreted growth and differentiation factor that influences vital cellular processes like proliferation, adhesion, motility, and apoptosis. Regulation of the TGFβ signaling pathway is of key importance to maintain tissue homeostasis. Perturbation of this signaling pathway has been implicated in a plethora of diseases, including cancer. The effect of TGFβ is dependent on cellular context, and TGFβ can perform both anti- and pro-oncogenic roles. TGFβ acts by binding to specific cell surface TGFβ type I and type II transmembrane receptors that are endowed with serine/threonine kinase activity. Upon ligand-induced receptor phosphorylation, SMAD proteins and other intracellular effectors become activated and mediate biological responses. The levels, localization, and function of TGFβ signaling mediators, regulators, and effectors are highly dynamic and regulated by a myriad of post-translational modifications. One such crucial modification is ubiquitination. The ubiquitin modification is also a mechanism by which crosstalk with other signaling pathways is achieved. Crucial effector components of the ubiquitination cascade include the very diverse family of E3 ubiquitin ligases. This review summarizes the diverse roles of E3 ligases that act on TGFβ receptor and intracellular signaling components. E3 ligases regulate TGFβ signaling both positively and negatively by regulating degradation of receptors and various signaling intermediates. We also highlight the function of E3 ligases in connection with TGFβ's dual role during tumorigenesis. We conclude with a perspective on the emerging possibility of defining E3 ligases as drug targets and how they may be used to selectively target TGFβ-induced pro-oncogenic responses.

    Topics: Animals; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Ubiquitin-Protein Ligases; Ubiquitination

2021
Autocrine TGF-β in Cancer: Review of the Literature and Caveats in Experimental Analysis.
    International journal of molecular sciences, 2021, Jan-19, Volume: 22, Issue:2

    Autocrine signaling is defined as the production and secretion of an extracellular mediator by a cell followed by the binding of that mediator to receptors on the same cell to initiate signaling. Autocrine stimulation often operates in autocrine loops, a type of interaction, in which a cell produces a mediator, for which it has receptors, that upon activation promotes expression of the same mediator, allowing the cell to repeatedly autostimulate itself (positive feedback) or balance its expression via regulation of a second factor that provides negative feedback. Autocrine signaling loops with positive or negative feedback are an important feature in cancer, where they enable context-dependent cell signaling in the regulation of growth, survival, and cell motility. A growth factor that is intimately involved in tumor development and progression and often produced by the cancer cells in an autocrine manner is transforming growth factor-β (TGF-β). This review surveys the many observations of autocrine TGF-β signaling in tumor biology, including data from cell culture and animal models as well as from patients. We also provide the reader with a critical discussion on the various experimental approaches employed to identify and prove the involvement of autocrine TGF-β in a given cellular response.

    Topics: Animals; Autocrine Communication; Biomedical Research; Feedback, Physiological; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Receptors, Transforming Growth Factor beta; Research Design; Transforming Growth Factor beta

2021
Translational Regulation of Cancer Metastasis.
    Cancer research, 2021, 02-01, Volume: 81, Issue:3

    Deregulation of the mRNA translational process has been observed during tumorigenesis. However, recent findings have shown that deregulation of translation also contributes specifically to cancer cell spread. During metastasis, cancer cells undergo changes in cellular state, permitting the acquisition of features necessary for cell survival, dissemination, and outgrowth. In addition, metastatic cells respond to external cues, allowing for their persistence under significant cellular and microenvironmental stresses. Recent work has revealed the importance of mRNA translation to these dynamic changes, including regulation of cell states through epithelial-to-mesenchymal transition and tumor dormancy and as a response to external stresses such as hypoxia and immune surveillance. In this review, we focus on examples of altered translation underlying these phenotypic changes and responses to external cues and explore how they contribute to metastatic progression. We also highlight the therapeutic opportunities presented by aberrant mRNA translation, suggesting novel ways to target metastatic tumor cells.

    Topics: Carcinogenesis; Cell Movement; Cell Survival; Epithelial-Mesenchymal Transition; Humans; Neoplasm Metastasis; Neoplasm Proteins; Neoplasms; Neovascularization, Pathologic; Phenotype; Protein Biosynthesis; TOR Serine-Threonine Kinases; Transforming Growth Factor beta; Tumor Escape; Tumor Hypoxia; Tumor Microenvironment

2021
Molecular Mechanisms of Epithelial to Mesenchymal Transition Regulated by ERK5 Signaling.
    Biomolecules, 2021, 01-29, Volume: 11, Issue:2

    Extracellular signal-regulated kinase (ERK5) is an essential regulator of cancer progression, tumor relapse, and poor patient survival. Epithelial to mesenchymal transition (EMT) is a complex oncogenic process, which drives cell invasion, stemness, and metastases. Activators of ERK5, including mitogen-activated protein kinase 5 (MEK5), tumor necrosis factor α (TNF-α), and transforming growth factor-β (TGF-β), are known to induce EMT and metastases in breast, lung, colorectal, and other cancers. Several downstream targets of the ERK5 pathway, such as myocyte-specific enhancer factor 2c (MEF2C), activator protein-1 (AP-1), focal adhesion kinase (FAK), and c-Myc, play a critical role in the regulation of EMT transcription factors SNAIL, SLUG, and β-catenin. Moreover, ERK5 activation increases the release of extracellular matrix metalloproteinases (MMPs), facilitating breakdown of the extracellular matrix (ECM) and local tumor invasion. Targeting the ERK5 signaling pathway using small molecule inhibitors, microRNAs, and knockdown approaches decreases EMT, cell invasion, and metastases via several mechanisms. The focus of the current review is to highlight the mechanisms which are known to mediate cancer EMT via ERK5 signaling. Several therapeutic approaches that can be undertaken to target the ERK5 pathway and inhibit or reverse EMT and metastases are discussed.

    Topics: Animals; Cell Adhesion; Cytoskeleton; Disease Progression; Epithelial-Mesenchymal Transition; Extracellular Matrix; Humans; Mitogen-Activated Protein Kinase 7; Mutation; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neoplastic Stem Cells; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor-alpha

2021
Actin Cytoskeleton and Regulation of TGFβ Signaling: Exploring Their Links.
    Biomolecules, 2021, 02-23, Volume: 11, Issue:2

    Human tissues, to maintain their architecture and function, respond to injuries by activating intricate biochemical and physical mechanisms that regulates intercellular communication crucial in maintaining tissue homeostasis. Coordination of the communication occurs through the activity of different actin cytoskeletal regulators, physically connected to extracellular matrix through integrins, generating a platform of biochemical and biomechanical signaling that is deregulated in cancer. Among the major pathways, a controller of cellular functions is the cytokine transforming growth factor β (TGFβ), which remains a complex and central signaling network still to be interpreted and explained in cancer progression. Here, we discuss the link between actin dynamics and TGFβ signaling with the aim of exploring their aberrant interaction in cancer.

    Topics: Actin Cytoskeleton; Actins; Animals; Biomechanical Phenomena; Cytokines; Disease Progression; Extracellular Matrix; Homeostasis; Humans; Integrins; Ligands; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2021
Epithelial plasticity, epithelial-mesenchymal transition, and the TGF-β family.
    Developmental cell, 2021, 03-22, Volume: 56, Issue:6

    Epithelial cells repress epithelial characteristics and elaborate mesenchymal characteristics to migrate to other locations and acquire new properties. Epithelial plasticity responses are directed through cooperation of signaling pathways, with TGF-β and TGF-β-related proteins playing prominent instructive roles. Epithelial-mesenchymal transitions (EMTs) directed by activin-like molecules, bone morphogenetic proteins, or TGF-β regulate metazoan development and wound healing and drive fibrosis and cancer progression. In carcinomas, diverse EMTs enable stem cell generation, anti-cancer drug resistance, genomic instability, and localized immunosuppression. This review discusses roles of TGF-β and TGF-β-related proteins, and underlying molecular mechanisms, in epithelial plasticity in development and wound healing, fibrosis, and cancer.

    Topics: Animals; Epithelial Cells; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Transforming Growth Factor beta

2021
TGF-β: Many Paths to CD103
    Cells, 2021, 04-23, Volume: 10, Issue:5

    CD8 tissue-resident memory T (T

    Topics: Antigens, CD; CD8-Positive T-Lymphocytes; Humans; Immunologic Memory; Integrin alpha Chains; Lymphocyte Activation; Neoplasms; Transforming Growth Factor beta

2021
The role of the extracellular matrix protein TGFBI in cancer.
    Cellular signalling, 2021, Volume: 84

    The secreted extracellular protein, transforming growth factor beta induced (TGFBI or βIGH3), has roles in regulating numerous biological functions, including cell adhesion and bone formation, both during embryonic development and during the pathogenesis of human disease. TGFBI has been most studied in the context of hereditary corneal dystrophies, where mutations in TGFBI result in accumulation of TGFBI in the cornea. In cancer, early studies focused on TGFBI as a tumor suppressor, in part by promoting chemotherapy sensitivity. However, in established tumors, TGFBI largely has a role in promoting tumor progression, with elevated levels correlating to poorer clinical outcomes. As an important regulator of cancer progression, TGFBI expression and function is tightly regulated by numerous mechanisms including epigenetic silencing through promoter methylation and microRNAs. Mechanisms to target TGFBI have potential clinical utility in treating advanced cancers, while assessing TGFBI levels could be a biomarker for chemotherapy resistance and tumor progression.

    Topics: Corneal Dystrophies, Hereditary; Epigenesis, Genetic; Extracellular Matrix Proteins; Humans; Neoplasms; Transforming Growth Factor beta

2021
The outstanding role of miR-132-3p in carcinogenesis of solid tumors.
    Human cell, 2021, Volume: 34, Issue:4

    MicroRNAs are a group of short non-coding RNAs (miRNAs), which are epigenetically involved in gene expression and other cellular biological processes and can be considered as potential biomarkers for cancer detection and support for treatment management. This review aims to amass the evidence to reach the molecular mechanism and clinical significance of miR-132 in different types of cancer. Dysregulation of miR-132 level in various types of malignancies, including hepatocellular carcinoma, breast cancer, colorectal cancer, gastric cancer, lung cancer, prostate cancer, osteosarcoma, pancreatic cancer, and ovarian cancer have reported, significantly decrease in its level, which can be indicated to its function as a tumor suppressor. miR-132 is involved in cell proliferation, migration, and invasion through cell cycle pathways, such as PI3K, TGFβ or hippo signaling pathways, or on oncogenes such as Ras, AKT, mTOR, glycolysis. miR-132 could be potentially a candidate as a valuable biomarker for prognosis in various cancers. Through this study, we proposed that miR-132 can potentially be a candidate as a prognostic marker for early detection of tumor development, progression, as well as metastasis.

    Topics: Biomarkers, Tumor; Carcinogenesis; Cell Cycle; Cell Movement; Cell Proliferation; Humans; MicroRNAs; Neoplasm Invasiveness; Neoplasms; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; ras Proteins; Signal Transduction; TOR Serine-Threonine Kinases; Transforming Growth Factor beta

2021
Emerging strategies for sensitization of therapy resistant tumors toward cancer therapeutics by targeting the Bcl-2 family, TGF-β, Wnt/β-Catenin, RASSF and miRNA regulated signaling pathways.
    The international journal of biochemistry & cell biology, 2021, Volume: 137

    Conventional chemotherapy relies on the cytotoxicity of chemo-drugs to inflict destructive effects on tumor cells. However, as most tumor cells develop resistance to chemo-drugs, small doses of chemo-drugs are unlikely to provide significant clinical benefits in cancer treatment while high doses of chemo-drugs have been shown to impact normal human cells negatively due to the non-specific nature and cytotoxicity associated with chemo-drugs. To overcome this challenge, sensitizations of tumor cells with bioactive molecules that specifically target the pro-survival and pro-apoptosis signaling pathways of the tumor cells are likely to increase the therapeutic impacts and improve the clinical outcomes by reducing the dependency and adverse effects associated with using high doses of chemo-drugs in cancer treatment. This review focuses on emerging strategies to enhance the sensitization of tumor cells toward cancer therapies based on our understanding of tumor cell biology and underlying signaling pathways.

    Topics: Animals; Antineoplastic Agents; beta Catenin; Drug Resistance, Neoplasm; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Neoplasms; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor beta; Wnt Signaling Pathway

2021
Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer.
    Signal transduction and targeted therapy, 2021, 06-10, Volume: 6, Issue:1

    To flourish, cancers greatly depend on their surrounding tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in TME are critical for cancer occurrence and progression because of their versatile roles in extracellular matrix remodeling, maintenance of stemness, blood vessel formation, modulation of tumor metabolism, immune response, and promotion of cancer cell proliferation, migration, invasion, and therapeutic resistance. CAFs are highly heterogeneous stromal cells and their crosstalk with cancer cells is mediated by a complex and intricate signaling network consisting of transforming growth factor-beta, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin, mitogen-activated protein kinase, Wnt, Janus kinase/signal transducers and activators of transcription, epidermal growth factor receptor, Hippo, and nuclear factor kappa-light-chain-enhancer of activated B cells, etc., signaling pathways. These signals in CAFs exhibit their own special characteristics during the cancer progression and have the potential to be targeted for anticancer therapy. Therefore, a comprehensive understanding of these signaling cascades in interactions between cancer cells and CAFs is necessary to fully realize the pivotal roles of CAFs in cancers. Herein, in this review, we will summarize the enormous amounts of findings on the signals mediating crosstalk of CAFs with cancer cells and its related targets or trials. Further, we hypothesize three potential targeting strategies, including, namely, epithelial-mesenchymal common targets, sequential target perturbation, and crosstalk-directed signaling targets, paving the way for CAF-directed or host cell-directed antitumor therapy.

    Topics: Cancer-Associated Fibroblasts; Cell Proliferation; Extracellular Matrix; Humans; Molecular Targeted Therapy; Neoplasms; Phosphatidylinositol 3-Kinases; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2021
Conservation of Epithelial-to-Mesenchymal Transition Process in Neural Crest Cells and Metastatic Cancer.
    Cells, tissues, organs, 2021, Volume: 210, Issue:3

    Epithelial to mesenchymal transition (EMT) is a highly conserved cellular process in several species, from worms to humans. EMT plays a fundamental role in early embryogenesis, wound healing, and cancer metastasis. For neural crest cell (NCC) development, EMT typically results in forming a migratory and potent cell population that generates a wide variety of cell and tissue, including cartilage, bone, connective tissue, endocrine cells, neurons, and glia amongst many others. The degree of conservation between the signaling pathways that regulate EMT during development and metastatic cancer (MC) has not been fully established, despite ample studies. This systematic review and meta-analysis dissects the major signaling pathways involved in EMT of NCC development and MC to unravel the similarities and differences. While the FGF, TGFβ/BMP, SHH, and NOTCH pathways have been rigorously investigated in both systems, the EGF, IGF, HIPPO, Factor Receptor Superfamily, and their intracellular signaling cascades need to be the focus of future NCC studies. In general, meta-analyses of the associated signaling pathways show a significant number of overlapping genes (particularly ligands, transcription regulators, and targeted cadherins) involved in each signaling pathway of both systems without stratification by body segments and cancer type. Lack of stratification makes it difficult to meaningfully evaluate the intracellular downstream effectors of each signaling pathway. Finally, pediatric neuroblastoma and melanoma are NCC-derived malignancies, which emphasize the importance of uncovering the EMT events that convert NCC into treatment-resistant malignant cells.

    Topics: Cell Movement; Child; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Neural Crest; Transforming Growth Factor beta

2021
Exploring the Crosstalk between Inflammation and Epithelial-Mesenchymal Transition in Cancer.
    Mediators of inflammation, 2021, Volume: 2021

    Tumor cells undergo invasion and metastasis through epithelial-to-mesenchymal cell transition (EMT) by activation of alterations in extracellular matrix (ECM) protein-encoding genes, enzymes responsible for the breakdown of ECM, and activation of genes that drive the transformation of the epithelial cell to the mesenchymal type. Inflammatory cytokines such as TGF

    Topics: Cadherins; Cell Line, Tumor; Cytokines; Disease Progression; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Inflammation; Interleukin-1beta; Interleukin-6; Neoplasms; Neoplastic Stem Cells; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor-alpha

2021
A cytokine in turmoil: Transforming growth factor beta in cancer.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2021, Volume: 139

    Cancer remains one of the debilitating health threats to mankind in view of its incurable nature. Many factors are complicit in the initiation, progression and establishment of cancers. Early detection of cancer is the only window of hope that allows for appreciable management and possible limited survival. However, understanding of cancer biology and knowledge of the key factors that interplay at multi-level in the initiation and progression of cancer may hold possible avenues for cancer treatment and management. In particular, dysregulation of growth factor signaling such as that of transforming growth factor beta (TGF-β) and its downstream mediators play key roles in various cancer subtypes. Expanded understanding of the context/cell type-dependent roles of TGF-β and its downstream signaling mediators in cancer may provide leads for cancer pharmacotherapy. Reliable information contained in original articles, reviews, mini-reviews and expert opinions on TGF-β, cancer and the specific roles of TGF-β signaling in various cancer subtypes were retrieved from major scientific data bases including PubMed, Scopus, Medline, Web of Science core collections just to mention but a sample by using the following search terms: TGF-β in cancer, TGF-β and colorectal cancer, TGF-β and brain cancer, TGF-β in cancer initiation, TGF-β and cell proliferation, TGF-β and cell invasion, and TGF-β-based cancer therapy. Retrieved information and reports were carefully examined, contextualized and synchronized into a coherent scientific content to highlight the multiple roles of TGF-β signaling in normal and cancerous cells. From a conceptual standpoint, development of pharmacologically active agents that exert non-specific inhibitory effects on TGF-β signaling on various cell types will undoubtedly lead to a plethora of serious side effects in view of the multi-functionality and pleiotropic nature of TGF-β. Such non-specific targeting of TGF-β could derail any beneficial therapeutic intention associated with TGF-β-based therapy. However, development of pharmacologically active agents designed specifically to target TGF-β signaling in cancer cells may improve cancer pharmacotherapy. Similarly, specific targeting of downstream mediators of TGF-β such as TGF-β type 1 and II receptors (TβRI and TβRII), receptor-mediated Smads, mitogen activated protein kinase (MAPK) and importing proteins in cancer cells may be crucial for cancer pharmacotherapy.

    Topics: Animals; Cytokines; Humans; Mitogen-Activated Protein Kinases; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2021
TGF-β Signaling: From Tissue Fibrosis to Tumor Microenvironment.
    International journal of molecular sciences, 2021, Jul-15, Volume: 22, Issue:14

    Transforming growth factor-β (TGF-β) signaling triggers diverse biological actions in inflammatory diseases. In tissue fibrosis, it acts as a key pathogenic regulator for promoting immunoregulation via controlling the activation, proliferation, and apoptosis of immunocytes. In cancer, it plays a critical role in tumor microenvironment (TME) for accelerating invasion, metastasis, angiogenesis, and immunosuppression. Increasing evidence suggest a pleiotropic nature of TGF-β signaling as a critical pathway for generating fibrotic TME, which contains numerous cancer-associated fibroblasts (CAFs), extracellular matrix proteins, and remodeling enzymes. Its pathogenic roles and working mechanisms in tumorigenesis are still largely unclear. Importantly, recent studies successfully demonstrated the clinical implications of fibrotic TME in cancer. This review systematically summarized the latest updates and discoveries of TGF-β signaling in the fibrotic TME.

    Topics: Animals; Cancer-Associated Fibroblasts; Fibrosis; Humans; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2021
The transformation of cancer-associated fibroblasts: Current perspectives on the role of TGF-β in CAF mediated tumor progression and therapeutic resistance.
    Cancer letters, 2021, 11-01, Volume: 520

    Over the last few years, the Transforming growth factor- β (TGF-β) has been significantly considered as an effective and ubiquitous mediator of cell growth. The cytokine, TGF-β is being increasingly recognized as the most potent inducer of cancer cell initiation, differentiation, migration as well as progression through both the SMAD-dependent and independent pathways. There is growing evidence that supports the role of secretory cytokine TGF-β as a crucial mediator of tumor-stroma crosstalk. Contextually, the CAFs are the prominent component of tumor stroma that helps in tumor progression and onset of chemoresistance. The interplay between the CAFs and the tumor cells through the paracrine signals is facilitated by cytokine TGF-β to induce the malignant progression. Here in this review, we have dissected the most recent advancements in understanding the mechanisms of TGF-β induced CAF activation, their multiple origins, and most importantly their role in conferring chemoresistance. Considering the pivotal role of TGF-β in tumor perogression and associated stemness, it is one the proven clinical targets We have also included the clinical trials going on, targeting the TGF-β and CAFs crosstalk with the tumor cells. Ultimately, we have underscored some of the outstanding issues that must be deciphered with utmost importance to unravel the successful strategies of anti-cancer therapies.

    Topics: Cancer-Associated Fibroblasts; Carcinogenesis; Cell Differentiation; Cell Proliferation; Drug Resistance, Neoplasm; Humans; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2021
The role of heparan sulfate maturation in cancer: A focus on the 3O-sulfation and the enigmatic 3O-sulfotransferases (HS3STs).
    Seminars in cancer biology, 2020, Volume: 62

    Heparansulfate (HS) modifications are master regulators of the cross-talk between cell and matrix and modulate the biological activity of an array of HS binding proteins, including growth factors and chemokines, morphogens and immunity cell receptors. This review will highlight the importance of HS maturation mediated by N-deactetylase/sulfotransferases, 2O- and 6O-sulfotransferases in cancer biology, and will focus on the 3O-sulfotransferases and on the terminal, rare 3O-sulfation, and their important but still enigmatic impact in cancer progression. The review will also discuss the molecular mechanisms of action of these HS modifications with regards to ligand interactions and signaling in the cancer process and their clinical significance.

    Topics: Animals; Biosynthetic Pathways; Carrier Proteins; Cell Proliferation; Disease Susceptibility; Fibroblast Growth Factors; Heparitin Sulfate; Humans; Ligands; Neoplasms; Protein Binding; Sulfotransferases; Transforming Growth Factor beta

2020
TGFβ-induced metabolic reprogramming during epithelial-to-mesenchymal transition in cancer.
    Cellular and molecular life sciences : CMLS, 2020, Volume: 77, Issue:11

    Metastasis is the most frequent cause of death in cancer patients. Epithelial-to-mesenchymal transition (EMT) is the process in which cells lose epithelial integrity and become motile, a critical step for cancer cell invasion, drug resistance and immune evasion. The transforming growth factor-β (TGFβ) signaling pathway is a major driver of EMT. Increasing evidence demonstrates that metabolic reprogramming is a hallmark of cancer and extensive metabolic changes are observed during EMT. The aim of this review is to summarize and interconnect recent findings that illustrate how changes in glycolysis, mitochondrial, lipid and choline metabolism coincide and functionally contribute to TGFβ-induced EMT. We describe TGFβ signaling is involved in stimulating both glycolysis and mitochondrial respiration. Interestingly, the subsequent metabolic consequences for the redox state and lipid metabolism in cancer cells are found to be in favor of EMT as well. Combined we illustrate that a better understanding of the mechanistic links between TGFβ signaling, cancer metabolism and EMT holds promising strategies for cancer therapy, some of which are already actively being explored in the clinic.

    Topics: Animals; Cell Respiration; Epithelial-Mesenchymal Transition; Glycolysis; Humans; Lipid Metabolism; Mitochondria; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2020
Smad2 linker region phosphorylation is an autonomous cell signalling pathway: Implications for multiple disease pathologies.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2020, Volume: 124

    This review will cover the signalling pathways leading to the phosphorylation of the Smad linker region independent of Smad carboxy terminal phosphorylation. Characterising Smad linker region as a signalling pathway in its own right will encourage comprehensive signalling studies to provide solutions for successful discovery and exploitation of drug targets. The review describes Smad transcription factor signalling distinct from Transforming Growth Factor (TGF)-β signalling. Novel signalling pathways represent new drug targets where these pathways are known to be involved in fibrosis, cancer and cardiovascular disease.

    Topics: Animals; Cardiovascular Diseases; Fibrosis; Humans; Neoplasms; Phosphorylation; Signal Transduction; Smad2 Protein; Transforming Growth Factor beta

2020
Epithelial to Mesenchymal Transition: A Mechanism that Fuels Cancer Radio/Chemoresistance.
    Cells, 2020, 02-12, Volume: 9, Issue:2

    Epithelial to mesenchymal transition (EMT) contributes to tumor progression, cancer cell invasion, and therapy resistance. EMT is regulated by transcription factors such as the protein products of the SNAI gene family, which inhibits the expression of epithelial genes. Several signaling pathways, such as TGF-beta1, IL-6, Akt, and Erk1/2, trigger EMT responses. Besides regulatory transcription factors, RNA molecules without protein translation, micro RNAs, and long non-coding RNAs also assist in the initialization of the EMT gene cluster. A challenging novel aspect of EMT research is the investigation of the interplay between tumor microenvironments and EMT. Several microenvironmental factors, including fibroblasts and myofibroblasts, as well as inflammatory, immune, and endothelial cells, induce EMT in tumor cells. EMT tumor cells change their adverse microenvironment into a tumor friendly neighborhood, loaded with stromal regulatory T cells, exhausted CD8

    Topics: Antineoplastic Agents, Phytogenic; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Humans; Mitogen-Activated Protein Kinase 3; Neoplasms; Signal Transduction; T-Lymphocytes, Regulatory; Transcription Factors; Transforming Growth Factor beta

2020
Endoglin: Beyond the Endothelium.
    Biomolecules, 2020, 02-12, Volume: 10, Issue:2

    Topics: Activin Receptors, Type II; Adaptive Immunity; Animals; Antibodies, Monoclonal; Clinical Trials as Topic; Endoglin; Endothelium; Epithelial Cells; Fibrosis; Growth Differentiation Factor 2; Hematopoiesis; Humans; Immunity, Innate; Ligands; Mesenchymal Stem Cells; Mice; Neoplasms; Neovascularization, Pathologic; Phosphorylation; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2020
Role of fibrillin-2 in the control of TGF-β activation in tumor angiogenesis and connective tissue disorders.
    Biochimica et biophysica acta. Reviews on cancer, 2020, Volume: 1873, Issue:2

    Fibrillins constitute a family of large extracellular glycoproteins which multimerize to form microfibrils, an important structure in the extracellular matrix. It has long been assumed that fibrillin-2 was barely present during postnatal life, but it is now clear that fibrillin-2 molecules form the structural core of microfibrils, and are masked by an outer layer of fibrillin-1. Mutations in fibrillins give rise to heritable connective tissue disorders, including Marfan syndrome and congenital contractural arachnodactyly. Fibrillins also play an important role in matrix sequestering of members of the transforming growth factor-β family, and in context of Marfan syndrome excessive TGF-β activation has been observed. TGF-β activation is highly dependent on integrin binding, including integrin αvβ8 and αvβ6, which are upregulated upon TGF-β exposure. TGF-β is also involved in tumor progression, metastasis, epithelial-to-mesenchymal transition and tumor angiogenesis. In several highly vascularized types of cancer such as hepatocellular carcinoma, a positive correlation was found between increased TGF-β plasma concentrations and tumor vascularity. Interestingly, fibrillin-1 has a higher affinity to TGF-β and, therefore, has a higher capacity to sequester TGF-β compared to fibrillin-2. The previously reported downregulation of fibrillin-1 in tumor endothelium affects the fibrillin-1/fibrillin-2 ratio in the microfibrils, exposing the normally hidden fibrillin-2. We postulate that fibrillin-2 exposure in the tumor endothelium directly stimulates tumor angiogenesis by influencing TGF-β sequestering by microfibrils, leading to a locally higher active TGF-β concentration in the tumor microenvironment. From a therapeutic perspective, fibrillin-2 might serve as a potential target for future anti-cancer therapies.

    Topics: Animals; Arachnodactyly; Connective Tissue; Contracture; Disease Models, Animal; Endothelium, Vascular; Fibrillin-2; Humans; Marfan Syndrome; Mutation; Neoplasms; Neovascularization, Pathologic; Transforming Growth Factor beta; Tumor Microenvironment

2020
TGF-β in radiotherapy: Mechanisms of tumor resistance and normal tissues injury.
    Pharmacological research, 2020, Volume: 155

    Emerging evidences show that changes in tumor stroma can adapt cancer cells to radiotherapy, thereby leading to a reduction in tumor response to treatment. On the other hand, radiotherapy is associated with severe reactions in normal tissues which limit the amount radiation dose received by tumor. These challenges open a window in radiobiology and radiation oncology to explore mechanisms for improving tumor response and also alleviate side effects of radiotherapy. Transforming growth factor beta (TGF-β) is a well-known and multitasking cytokine that regulates a wide range of reactions and interactions within tumor and normal tissues. Within tumor microenvironment (TME), TGF-β is the most potent suppressor of immune system activity against cancer cells. This effect is mediated through stimulation of CD4+ which differentiates to T regulatory cells (Tregs), infiltration of fibroblasts and differentiation into cancer associated fibroblasts (CAFs), and also polarization of macrophages to M2 cells. These changes lead to suppression of cytotoxic CD8 + T lymphocytes (CTLs) and natural killer (NK) cells to kill cancer cells. TGF-β also plays a key role in the angiogenesis, invasion and DNA damage responses (DDR) in cancer cells. In normal tissues, TGF-β triggers the expression of a wide range of pro-oxidant and pro-fibrosis genes, leading to fibrosis, genomic instability and some other side effects. These properties of TGF-β make it a potential target to preserve normal tissues and sensitize tumor via its inhibition. In the current review, we aim to explain the mechanisms of upregulation of TGF-β and its consequences in both tumor and normal tissues.

    Topics: Animals; Humans; Neoplasms; Transforming Growth Factor beta

2020
Immunotherapeutic Potential of TGF-β Inhibition and Oncolytic Viruses.
    Trends in immunology, 2020, Volume: 41, Issue:5

    In cancer immunotherapy, a patient's own immune system is harnessed against cancer. Immune checkpoint inhibitors release the brakes on tumor-reactive T cells and, therefore, are particularly effective in treating certain immune-infiltrated solid tumors. By contrast, solid tumors with immune-silent profiles show limited efficacy of checkpoint blockers due to several barriers. Recent discoveries highlight transforming growth factor-β (TGF-β)-induced immune exclusion and a lack of immunogenicity as examples of these barriers. In this review, we summarize preclinical and clinical evidence that illustrates how the inhibition of TGF-β signaling and the use of oncolytic viruses (OVs) can increase the efficacy of immunotherapy, and discuss the promise and challenges of combining these approaches with immune checkpoint blockade.

    Topics: Humans; Immunotherapy; Neoplasms; Oncolytic Virotherapy; Oncolytic Viruses; Transforming Growth Factor beta

2020
TGF-beta: a master immune regulator.
    Expert opinion on therapeutic targets, 2020, Volume: 24, Issue:5

    Topics: Animals; Gene Expression Regulation, Neoplastic; Humans; Immune Checkpoint Inhibitors; Immunity, Cellular; Immunotherapy; Molecular Targeted Therapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2020
LncRNA-ATB in cancers: what do we know so far?
    Molecular biology reports, 2020, Volume: 47, Issue:5

    Cancer-related deaths did not apparently decrease in the past decades despite aggressive treatments. It's reported that cancer will become the leading cause of death worldwide in the twenty-first century. Increasing evidence has revealed that lncRNAs will emerge as promising cancer biomarkers or therapeutic targets in cancer treatment. LncRNA-ATB, a long noncoding RNA activated by TGF-β, was found to be abnormally expressed in certain cancers and participate in the development and progression of tumors. In addition, aberrant lncRNA-ATB expression was also associated with clinical characteristics of tumors. The purpose of this review is to summarize functions and underlying mechanisms of lncRNA-ATB in tumors, and discuss whether lncRNA-ATB can be a biomarker and therapeutic target in cancers.

    Topics: Biomarkers, Tumor; Cell Proliferation; Disease Progression; Humans; Neoplasms; RNA, Long Noncoding; Transforming Growth Factor beta

2020
The Distinct Roles of Transcriptional Factor KLF11 in Normal Cell Growth Regulation and Cancer as a Mediator of TGF-β Signaling Pathway.
    International journal of molecular sciences, 2020, Apr-22, Volume: 21, Issue:8

    KLF11 (Krüppel-like factor 11) belongs to the family of Sp1/Krüppel-like zinc finger transcription factors that play important roles in a variety of cell types and tissues. KLF11 was initially described as a transforming growth factor-beta (TGF-β) inducible immediate early gene (TIEG). KLF11 promotes the effects of TGF-β on cell growth control by influencing the TGFβ-Smads signaling pathway and regulating the transcription of genes that induce either apoptosis or cell cycle arrest. In carcinogenesis, KLF11 can show diverse effects. Its function as a tumor suppressor gene can be suppressed by phosphorylation of its binding domains via oncogenic pathways. However, KLF 11 can itself also show tumor-promoting effects and seems to have a crucial role in the epithelial-mesenchymal transition process. Here, we review the current knowledge about the function of KLF11 in cell growth regulation. We focus on its transcriptional regulatory function and its influence on the TGF-β signaling pathway. We further discuss its possible role in mediating crosstalk between various signaling pathways in normal cell growth and in carcinogenesis.

    Topics: Animals; Apoptosis; Apoptosis Regulatory Proteins; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Repressor Proteins; Signal Transduction; Trans-Activators; Transcription Factors; Transforming Growth Factor beta

2020
Rational targeting of immunosuppressive neutrophils in cancer.
    Pharmacology & therapeutics, 2020, Volume: 212

    Neutrophils, the most abundant circulating leukocytes in human, play an indispensable role in the innate immune response to microbial infections. However, the contribution of tumor-associated neutrophils (TANs) to cancer progression and tumor immunity has been a matter of debate for decades. A higher neutrophil-to-lymphocyte ratio is associated with adverse overall survival in many solid tumors. Preclinical evidence exists to support both anti-tumor and pro-tumor activities of TANs, and TANs employ diverse mechanisms to influence tumor progression and metastasis. Here, we focus our review on the immunosuppressive mechanism of TANs and highlight how neutrophils can operate to dampen both innate and adaptive immunity to promote tumorigenesis. Here we discuss the intriguing and sometimes controversial connection between TANs and granulocytic/polymorphonuclear myeloid-derived suppressor cells (G/PMN-MDSCs). The molecular mechanisms underlying neutrophils' role in immunosuppression provide potential therapeutic targets for cancer treatment, either as monotherapies or as a part of combinatorial regimens. Therefore, we also highlight a number of neutrophil-targeting approaches that may improve the efficacy of current anticancer therapies, especially cancer immunotherapy. Currently interest is surging in the understanding and targeting of immunosuppressive neutrophils, with the goal of developing novel therapeutic strategies in the battle against cancer.

    Topics: Extracellular Traps; Granulocyte Colony-Stimulating Factor; Humans; Immune Tolerance; Neoplasms; Neutrophils; Proto-Oncogene Proteins c-met; Transforming Growth Factor beta; Tumor Microenvironment

2020
New insights into the core Hippo signaling and biological macromolecules interactions in the biology of solid tumors.
    BioFactors (Oxford, England), 2020, Volume: 46, Issue:4

    As an evolutionarily conserved pathway, Hippo signaling pathway impacts different pathology and physiology processes such as wound healing, tissue repair/size and regeneration. When some components of Hippo signaling dysregulated, it affects cancer cells proliferation. Moreover, the relation Hippo pathway with other signaling including Wnt, TGFβ, Notch, and EGFR signaling leaves effect on the proliferation of cancer cells. Utilizing a number of therapeutic approaches, such as siRNAs and long noncoding RNA (lncRNA) to prevent cancer cells through the targeting of Hippo pathways, can provide new insights into cancer target therapy. The purpose of present review, first of all, is to demonstrate the importance of Hippo signaling and its relation with other signaling pathways in cancer. It also tries to demonstrate targeting Hippo signaling progress in cancer therapy.

    Topics: Animals; Antineoplastic Agents; Drosophila melanogaster; ErbB Receptors; Gene Expression Regulation, Neoplastic; Hippo Signaling Pathway; Humans; MicroRNAs; Molecular Targeted Therapy; Neoplasm Proteins; Neoplasms; Protein Interaction Mapping; Protein Serine-Threonine Kinases; Receptors, Notch; RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta; Wnt Proteins

2020
Long non-coding RNAs and TGF-β signaling in cancer.
    Cancer science, 2020, Volume: 111, Issue:8

    Cancer is driven by genetic mutations in oncogenes and tumor suppressor genes and by cellular events that develop a misregulated molecular microenvironment in the growing tumor tissue. The tumor microenvironment is guided by the excessive action of specific cytokines including transforming growth factor-β (TGF-β), which normally controls embryonic development and the homeostasis of young or adult tissues. As a consequence of the genetic alterations generating a given tumor, TGF-β can preserve its homeostatic function and attempt to limit neoplastic expansion, whereas, once the tumor has progressed to an aggressive stage, TGF-β can synergize with various oncogenic stimuli to facilitate tumor invasiveness and metastasis. TGF-β signaling mechanisms via Smad proteins, various ubiquitin ligases, and protein kinases are relatively well understood. Such mechanisms regulate the expression of genes encoding proteins or non-coding RNAs. Among non-coding RNAs, much has been understood regarding the regulation and function of microRNAs, whereas the role of long non-coding RNAs is still emerging. This article emphasizes TGF-β signaling mechanisms leading to the regulation of non-coding genes, the function of such non-coding RNAs as regulators of TGF-β signaling, and the contribution of these mechanisms in specific hallmarks of cancer.

    Topics: Animals; Carcinogenesis; Cell Line, Tumor; Feedback, Physiological; Gene Expression Regulation, Neoplastic; Humans; Neoplasm Invasiveness; Neoplasms; RNA, Long Noncoding; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2020
Extracellular BMP Antagonists, Multifaceted Orchestrators in the Tumor and Its Microenvironment.
    International journal of molecular sciences, 2020, May-29, Volume: 21, Issue:11

    The bone morphogenetic proteins (BMPs), a subgroup of the transforming growth factor-β (TGF-β) superfamily, are involved in multiple biological processes such as embryonic development and maintenance of adult tissue homeostasis. The importance of a functional BMP pathway is underlined by various diseases, including cancer, which can arise as a consequence of dysregulated BMP signaling. Mutations in crucial elements of this signaling pathway, such as receptors, have been reported to disrupt BMP signaling. Next to that, aberrant expression of BMP antagonists could also contribute to abrogated signaling. In this review we set out to highlight how BMP antagonists affect not only the cancer cells, but also the other cells present in the microenvironment to influence cancer progression.

    Topics: Animals; Bone Morphogenetic Proteins; Carrier Proteins; Cell Line, Tumor; Disease Progression; Endothelial Cells; Gene Expression Regulation, Neoplastic; Humans; Intercellular Signaling Peptides and Proteins; Mice; Mutation; Neoplasms; Phylogeny; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2020
Fibroblasts: The arbiters of extracellular matrix remodeling.
    Matrix biology : journal of the International Society for Matrix Biology, 2020, Volume: 91-92

    Extracellular matrix (ECM) is the foundation on which all cells and organs converge to orchestrate normal physiological functions. In the setting of pathology, the ECM is modified to incorporate additional roles, with modifications including turnover of existing ECM and deposition of new ECM. The fibroblast is center stage in coordinating both normal tissue homeostasis and response to disease. Understanding how fibroblasts work under normal conditions and are activated in response to injury or stress will provide mechanistic insight that triggers discovery of new therapeutic treatments for a wide range of disease. We highlight here fibroblast roles in the cancer, lung, and heart as example systems where fibroblasts are major contributors to homeostasis and pathology.

    Topics: Extracellular Matrix; Extracellular Matrix Proteins; Fibroblasts; Fibrosis; Gene Expression Regulation; Homeostasis; Humans; Hypertension; Inflammation; Lung; Myocardial Infarction; Myocardium; Neoplasms; Stromal Cells; Transforming Growth Factor beta; Wnt Signaling Pathway

2020
SMAD-oncoprotein interplay: Potential determining factors in targeted therapies.
    Biochemical pharmacology, 2020, Volume: 180

    The Transforming Growth Factor-β (TGF-β) signaling pathway plays a versatile role in diverse physiological and disease conditions. Outcomes of TGF-β signaling are divergent, sometimes even opposite, on cellular functions and disease progression through context-dependent transcriptional programs. For example, TGF-β signaling is well known for its dichotomous roles in cancer progression, serving both as an inhibitor of tumor cell growth and an inducer of tumor metastasis. This is achieved mainly through the interplay between Smad proteins (Smads) and cofactors/modulators in a context-determined manner. Smad proteins can be post-translationally modified through several mechanisms, and are able to interact with many transcription coactivators/corepressors during a wide range of biological functions. As of such, Smads influence on many oncogenic processes through the interplay with proteins in oncogenic pathways. Because of the importance of the TGF-β pathway in cancer progression, the interplay between Smads and oncogenic drug targets has considerable impact on the outcome of targeted therapies. In this review, we focus on the interplay between Smads and oncogenic drug targets, with a discussion on how this interplay guides targeted anticancer therapies.

    Topics: Animals; Antineoplastic Agents; Drug Delivery Systems; Humans; Neoplasms; Oncogene Proteins; Protein Binding; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2020
Clinical development of therapies targeting TGFβ: current knowledge and future perspectives.
    Annals of oncology : official journal of the European Society for Medical Oncology, 2020, Volume: 31, Issue:10

    Transforming growth factor beta (TGFβ) is a pleiotropic cytokine that plays a key role in both physiologic and pathologic conditions, including cancer. Importantly, TGFβ can exhibit both tumor-suppressive and oncogenic functions. In normal epithelial cells TGFβ acts as an antiproliferative and differentiating factor, whereas in advanced tumors TGFβ can act as an oncogenic factor by creating an immune-suppressive tumor microenvironment, and inducing cancer cell proliferation, angiogenesis, invasion, tumor progression, and metastatic spread. A wealth of preclinical findings have demonstrated that targeting TGFβ is a promising means of exerting antitumor activity. Based on this rationale, several classes of TGFβ inhibitors have been developed and tested in clinical trials, namely, monoclonal, neutralizing, and bifunctional antibodies; antisense oligonucleotides; TGFβ-related vaccines; and receptor kinase inhibitors. It is now >15 years since the first clinical trial testing an anti-TGFβ agent was engaged. Despite the promising preclinical studies, translation of the basic understanding of the TGFβ oncogenic response into the clinical setting has been slow and challenging. Here, we review the conclusions and status of all the completed and ongoing clinical trials that test compounds that inhibit the TGFβ pathway, and discuss the challenges that have arisen during their clinical development. With none of the TGFβ inhibitors evaluated in clinical trials approved for cancer therapy, clinical development for TGFβ blockade therapy is primarily oriented toward TGFβ inhibitor combinations. Immune checkpoint inhibitors are considered candidates, albeit with efficacy anticipated to be restricted to specific populations. In this context, we describe current efforts in the search for biomarkers for selecting the appropriate cancer patients who are likely to benefit from anti-TGFβ therapies. The knowledge accumulated during the last 15 years of clinical research in the context of the TGFβ pathway is crucial to design better, innovative, and more successful trials.

    Topics: Humans; Immunotherapy; Neoplasms; Oncogenes; Transforming Growth Factor beta; Tumor Microenvironment

2020
Thrombospondin in Tumor Microenvironment.
    Advances in experimental medicine and biology, 2020, Volume: 1272

    Thrombospondins (TSPs) are multifaceted proteins that contribute to physiologic as well as pathologic conditions. Due to their multiple receptor-binding domains, TSPs display both oncogenic and tumor-suppressive qualities and are thus essential components of the extracellular matrix. Known for their antiangiogenic capacity, TSPs are an important component of the tumor microenvironment. The N- and C-terminal domains of TSP are, respectively, involved in cell adhesion and spreading, an important feature of wound healing as well as cancer cell migration. Previously known for the activation of TGF-β to promote tumor growth and inflammation, TSP-1 has recently been found to be transcriptionally induced by TGF-β, implying the presence of a possible feedback loop. TSP-1 is an endogenous inhibitor of T cells and also mediates its immunosuppressive effects via induction of Tregs. Given the diverse roles of TSPs in the tumor microenvironment, many therapeutic strategies have utilized TSP-mimetic peptides or antibody blockade as anti-metastatic approaches. This chapter discusses the diverse structural domains, functional implications, and anti-metastatic therapies in the context of the role of TSP in the tumor microenvironment.

    Topics: Angiogenesis Inhibitors; Cell Movement; Humans; Neoplasms; Thrombospondins; Transforming Growth Factor beta; Tumor Microenvironment

2020
Roles of TGF-β signaling pathway in tumor microenvirionment and cancer therapy.
    International immunopharmacology, 2020, Volume: 89, Issue:Pt B

    Transforming growth factor β (TGF- β) signaling pathway has pleiotropic effects on cell proliferation, differentiation, adhesion, senescence, and apoptosis. TGF-β can be widely produced by various immune or non-immune cells and regulate cell behaviors through autocrine and paracrine. It plays essential roles in biological processes including embryological development, immune response, and tumor progression. Few cell signalings can contribute to so many pleiotropic functions as the TGF- β signaling pathway in mammals. The significant function of TGF-β signaling in tumor progression and evasion leading it to draw great attention in scientific and clinical research. Understanding the mechanism of TGF- β signaling provides us with chances to potentiate the effectiveness and selectivity of this therapeutic method. Herein, we review the molecular and cellular mechanisms of TGF-β signaling in carcinomas and tumor microenvironment. Then, we enumerate main achievements of TGF-β blockades used or being evaluated in cancer therapy, providing us opportunities to improve therapeutical approaches in the tumor which thrive in a TGF-β-rich environment.

    Topics: Animals; Humans; Immune Checkpoint Inhibitors; Neoplasms; Receptors, Transforming Growth Factor beta; RNA, Messenger; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2020
Transforming growth factor β signaling pathway: A promising therapeutic target for cancer.
    Journal of cellular physiology, 2020, Volume: 235, Issue:3

    Transforming growth factor β (TGF-β) is part of the transforming growth factor β superfamily which is involved in many physiological processes and closely related to the carcinogenesis. Here, we discuss the TGF-β structure, function, and its canonical Smads signaling pathway. Importantly, TGF-β has been proved that it plays both tumor suppressor as well as an activator role in tumor progression. In an early stage, TGF-β inhibits cell proliferation and is involved in cell apoptosis. In an advanced tumor, TGF-β signaling pathway induces tumor invasion and metastasis through promoting angiogenesis, epithelial-mesenchymal transition, and immune escape. Furthermore, we are centered on updated research results into the inhibitors as drugs which have been studied in preclinical or clinical trials in tumor carcinogenesis to prevent the TGF-β synthesis and block its signaling pathways such as antibodies, antisense molecules, and small-molecule tyrosine kinase inhibitors. Thus, it is highlighting the crucial role of TGF-β in tumor therapy and may provide opportunities for the new antitumor strategies in patients with cancer.

    Topics: Animals; Antineoplastic Agents; Apoptosis; Cell Proliferation; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2020
T Helper 9 Cells: A New Player in Immune-Related Diseases.
    DNA and cell biology, 2019, Volume: 38, Issue:10

    The helper T cell 9 (Thelper-9, Th9), as a functional subgroup of CD4

    Topics: Animals; Cell Differentiation; GATA3 Transcription Factor; Gene Expression Regulation; Humans; Inflammation; Interleukin-4; Interleukin-9; Lupus Erythematosus, Systemic; Mice; Mice, Knockout; Neoplasms; Proto-Oncogene Proteins; Signal Transduction; STAT6 Transcription Factor; T-Lymphocytes, Helper-Inducer; Trans-Activators; Transforming Growth Factor beta

2019
The Interactivity between TGFβ and BMP Signaling in Organogenesis, Fibrosis, and Cancer.
    Cells, 2019, 09-23, Volume: 8, Issue:10

    The Transforming Growth Factor beta (TGFβ) and Bone Morphogenic Protein (BMP) pathways intersect at multiple signaling hubs and cooperatively or counteractively participate to bring about cellular processes which are critical not only for tissue morphogenesis and organogenesis during development, but also for adult tissue homeostasis. The proper functioning of the TGFβ/BMP pathway depends on its communication with other signaling pathways and any deregulation leads to developmental defects or diseases, including fibrosis and cancer. In this review we explore the cellular and physio-pathological contexts in which the synergism or antagonism between the TGFβ and BMP pathways are crucial determinants for the normal developmental processes, as well as the progression of fibrosis and malignancies.

    Topics: Animals; Bone Morphogenetic Proteins; Disease Progression; Fibrosis; Gene Regulatory Networks; Humans; Neoplasms; Organogenesis; Signal Transduction; Transforming Growth Factor beta

2019
Double-edge Role of B Cells in Tumor Immunity: Potential Molecular Mechanism.
    Current medical science, 2019, Volume: 39, Issue:5

    B cells are a heterogeneous population, which have distinct functions of antigen presentation, activating T cells, and secreting antibodies, cytokines as well as protease. It is supposed that the balance among these B cells subpopulation (resting B cells, activated B cells, Bregs, and other differentiated B cells) will determine the ultimate role of B cells in tumor immunity. There has been increasing evidence supporting opposite roles of B cells in tumor immunity, though there are no general acceptable phenotypes for them. Recent years, a new designated subset of B cells identified as Bregs has emerged from immunosuppressive and/or regulatory functions in tumor immune responses. Therefore, transferring activated B cells would be possible to become a promising strategy against tumor via conquering the immunosuppressive status of B cells in future. Understanding the potential mechanism of double-edge role of B cells will help researchers utilize activated B cells to improve their anti-tumor response. Moreover, the molecular pathways related to B cell differentiation are involved in its tumor-promoting effect, such as NF-κB, STAT3, BTK. So, we review the molecular and signaling pathway mechanisms of B cells involved in both tumor-promoting and tumor-suppressive immunity, in order to help researchers optimize B cells to fight cancer better.

    Topics: Agammaglobulinaemia Tyrosine Kinase; Animals; B-Lymphocyte Subsets; Cell Differentiation; Gene Expression Regulation, Neoplastic; Humans; Immunophenotyping; Interleukin-10; Lymphocyte Activation; Mice; Neoplasms; NF-kappa B; Signal Transduction; STAT3 Transcription Factor; Transforming Growth Factor beta; Tumor Escape

2019
Cell Signaling Pathways Related to Epithelial Mesenchymal Transition in Cancer Metastasis.
    Critical reviews in oncogenesis, 2019, Volume: 24, Issue:1

    The epithelial mesenchymal transition (EMT) is a highly complex and dynamic morphogenetic process in which epithelial cells change their cellular characteristics and transform to mesenchymal cells. It plays a key role in tissue remodeling, not only during embryonic development and stem cell biology but also during wound healing, fibrosis, and cancer progression. In EMT, under different extracellular cell signals and stimuli, epithelial cells undergo an orchestrated series of morphological, cellular, biochemical, and molecular changes. The mechanism of cancer cell metastasis is similar to embryonic development, like epithelial cells migrating and differentiating into all tissues of the embryo. In this period, epithelial cells differentiate into mesenchymal cells, migrate, and return to the epithelial cells when necessary. The important cell signaling pathways in embryonic development, such as growth factor receptor tyrosine kinases (FGF, HGF, TGF-α), TGF-β, Wnt, NOTCH, MAPK, JAK/STAT and inflammatory cytokines such as NF-κB, TNF-α, IL6, cathepsin and HIF-1α. All play crucial roles in EMT during cancer cell invasion and metastasis. In cancer progression, similar cell signaling pathways and molecular activation occur in embryonic development; therefore, it is important to understand the molecular mechanisms of cell signaling pathways related to cancer metastasis and improve new diagnostic and therapeutic approaches for resistance to drugs.

    Topics: Animals; Biomarkers; Cell Movement; Epithelial-Mesenchymal Transition; Extracellular Space; Gene Expression Regulation; Humans; Neoplasm Metastasis; Neoplasms; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Signal Transduction; TOR Serine-Threonine Kinases; Transcription Factors; Transforming Growth Factor beta; Wnt Signaling Pathway

2019
A Perspective on the Development of TGF-β Inhibitors for Cancer Treatment.
    Biomolecules, 2019, 11-17, Volume: 9, Issue:11

    Transforming growth factor (TGF)-β is a secreted multifunctional cytokine that signals via plasma membrane TGF-β type I and type II receptors and intercellular SMAD transcriptional effectors. Aberrant inter- and intracellular TGF-β signaling can contribute to cancer progression. In normal cells and early stages of cancer, TGF-β can stimulate epithelial growth arrest and elicit a tumor suppressor function. However, in late stages of cancer, when the cytostatic effects of TGF-β in cancer cells are blocked, TGF-β signaling can act as tumor promoter by its ability to stimulate epithelial-to-mesenchymal transition of cancer cells, by stimulating angiogenesis, and by promoting evasion of immune responses. In this review, we will discuss the rationale and challenges of targeting TGF-β signaling in cancer and summarize the clinical status of TGF-β signaling inhibitors that interfere with TGFβ bioavailability, TGF-βreceptor interaction, or TGF-β receptor kinase function. Moreover, we will discuss targeting of TGF-β signaling modulators and downstream effectors as well as alternative approaches by using promising technologies that may lead to entirely new classes of drugs.

    Topics: Antineoplastic Agents; Humans; Neoplasm Proteins; Neoplasms; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Signal Transduction; Transforming Growth Factor beta

2019
Molecular mechanisms of epithelial to mesenchymal transition in tumor metastasis.
    Acta biochimica Polonica, 2019, Dec-28, Volume: 66, Issue:4

    Epithelial to mesenchymal transition (EMT) is a process during which cancer cells lose epithelial features, cytoskeletal architecture is re-organized, cell shape changes and cells activate genes that help to define mesenchymal phenotype, what leads to an increased cell motility and dissemination of tumor to distant metastatic sites. This review describes different signaling networks between microRNAs and proteins that regulate EMT in tumor growth. Activation of EMT is mediated via series of paracrine signaling molecules. WNT, TGF-b, NOTCH and Shh signaling pathways play crucial roles in activation of EMT-related transcription factors, such as SNAIL, SLUG, ZEB1/2 or TWIST. Recent data provide evidence that crosstalk between microRNAs, long non-coding RNAs and EMT-transcription factors is crucial event in EMT regulation. MicroRNAs affect also level of proteins responsible for cellular contact, adhesion and cytoskeletal proteins, what induces changes of epithelial to mesenchymal phenotype. Understanding of those signaling networks may help to identify novel biomarkers or develop new treatment strategies based on microRNA therapeutics in future.

    Topics: Cell Movement; Epithelial-Mesenchymal Transition; Hedgehog Proteins; Humans; MicroRNAs; Neoplasm Metastasis; Neoplasms; Receptors, Notch; Snail Family Transcription Factors; Transforming Growth Factor beta; Twist-Related Protein 1; Wnt Signaling Pathway; Zinc Finger E-box-Binding Homeobox 1

2019
TGF-beta signaling in cancer: post-transcriptional regulation of EMT via hnRNP E1.
    Cytokine, 2019, Volume: 118

    The TGFβ signaling pathway is a critical regulator of cancer progression in part through induction of the epithelial to mesenchymal transition (EMT). This process is aberrantly activated in cancer cells, facilitating invasion of the basement membrane, survival in the circulatory system, and dissemination to distant organs. The mechanisms through which epithelial cells transition to a mesenchymal state involve coordinated transcriptional and post-transcriptional control of gene expression. One such mechanism of control is through the RNA binding protein hnRNP E1, which regulates splicing and translation of a cohort of EMT and stemness-associated transcripts. A growing body of evidence indicates a major role for hnRNP E1 in the control of epithelial cell plasticity, especially in the context of carcinoma progression. Here, we review the multiple mechanisms through which hnRNP E1 functions to control EMT and metastatic progression.

    Topics: Animals; Epithelial-Mesenchymal Transition; Humans; Neoplasms; RNA Processing, Post-Transcriptional; RNA-Binding Proteins; Signal Transduction; Transforming Growth Factor beta

2019
Genetic status of KRAS influences Transforming Growth Factor-beta (TGF-β) signaling: An insight into Neuropilin-1 (NRP1) mediated tumorigenesis.
    Seminars in cancer biology, 2019, Volume: 54

    Oncogenic RAS and deregulated transforming growth factor-beta (TGF)-β signaling have been implicated in several cancers. So far, attempts to target either one of them therapeutically have been futile as both of them are involved in multiple fundamental cellular processes and the normal forms are expressed by almost all cells. Hence, their inhibition would disrupt several physiological processes. Besides, their downregulation stimulates the tumor cells to develop adaptive mechanisms and would most likely be ineffective as therapeutic targets. Furthermore, growing literature suggests that both of these signaling pathways converge to enhance tumor development. Therefore, a lot of interest has been generated to explore the areas where these pathways interface that might identify new molecules that could potentially serve as novel therapeutic targets. In this review, we focus on such convergent signaling and cross-interaction that is mediated by neuropilin-1 (NRP1), a receptor that can interact with multiple growth factors including TGF-β for promoting tumorigenesis process.

    Topics: Animals; Cell Transformation, Neoplastic; Cytokines; Gene Expression Regulation, Neoplastic; Genetic Variation; Humans; Intercellular Signaling Peptides and Proteins; Neoplasms; Neuropilin-1; Proto-Oncogene Proteins p21(ras); Signal Transduction; Transforming Growth Factor beta

2019
H19X-encoded miR-424(322)/-503 cluster: emerging roles in cell differentiation, proliferation, plasticity and metabolism.
    Cellular and molecular life sciences : CMLS, 2019, Volume: 76, Issue:5

    miR-424(322)/-503 are mammal-specific members of the extended miR-15/107 microRNA family. They form a co-expression network with the imprinted lncRNA H19 in tetrapods. miR-424(322)/-503 regulate fundamental cellular processes including cell cycle, epithelial-to-mesenchymal transition, hypoxia and other stress response. They control tissue differentiation (cardiomyocyte, skeletal muscle, monocyte) and remodeling (mammary gland involution), and paradoxically participate in tumor initiation and progression. Expression of miR-424(322)/-503 is governed by unique mechanisms involving sex hormones. Here, we summarize current literature and provide a primer for future endeavors.

    Topics: Animals; Apoptosis; Biomarkers; Cell Differentiation; Cell Plasticity; Cell Proliferation; Endoplasmic Reticulum Stress; Epithelial-Mesenchymal Transition; Genes, Tumor Suppressor; Glycolysis; Humans; MicroRNAs; Neoplasms; RNA, Long Noncoding; Transforming Growth Factor beta

2019
CXXC5: A novel regulator and coordinator of TGF-β, BMP and Wnt signaling.
    Journal of cellular and molecular medicine, 2019, Volume: 23, Issue:2

    CXXC5 is a member of the CXXC-type zinc-finger protein family. Proteins in this family play a pivotal role in epigenetic regulation by binding to unmethylated CpG islands in gene promoters through their characteristic CXXC domain. CXXC5 is a short protein (322 amino acids in length) that does not have any catalytic domain, but is able to bind to DNA and act as a transcription factor and epigenetic factor through protein-protein interactions. Intriguingly, increasing evidence indicates that expression of the CXXC5 gene is controlled by multiple signaling pathways and a variety of transcription factors, positioning CXXC5 as an important signal integrator. In addition, CXXC5 is capable of regulating various signal transduction processes, including the TGF-β, Wnt and ATM-p53 pathways, thereby acting as a novel and crucial signaling coordinator. CXXC5 plays an important role in embryonic development and adult tissue homeostasis by regulating cell proliferation, differentiation and apoptosis. In keeping with these functions, aberrant expression or altered activity of CXXC5 has been shown to be involved in several human diseases including tumourigenesis. This review summarizes the current understanding of CXXC5 as a transcription factor and signaling regulator and coordinator.

    Topics: Amino Acid Sequence; beta Catenin; Bone Morphogenetic Protein 4; Carcinogenesis; DNA-Binding Proteins; Epigenesis, Genetic; Humans; Neoplasms; Protein Binding; Protein Domains; Signal Transduction; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Wnt3A Protein

2019
Transforming growth factor-β signaling: Tumorigenesis and targeting for cancer therapy.
    Journal of cellular physiology, 2019, Volume: 234, Issue:8

    Transforming growth factor (TGF)-β is a multitasking cytokine such that its aberrant expression is related to cancer progression and metastasis. TGF-β is produced by a variety of cells within the tumor microenvironment (TME), and it is responsible for regulation of the activity of cells within this milieu. TGF-β is a main inducer of epithelial-mesenchymal transition (EMT), immune evasion, and metastasis during cancer progression. TGF-β exerts most of its functions by acting on TβRI and TβRII receptors in canonical (Smad-dependent) or noncanonical (Smad-independent) pathways. Members of mitogen-activated protein kinase, phosphatidylinositol 3-kinase/protein kinase B, and nuclear factor κβ are involved in the non-Smad TGF-β pathway. TGF-β acts by complex signaling, and deletion in one of the effectors in this pathway may influence the outcome in a diverse way by taking even an antitumor role. The stage and the type of tumor (contextual cues from cancer cells and/or the TME) and the concentration of TGF-β are other important factors determining the fate of cancer (progression or repression). There are a number of ways for targeting TGF-β signaling in cancer, among them the special focus is on TβRII suppression.

    Topics: Carcinogenesis; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2019
Recent advances in understanding the roles of matrix metalloproteinases in tumour invasion and metastasis.
    The Journal of pathology, 2019, Volume: 247, Issue:5

    This review aims to provide an overview of recent developments regarding the roles of MMPs in tumour invasion and metastasis. Much of the mortality burden belonging to cancer relates to its ability to invade adjacent tissue and form metastases at distant sites. This would not be possible without remodelling of the ECM, a process which is enabled by the functions of MMPs. Recent studies provide a better understanding of the importance of the biophysical nature of the ECM, how this influences cancer cell motility, and how MMPs act to modify matrix stiffness. The regulation of MMPs and the role of immune cell generated MMPs has also become better understood. All of this provides a framework for the therapeutic targeting of MMPs and recent advances in the development of selective MMPs inhibitors are also reviewed. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

    Topics: Antigens, CD; beta-D-Galactoside alpha 2-6-Sialyltransferase; Extracellular Matrix; Humans; Immune System; Matrix Metalloproteinase Inhibitors; Matrix Metalloproteinases; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Receptors, G-Protein-Coupled; Sialyltransferases; Terminology as Topic; Thrombospondins; Transforming Growth Factor beta

2019
Phosphorylation status at Smad3 linker region modulates transforming growth factor-β-induced epithelial-mesenchymal transition and cancer progression.
    Cancer science, 2019, Volume: 110, Issue:2

    Smad3, a major transcription factor in transforming growth factor-β (TGF-β) signaling, plays critical roles in both tumor-suppressive and pro-oncogenic functions. Upon TGF-β stimulation, the C-terminal tail of Smad3 undergoes phosphorylation that is essential for canonical TGF-β signaling. The Smad3 linker region contains serine/threonine phosphorylation sites and can be phosphorylated by intracellular kinases, such as the MAPK family, cyclin-dependent kinase (CDK) family and glycogen synthase kinase-3β (GSK-3β). Previous reports based on cell culture studies by us and others showed that mutation of Smad3 linker phosphorylation sites dramatically intensifies TGF-β responses as well as growth-inhibitory function and epithelial-mesenchymal transition (EMT), suggesting that Smad3 linker phosphorylation suppresses TGF-β transcriptional activities. However, recent discoveries of Smad3-interacting molecules that preferentially bind phosphorylated Smad3 linker serine/threonine residues have shown a multitude of signal transductions that either enhance or suppress TGF-β responses associated with Smad3 turnover or cancer progression. This review aims at providing new insight into the perplexing mechanisms of TGF-β signaling affected by Smad3 linker phosphorylation and further attempts to gain insight into elimination and protection of TGF-β-mediated oncogenic and growth-suppressive signals, respectively.

    Topics: Animals; Disease Progression; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Phosphorylation; Smad3 Protein; Transforming Growth Factor beta

2019
TGF-β Family Signaling Pathways in Cellular Dormancy.
    Trends in cancer, 2019, Volume: 5, Issue:1

    Individual cancer cells can switch, reversibly, to a non-proliferative dormant state, a process characterized by two principal stages: (i) establishment and maintenance, and (ii) the breaking of dormancy. This phenomenon is of clinical importance because dormant cells resist chemotherapy, and this can result in cancer relapse following years, if not decades, of clinical remission. Although the molecular mechanisms governing tumor cell dormancy have not been clearly delineated, accumulating evidence suggests that members of the transforming growth factor-β (TGF-β) family are integral. We summarize here recent findings which support the view that TGF-β family signaling pathways play a pivotal role in cellular dormancy, and discuss how affected cells could be therapeutically targeted to prevent cancer relapse.

    Topics: Animals; Biomarkers; Cell Cycle Checkpoints; Epithelial-Mesenchymal Transition; Humans; Matrix Metalloproteinases; Neoplasms; Neoplastic Stem Cells; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2019
Targeting Hedgehog signaling pathway: Paving the road for cancer therapy.
    Pharmacological research, 2019, Volume: 141

    The Hedgehog pathway is essential for embryonic development but also for tissue and organ homeostasis in adult organisms. Activation of this pathway leads to the expression of target genes involved in proliferation, angiogenesis and stem cell self-renewal. Moreover, abnormal persistence of Hedgehog signaling is directly involved in a wide range of human cancers. Development of novel strategies targeting the Hedgehog pathway has become a subject of increased interest in anticancer therapy. These data are sustained by pre-clinical studies demonstrating that Hedgehog pathway inhibitors could represent an effective strategy against a heterogeneous panel of malignancies. Limited activity in other tumor types could be explained by the existence of crosstalk between the Hedgehog pathway and other signaling pathways that can compensate for its function. This review describes the Hedgehog pathway in detail, with its physiological roles during embryogenesis and adult tissues, and summarizing the preclinical evidence on its inhibition, the crosstalk between Hedgehog and other cancer-related pathways and finally the potential therapeutic effects of emerging compounds.

    Topics: Animals; Antineoplastic Agents; Hedgehog Proteins; Humans; Molecular Targeted Therapy; Neoplasms; Receptors, Notch; Receptors, Platelet-Derived Growth Factor; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2019
Dual role of TGF-β in early pregnancy: clues from tumor progression.
    Biology of reproduction, 2019, 06-01, Volume: 100, Issue:6

    TGF-β signaling in the endometrium is active during the implantation period and has a pivotal role in regulating endometrial receptivity and embryo implantation. During embryo implantation, both apoptosis and proliferation of endometrial cells happen at the same time and it seems TGF-β is the factor that controls both of these processes. As shown in cancer cells, in special conditions this cytokine can have a dual effect and switch the action from apoptosis to proliferation. Owing to the similarity between embryo implantation and cancer development and also unusual pattern of proliferation and remodeling in the uterus, in this review we suggest the existence of such a switching in endometrium during the early pregnancy. Moreover, we address some potential mechanisms that could regulate the switching. A better understanding of the molecular mechanisms regulating TGF-β action and signaling during the implantation period could pave the way for introducing novel therapeutic strategies in order to solve implantation-associated issues such as repeated implantation failure.

    Topics: Abortion, Spontaneous; Animals; Embryo Implantation; Female; Gene Expression Regulation; Humans; Neoplasms; Pregnancy; Transforming Growth Factor beta

2019
Opposing roles and potential antagonistic mechanism between TGF-β and BMP pathways: Implications for cancer progression.
    EBioMedicine, 2019, Volume: 41

    The transforming growth factor β (TGF-β) superfamily participates in tumour proliferation, apoptosis, differentiation, migration, invasion, immune evasion and extracellular matrix remodelling. Genetic deficiency in distinct components of TGF-β and BMP-induced signalling pathways or their excessive activation has been reported to regulate the development and progression of some cancers. As more in-depth studies about this superfamily have been conducted, more evidence suggests that the TGF-β and BMP pathways play an opposing role. The cross-talk of these 2 pathways has been widely studied in kidney disease and bone formation, and the opposing effects have also been observed in some cancers. However, the antagonistic mechanisms are still insufficiently investigated in cancer. In this review, we aim to display more evidences and possible mechanisms accounting for the antagonism between these 2 pathways, which might provide some clues for further study in cancer.

    Topics: Bone Morphogenetic Proteins; Carrier Proteins; CCCTC-Binding Factor; Humans; MicroRNAs; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2019
Specificity, versatility, and control of TGF-β family signaling.
    Science signaling, 2019, 02-26, Volume: 12, Issue:570

    Encoded in mammalian cells by 33 genes, the transforming growth factor-β (TGF-β) family of secreted, homodimeric and heterodimeric proteins controls the differentiation of most, if not all, cell lineages and many aspects of cell and tissue physiology in multicellular eukaryotes. Deregulation of TGF-β family signaling leads to developmental anomalies and disease, whereas enhanced TGF-β signaling contributes to cancer and fibrosis. Here, we review the fundamentals of the signaling mechanisms that are initiated upon TGF-β ligand binding to its cell surface receptors and the dependence of the signaling responses on input from and cooperation with other signaling pathways. We discuss how cells exquisitely control the functional presentation and activation of heteromeric receptor complexes of transmembrane, dual-specificity kinases and, thus, define their context-dependent responsiveness to ligands. We also introduce the mechanisms through which proteins called Smads act as intracellular effectors of ligand-induced gene expression responses and show that the specificity and impressive versatility of Smad signaling depend on cross-talk from other pathways. Last, we discuss how non-Smad signaling mechanisms, initiated by distinct ligand-activated receptor complexes, complement Smad signaling and thus contribute to cellular responses.

    Topics: Animals; Cell Differentiation; Gene Expression Regulation; Humans; Ligands; Neoplasms; Protein Binding; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2019
Transforming Growth Factor-β Signaling in Immunity and Cancer.
    Immunity, 2019, 04-16, Volume: 50, Issue:4

    Transforming growth factor (TGF)-β is a crucial enforcer of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Perturbations in TGF-β signaling underlie inflammatory diseases and promote tumor emergence. TGF-β is also central to immune suppression within the tumor microenvironment, and recent studies have revealed roles in tumor immune evasion and poor responses to cancer immunotherapy. Here, we present an overview of the complex biology of the TGF-β family and its context-dependent nature. Then, focusing on cancer, we discuss the roles of TGF-β signaling in distinct immune cell types and how this knowledge is being leveraged to unleash the immune system against the tumor.

    Topics: Adaptive Immunity; Animals; Dendritic Cells; Disease Progression; Epithelial-Mesenchymal Transition; Fibroblasts; Humans; Immunity, Innate; Inflammation; Macrophages; Mice, Knockout; Neoplasms; Neutrophils; Receptors, Transforming Growth Factor beta; Signal Transduction; T-Lymphocyte Subsets; Transforming Growth Factor beta; Tumor Escape; Tumor Microenvironment

2019
Perspectives of small molecule inhibitors of activin receptor‑like kinase in anti‑tumor treatment and stem cell differentiation (Review).
    Molecular medicine reports, 2019, Volume: 19, Issue:6

    Activin receptor‑like kinases (ALKs), members of the type I activin receptor family, belong to the serine/threonine kinase receptors of the transforming growth factor‑β (TGF‑β) superfamily. ALKs mediate the roles of activin/TGF‑β in a wide variety of physiological and pathological processes, ranging from cell differentiation and proliferation to apoptosis. For example, the activities of ALKs are associated with an advanced tumor stage in prostate cancer and the chondrogenic differentiation of mesenchymal stem cells. Therefore, potent and selective small molecule inhibitors of ALKs would not only aid in investigating the function of activin/TGF‑β, but also in developing treatments for these diseases via the disruption of activin/TGF‑β. In recent studies, several ALK inhibitors, including LY‑2157299, SB‑431542 and A‑83‑01, have been identified and have been confirmed to affect stem cell differentiation and tumor progression in animal models. This review discusses the therapeutic perspective of small molecule inhibitors of ALKs as drug targets in tumor and stem cells.

    Topics: Activin Receptors; Activins; Animals; Cell Differentiation; Humans; Mesenchymal Stem Cells; Neoplasms; Signal Transduction; Small Molecule Libraries; Transforming Growth Factor beta

2019
Blockade of TGF-β signaling: a potential target for cancer immunotherapy?
    Expert opinion on therapeutic targets, 2019, Volume: 23, Issue:8

    Topics: Animals; Disease Models, Animal; Humans; Immunotherapy; Neoplasms; Signal Transduction; T-Lymphocytes; Transforming Growth Factor beta; Tumor Microenvironment

2019
A New Switch for TGFβ in Cancer.
    Cancer research, 2019, 08-01, Volume: 79, Issue:15

    The TGFβ cytokine plays dichotomous roles during tumor progression. In normal and premalignant cancer cells, the TGFβ signaling pathway inhibits proliferation and promotes cell-cycle arrest and apoptosis. However, the activation of this pathway in late-stage cancer cells could facilitate the epithelial-to-mesenchymal transition, stemness, and mobile features to enhance tumorigenesis and metastasis. The opposite functions of TGFβ signaling during tumor progression make it a challenging target to develop anticancer interventions. Nevertheless, the recent discovery of cellular contextual determinants, especially the binding partners of the transcription modulators Smads, is critical to switch TGFβ responses from proapoptosis to prometastasis. In this review, we summarize the recently identified contextual determinants (such as PSPC1, KLF5, 14-3-3ζ, C/EBPβ, and others) and the mechanisms of how tumor cells manage the context-dependent autonomous TGFβ responses to potentiate tumor progression. With the altered expression of some contextual determinants and their effectors during tumor progression, the aberrant molecular prometastatic switch might serve as a new class of theranostic targets for developing anticancer strategies.

    Topics: Humans; Neoplasms; Transforming Growth Factor beta

2019
Dual Roles of the AMP-Activated Protein Kinase Pathway in Angiogenesis.
    Cells, 2019, 07-19, Volume: 8, Issue:7

    Angiogenesis plays important roles in development, stress response, wound healing, tumorigenesis and cancer progression, diabetic retinopathy, and age-related macular degeneration. It is a complex event engaging many signaling pathways including vascular endothelial growth factor (VEGF), Notch, transforming growth factor-beta/bone morphogenetic proteins (TGF-β/BMPs), and other cytokines and growth factors. Almost all of them eventually funnel to two crucial molecules, VEGF and hypoxia-inducing factor-1 alpha (HIF-1α) whose expressions could change under both physiological and pathological conditions. Hypoxic conditions stabilize HIF-1α, while it is upregulated by many oncogenic factors under normaxia. HIF-1α is a critical transcription activator for VEGF. Recent studies have shown that intracellular metabolic state participates in regulation of sprouting angiogenesis, which may involve AMP-activated protein kinase (AMPK). Indeed, AMPK has been shown to exert both positive and negative effects on angiogenesis. On the one hand, activation of AMPK mediates stress responses to facilitate autophagy which stabilizes HIF-1α, leading to increased expression of VEGF. On the other hand, AMPK could attenuate angiogenesis induced by tumor-promoting and pro-metastatic factors, such as the phosphoinositide 3-kinase /protein kinase B (Akt)/mammalian target of rapamycin (PI3K/Akt/mTOR), hepatic growth factor (HGF), and TGF-β/BMP signaling pathways. Thus, this review will summarize research progresses on these two opposite effects and discuss the mechanisms behind the discrepant findings.

    Topics: AMP-Activated Protein Kinases; Animals; Carcinogenesis; Diabetic Retinopathy; Eye; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Macular Degeneration; Mice; Neoplasms; Neovascularization, Pathologic; Neovascularization, Physiologic; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; TOR Serine-Threonine Kinases; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2019
TGF-β signaling in cell fate control and cancer.
    Current opinion in cell biology, 2019, Volume: 61

    Members of the transforming growth factor-β (TGF-β) family regulate cell fate decisions during early embryonic development and tissue homeostasis in the adult. Deregulation of TGF-β family signaling contributes to developmental anomalies, fibrotic disorders, tumorigenesis and immune diseases. TGF-β exerts a wide spectrum of cellular functions by activating canonical (SMAD-dependent) or non-canonical (SMAD-independent) pathways in a cell type-specific and context-dependent manner. Here, we focus on recent advances in the understanding of the mechanisms and functions of SMAD and non-SMAD pathways in physiology and pathology.

    Topics: Animals; Cell Lineage; Humans; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2019
The role of small GTPases of the Rho/Rac family in TGF-β-induced EMT and cell motility in cancer.
    Developmental dynamics : an official publication of the American Association of Anatomists, 2018, Volume: 247, Issue:3

    This article focuses on the role of Rho family GTPases, particularly Rac1 and Rac1b in TGF-β-induced epithelial-mesenchymal transition (EMT) and EMT-associated responses such as cell migration, invasion, and metastasis in cancer. EMT is considered a prerequisite for cells to adopt a motile and invasive phenotype and eventually become metastatic. A major regulator of EMT and metastasis in cancer is TGF-β, and its specific functions on tumor cells are mediated beside Smad proteins and mitogen-activated protein kinases (MAPKs) by small GTPases of the Rho/Rac1 family. Available data point to extensive signaling crosstalk between TGF-β and various Rho GTPases, and in particular a synergistic role of Rho and Rac1 during EMT and cell motility in normal and neoplastic epithelial cells. In contrast, the Rac1-related isoform, Rac1b, emerges as an endogenous inhibitor of Rac1 in TGF-β signaling, at least in pancreatic carcinoma cells. Given the tumor-promoting role of TGF-β in late-stage carcinomas and the intimate crosstalk of Rho/Rac1/Rac1b and TGF-β signaling in various tumor cell responses, targeting specific Rho GTPases may allow for selective interference with prooncogenic TGF-β responses to aid in anticancer treatments. Developmental Dynamics 247:451-461, 2018. © 2017 Wiley Periodicals, Inc.

    Topics: Cell Movement; Epithelial-Mesenchymal Transition; Monomeric GTP-Binding Proteins; Neoplasms; rac1 GTP-Binding Protein; Receptor Cross-Talk; rho GTP-Binding Proteins; Transforming Growth Factor beta

2018
Transforming growth factor-β, matrix metalloproteinases, and urokinase-type plasminogen activator interaction in the cancer epithelial to mesenchymal transition.
    Developmental dynamics : an official publication of the American Association of Anatomists, 2018, Volume: 247, Issue:3

    Transforming growth factor-β (TGF-β) is a pleiotropic factor that acts as a tumor suppressor in the early stages, while it exerts tumor promoting activities in advanced stages of cancer development. One of the hallmarks of cancer progression is the capacity of cancer cells to migrate and invade surrounding tissues with subsequent metastasis to different organs. Matrix metalloproteinases (MMPs) together with urokinase-type plasminogen activator (uPA) and its receptor (uPAR), whose main original function described is the proteolytic degradation of the extracellular matrix, play key cellular roles in the enhancement of cell malignancy during cancer progression. TGF-β tightly regulates the expression of several MMPs and uPA/uPAR in cancer cells, which in return can participate in TGF-β activation, thus contributing to tumor malignancy. TGF-β is one of the master factors in the induction of cancer-associated epithelial to mesenchymal transition (EMT), and recently both MMPs and uPA/uPAR have also been shown to be implicated in the cancer-associated EMT process. In this review, we analyze the main molecular mechanisms underlying MMPs and uPA/uPAR regulation by TGF-β, as well as their mutual implication in the development of EMT in cancer cells. Developmental Dynamics 247:382-395, 2018. © 2017 Wiley Periodicals, Inc.

    Topics: Animals; Cell Movement; Disease Progression; Epithelial-Mesenchymal Transition; Extracellular Matrix; Humans; Matrix Metalloproteinases; Neoplasms; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator

2018
Transforming Growth Factor-Beta and Urokinase Type Plasminogen Interplay in Cancer.
    Current protein & peptide science, 2018, Volume: 19, Issue:12

    Transforming growth factor-beta (TGF-β) is well recognized as playing a double role in tumor progression. Its antitumor role takes place in the early stages of cancer development, when TGF-β acts as a repressor of epithelial tumor growth. In advanced stages of cancer development, TGF-β has a tumor stimulating role, acting concomitantly with the increase of cancer cell migration and metastasis. One of the critical features of cancer cells is their ability to migrate and invade the surrounding tissues leading to metastases in different organs. Cancer cells that leave the tumor to infiltrate neighboring tissues and ultimately overtake a distant organ, need a complex and fine-regulated mechanism to move through the barrier imposed by the extracellular matrix (ECM). Therefore, cancer cells express a set of proteinases which are involved in the degradation and turnover of ECM. In particular, the urokinase type plasminogen activator (uPA) and the uPA cell surface receptor play key cellular roles in the enhancement of cell malignance during tumor progression. In normal cells uPA system is finely regulated, while in tumor cells its expression and activity are dysregulated in a way to enhance cells' invasion capacity during tumor progression. TGF-β strongly regulates uPA in cancer from transcriptional expression to enzyme activity. In turn, uPA participates in the activation of secreted latent TGF-β, thus producing a malicious loop which contributes to tumor progression and metastasis. In this review we will analyze the main molecular mechanisms implicated in uPA regulation by TGF-β. Moreover, the specific roles and interaction between TGF-β and uPA system in cancer cells and their impact on tumorigenesis will be portrayed.

    Topics: Animals; Carcinogenesis; Cell Membrane; Cell Movement; Extracellular Matrix; Humans; Neoplasm Metastasis; Neoplasms; Receptors, Urokinase Plasminogen Activator; Signal Transduction; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator

2018
TGF‑β signaling: A complex role in tumorigenesis (Review).
    Molecular medicine reports, 2018, Volume: 17, Issue:1

    Tumor progression can be affected by various cellular components of tumor cells and/or by tumor microenvironmental factors. The tumor microenvironment comprises a variety of nonmalignant stromal cells and inflammatory cytokines, which are pivotal in tumor promotion and progression. The transforming growth factor‑β (TGF‑β) ligands (TGF‑β1, 2 and 3) are secreted inflammatory cytokines, which are known to be involved in various aspects of tumor development through two transmembrane serine‑threonine kinase receptors, TGFβR1 and TGFβR2. TGF‑β promotes or inhibits tumorigenesis depending on the concurrent gene mutations and tissue microenvironment present through the small mothers against decapentaplegic (Smad) and non‑Smad pathways. This review aims to provide a comprehensive overview of the role of the TGF‑β pathway in tumor initiation and progression.

    Topics: Carcinogenesis; Humans; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Stromal Cells; Transforming Growth Factor beta; Tumor Microenvironment

2018
TGF-β signaling in cancer metastasis.
    Acta biochimica et biophysica Sinica, 2018, Jan-01, Volume: 50, Issue:1

    The transforming growth factor (TGF)-β signaling events are well known to control diverse processes and numerous responses, such as cell proliferation, differentiation, apoptosis, and migration. TGF-β signaling plays context-dependent roles in cancer: in pre-malignant cells TGF-β primarily functions as a tumor suppressor, while in the later stages of cancer TGF-β signaling promotes invasion and metastasis. Recent studies have also suggested that the cross-talk between TGF-β signaling and other signaling pathways, such as Hippo, Wnt, EGFR/RAS, and PI3K/AKT pathways, may substantially contribute to our current understanding of TGF-β signaling and cancer. As a result of the wide-ranging effects of TGF-β, blockade of TGF-β and its downstream signaling components provides multiple therapeutic opportunities. Therefore, the outlook for anti-TGF-β signaling therapy for numerous diseases appears bright and will provide valuable information and thinking on the drug molecular design. In this review, we focus on recent insights into the regulation of TGF-β signaling in cancer metastasis which may contribute to the development of novel cancer-targeting therapies.

    Topics: Antineoplastic Agents; Disease Progression; Humans; Models, Biological; Molecular Targeted Therapy; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2018
Dually modified transmembrane proteoglycans in development and disease.
    Cytokine & growth factor reviews, 2018, Volume: 39

    Aberrant cell signaling in response to secreted growth factors has been linked to the development of multiple diseases, including cancer. As such, understanding mechanisms that control growth factor availability and receptor-growth factor interaction is vital. Dually modified transmembrane proteoglycans (DMTPs), which are classified as cell surface macromolecules composed of a core protein decorated with covalently linked heparan sulfated (HS) and/or chondroitin sulfated (CS) glycosaminoglycan (GAG) chains, provide one type of regulatory mechanism. Specifically, DMTPs betaglycan and syndecan-1 (SDC1) play crucial roles in modulating key cell signaling pathways, such as Wnt, transforming growth factor-β and fibroblast growth factor signaling, to affect epithelial cell biology and cancer progression. This review outlines current and potential functions for betaglycan and SDC1, with an emphasis on comparing individual roles for HS and CS modified DMTPs. We highlight the mutual dependence of DMTPs' GAG chains and core proteins and provide comprehensive knowledge on how these DMTPs, through regulation of ligand availability and receptor internalization, control cell signaling pathways involved in development and disease.

    Topics: Animals; Chondroitin Sulfates; Epithelial Cells; Glycosaminoglycans; Heparitin Sulfate; Humans; Intercellular Signaling Peptides and Proteins; Mice; Neoplasms; Proteoglycans; Receptors, Transforming Growth Factor beta; Signal Transduction; Syndecan-1; Transforming Growth Factor beta; Wnt Proteins

2018
Viruses as key modulators of the TGF-β pathway; a double-edged sword involved in cancer.
    Reviews in medical virology, 2018, Volume: 28, Issue:2

    Transforming growth factor-β (TGF-β) signaling pathway is a key network in cell signaling that controls vital processes such as proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and migration, thus acting as a double-edged sword in normal development and diseases, in particular organ fibrosis, vascular disorders, and cancer. Early in tumorigenesis, the pathway exerts anti-tumor effects through suppressing cell cycle and inducing apoptosis, while during late stages, it functions as a tumor promoter by enhancing tumor invasiveness and metastasis. This signaling pathway can be perturbed by environmental and genetic factors such as microbial interference and mutation, respectively. In this way, the present review describes the modulation of the TGF-β pathway by oncogenic human viral pathogens and other viruses. The main mechanisms by which viruses interferes with TGF-β signaling seems to be through (1) the alteration of either TGF-β protein expression or activation, (2) the modulation of the TGF-β receptors or SMADs factors (by interfering with their levels and functions), (3) the alteration of none-SMAD pathways, and (4) indirect interaction with the pathway by the modulation of transcriptional co-activator/repressor and regulators of the pathway. Given the axial role of this pathway in tumorigenesis, it can be regarded as an attractive target for cancer therapy. Hence, further investigations on this subject may represent molecular targets among either TGF-β signaling molecules or viral factors for the treatment and management of viral infection consequences such as cancer.

    Topics: Animals; Cell Transformation, Viral; Host-Pathogen Interactions; Humans; Neoplasms; Oncogenic Viruses; Signal Transduction; Transforming Growth Factor beta; Tumor Virus Infections; Virus Physiological Phenomena

2018
Immunoregulatory functions and the therapeutic implications of GARP-TGF-β in inflammation and cancer.
    Journal of hematology & oncology, 2018, 02-20, Volume: 11, Issue:1

    GARP (glycoprotein-A repetitions predominant) is a type I transmembrane cell surface docking receptor for latent transforming growth factor-β (TGF-β) that is abundantly expressed on regulatory T lymphocytes and platelets. GARP regulates the availability of membrane-bound latent TGF-β and modulates its activation. For this reason, GARP expression on immune and non-immune cells is involved in maintaining peripheral tolerance. It plays an important role in preventing inflammatory diseases such as allergy and graft versus host disease (GvHD). GARP is also frequently hijacked by cancer cells to promote oncogenesis. This review summarizes the most important features of GARP biology described to date including gene regulation, protein expression and mechanism in activating latent TGF-β, and the function of GARP in regulatory T cell biology and peripheral tolerance, as well as GARP's increasingly recognized roles in platelet-mediated cancer immune evasion. The promise for GARP-targeted strategy as a novel immunotherapy of cancer is also highlighted.

    Topics: Animals; Blood Platelets; Gene Expression Regulation, Neoplastic; Humans; Immune Tolerance; Inflammation; Membrane Proteins; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2018
The role of TGF-β/SMAD4 signaling in cancer.
    International journal of biological sciences, 2018, Volume: 14, Issue:2

    Transforming growth factor β (TGF-β) signaling pathway plays important roles in many biological processes, including cell growth, differentiation, apoptosis, migration, as well as cancer initiation and progression. SMAD4, which serves as the central mediator of TGF-β signaling, is specifically inactivated in over half of pancreatic duct adenocarcinoma, and varying degrees in many other types of cancers. In the past two decades, multiple studies have revealed that SMAD4 loss on its own does not initiate tumor formation, but can promote tumor progression initiated by other genes, such as KRAS activation in pancreatic duct adenocarcinoma and APC inactivation in colorectal cancer. In other cases, such as skin cancer, loss of SMAD4 plays an important initiating role by disrupting DNA damage response and repair mechanisms and enhance genomic instability, suggesting its distinct roles in different types of tumors. This review lists SMAD4 mutations in various types of cancer and summarizes recent advances on SMAD4 with focuses on the function, signaling pathway, and the possibility of SMAD4 as a prognostic indicator.

    Topics: Animals; DNA Damage; DNA Repair; Epithelial-Mesenchymal Transition; Mice; Models, Molecular; Mutation; Neoplasms; Prognosis; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta

2018
Transcriptional and post-transcriptional regulation of the genes encoding the small GTPases RhoA, RhoB, and RhoC: implications for the pathogenesis of human diseases.
    Cellular and molecular life sciences : CMLS, 2018, Volume: 75, Issue:12

    Rho GTPases are highly conserved proteins that play critical roles in many cellular processes including actin dynamics, vesicular trafficking, gene transcription, cell-cycle progression, and cell adhesion. The main mode of regulation of Rho GTPases is through guanine nucleotide binding (cycling between an active GTP-bound form and an inactive GDP-bound form), but transcriptional, post-transcriptional, and post-translational modes of Rho regulation have also been described. In the present review, we summarize recent progress on the mechanisms that control the expression of the three members of the Rho-like subfamily (RhoA, RhoB, and RhoC) at the level of gene transcription as well as their post-transcriptional regulation by microRNAs. We also discuss the progress made in deciphering the mechanisms of cross-talk between Rho proteins and the transforming growth factor β signaling pathway and their implications for the pathogenesis of human diseases such as cancer metastasis and fibrosis.

    Topics: Animals; Gene Expression Regulation; Humans; MicroRNAs; Neoplasms; rhoA GTP-Binding Protein; rhoB GTP-Binding Protein; rhoC GTP-Binding Protein; Signal Transduction; Transcriptional Activation; Transforming Growth Factor beta

2018
Microfibril-associated glycoproteins MAGP-1 and MAGP-2 in disease.
    Matrix biology : journal of the International Society for Matrix Biology, 2018, Volume: 71-72

    Microfibril-associated glycoproteins 1 and 2 (MAGP-1, MAGP-2) are protein components of extracellular matrix microfibrils. These proteins interact with fibrillin, the core component of microfibrils, and impart unique biological properties that influence microfibril function in vertebrates. MAGPs bind active forms of TGFβ and BMPs and are capable of modulating Notch signaling. Mutations in MAGP-1 or MAGP-2 have been linked to thoracic aneurysms and metabolic disease in humans. MAGP-2 has also been shown to be an important biomarker in several human cancers. Mice lacking MAGP-1 or MAGP-2 have defects in multiple organ systems, which reflects the widespread distribution of microfibrils in vertebrate tissues. This review summarizes our current understanding of the function of the MAGPs and their relationship to human disease.

    Topics: Animals; Aortic Aneurysm, Thoracic; Biomarkers; Bone Morphogenetic Proteins; Contractile Proteins; Extracellular Matrix; Extracellular Matrix Proteins; Humans; Metabolic Diseases; Mice; Mutation; Neoplasms; Receptors, Notch; RNA Splicing Factors; Signal Transduction; Transforming Growth Factor beta

2018
The Long (lncRNA) and Short (miRNA) of It: TGFβ-Mediated Control of RNA-Binding Proteins and Noncoding RNAs.
    Molecular cancer research : MCR, 2018, Volume: 16, Issue:4

    RNA-binding proteins (RBP) and noncoding RNAs (ncRNA), such as long noncoding RNAs (lncRNA) and microRNAs (miRNA), control co- and posttranscriptional gene regulation (PTR). At the PTR level, RBPs and ncRNAs contribute to pre-mRNA processing, mRNA maturation, transport, localization, turnover, and translation. Deregulation of RBPs and ncRNAs promotes the onset of cancer progression and metastasis. Both RBPs and ncRNAs are altered by signaling cascades to cooperate or compete with each other to bind their nucleic acid targets. Most importantly, transforming growth factor-beta (TGFβ) signaling plays a significant role in controlling gene expression patterns by targeting RBPs and ncRNAs. Because of TGFβ signaling in cancer, RBP-RNA or RNA-RNA interactions are altered and cause enhanced cell growth and tumor cell dissemination. This review focuses on the emerging concepts of TGFβ signaling on posttranscriptional gene regulation and highlights the implications of RBPs and ncRNAs in cancer progression and metastasis.

    Topics: Animals; Disease Progression; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Neoplasm Metastasis; Neoplasms; RNA Processing, Post-Transcriptional; RNA-Binding Proteins; RNA, Long Noncoding; Signal Transduction; Transforming Growth Factor beta

2018
Regulatory T cells: a potential target in cancer immunotherapy.
    Annals of the New York Academy of Sciences, 2018, Volume: 1417, Issue:1

    Cancer immunotherapy involving blockade of immune checkpoint molecules, such as CTLA-4 and PD-1, has shown remarkable clinical success across several types of malignancies. However, a fraction of patients experience disease progression after treatment; thus, exploring resistant mechanisms for immune checkpoint inhibitors and improving their treatment outcome with additional modalities are of great importance. CD4

    Topics: Antibodies, Monoclonal; CTLA-4 Antigen; Glucocorticoid-Induced TNFR-Related Protein; HSP70 Heat-Shock Proteins; Humans; Immunotherapy; Indoleamine-Pyrrole 2,3,-Dioxygenase; Interleukin-2 Receptor alpha Subunit; Models, Immunological; Neoplasms; Phosphoinositide-3 Kinase Inhibitors; Receptors, CCR4; Receptors, OX40; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2018
Signaling Crosstalk of TGF-β/ALK5 and PAR2/PAR1: A Complex Regulatory Network Controlling Fibrosis and Cancer.
    International journal of molecular sciences, 2018, May-24, Volume: 19, Issue:6

    Both signaling by transforming growth factor-β (TGF-β) and agonists of the G Protein-coupled receptors proteinase-activated receptor-1 (PAR1) and -2 (PAR2) have been linked to tissue fibrosis and cancer. Intriguingly, TGF-β and PAR signaling either converge on the regulation of certain matrix genes overexpressed in these pathologies or display mutual regulation of their signaling components, which is mediated in part through sphingosine kinases and sphingosine-1-phosphate and indicative of an intimate signaling crosstalk between the two pathways. In the first part of this review, we summarize the various regulatory interactions that have been discovered so far according to the organ/tissue in which they were described. In the second part, we highlight the types of signaling crosstalk between TGF-β on the one hand and PAR2/PAR1 on the other hand. Both ligand⁻receptor systems interact at various levels and by several mechanisms including mutual regulation of ligand⁻ligand, ligand⁻receptor, and receptor⁻receptor at the transcriptional, post-transcriptional, and receptor transactivation levels. These mutual interactions between PAR2/PAR1 and TGF-β signaling components eventually result in feed-forward loops/vicious cycles of matrix deposition and malignant traits that exacerbate fibrosis and oncogenesis, respectively. Given the crucial role of PAR2 and PAR1 in controlling TGF-β receptor activation, signaling, TGF-β synthesis and bioactivation, combining PAR inhibitors with TGF-β blocking agents may turn out to be more efficient than targeting TGF-β alone in alleviating unwanted TGF-β-dependent responses but retaining the beneficial ones.

    Topics: Animals; Humans; Kidney; Liver Cirrhosis; Lung; Neoplasms; Protein Serine-Threonine Kinases; Receptor, PAR-1; Receptor, PAR-2; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2018
Negative regulation of TGF-β by AMPK and implications in the treatment of associated disorders.
    Acta biochimica et biophysica Sinica, 2018, Jun-01, Volume: 50, Issue:6

    Transforming growth factor beta (TGF-β) regulates a large number of biological processes, including proliferation, differentiation, immune response, and development. In addition, TGF-β plays important roles in some pathological processes, for instance, it is upregulated and activated in fibrosis and advanced cancer. Adenosine monophosphate-activated protein kinase (AMPK) acts as a fuel gauge that is activated when cells sense shortage of ATP and increase in AMP or AMP:ATP ratio. Activation of AMPK slows down anabolic processes and stimulates catabolic processes, leading to increased production of ATP. Furthermore, the functions of AMPK have been extended beyond energy homeostasis. In fact, AMPK has been shown to exert a tumor suppressive effect. Recent studies have demonstrated negative impacts of AMPK on TGF-β function. Therefore, in this review, we will discuss the differences in the biological functions of TGF-β and AMPK, and some pathological processes such as fibrosis, epithelial-mesenchymal transition (EMT) and cancer metastasis, as well as angiogenesis and heterotopic ossifications where TGF-β and AMPK exert opposite effects.

    Topics: AMP-Activated Protein Kinases; Animals; Energy Metabolism; Epithelial-Mesenchymal Transition; Humans; Models, Biological; Neoplasms; Phosphorylation; Signal Transduction; Transforming Growth Factor beta

2018
TMEPAI family: involvement in regulation of multiple signalling pathways.
    Journal of biochemistry, 2018, Sep-01, Volume: 164, Issue:3

    The TMEPAI family, composed of TMEPAI and C18ORF1, is known to inhibit transforming growth factor-β (TGF-β) signalling via its competition for binding of receptor-regulated Smad with Smad anchor for receptor activation. However, TMEPAI has also been reported to be involved in androgen receptor signalling, phosphatase and tensin homologue deleted on chromosome 10 signalling, and formation of autophagosomes in addition to degradation of TβRI (TGF-β type I receptor) through lysosomes. Thus, TMEPAI seems to act as a regulator of multiple signalling pathways. A great deal of attention has already been paid to the relationship between the TMEPAI family and tumourigenicity. In this paper, therefore, we describe recent progresses in the understanding of how the TMEPAI family physiologically contributes to cellular functions and diseases.

    Topics: Gene Expression Regulation; Humans; Membrane Proteins; Neoplasms; Proteolysis; Receptors, Androgen; Receptors, Transforming Growth Factor beta; Signal Transduction; Subcellular Fractions; Transcription, Genetic; Transforming Growth Factor beta

2018
TGF-β and mesenchymal stromal cells in regenerative medicine, autoimmunity and cancer.
    Cytokine & growth factor reviews, 2018, Volume: 43

    Multipotent mesenchymal stromal cells (MSCs) represent a promising cell-based therapy in regenerative medicine and for the treatment of inflammatory/autoimmune diseases. Importantly, MSCs have emerged as an important contributor to the tumor stroma with both pro- and anti-tumorigenic effects. However, the successful translation of MSCs to the clinic and the prevention of their tumorigenic and metastatic effect require a greater understanding of factors controlling their proliferation, differentiation, migration and immunomodulation in vitro and in vivo. The transforming growth factor(TGF)-β1, 2 and 3 are involved in almost every aspect of MSC function. The aim of this review is to highlight the roles that TGF-β play in the biology and therapeutic applications of MSCs. We will discuss the how TGF-β modulate MSC function as well as the paracrine effects of MSC-derived TGF-β on other cell types in the context of tissue regeneration, immune responses and cancer. Finally, taking all these aspects into consideration we discuss how modulation of TGF-β signaling/production in MSCs could be of clinical interest.

    Topics: Animals; Autoimmunity; Cell Differentiation; Cell Proliferation; Humans; Immunomodulation; Mesenchymal Stem Cells; Neoplasms; Regenerative Medicine; Transforming Growth Factor beta

2018
MicroRNA Control of TGF-β Signaling.
    International journal of molecular sciences, 2018, 06-28, Volume: 19, Issue:7

    Transcriptional and post-transcriptional regulation shapes the transcriptome and proteome changes induced by various cellular signaling cascades. MicroRNAs (miRNAs) are small regulatory RNAs that are approximately 22 nucleotides long, which direct the post-transcriptional regulation of diverse target genes and control cell states. Transforming growth factor (TGF)-β family is a multifunctional cytokine family, which plays many regulatory roles in the development and pathogenesis of diverse diseases, including fibrotic disease, cardiovascular disease and cancer. Previous studies have shown that the TGF-β pathway includes the miRNA pathway as an important component of its downstream signaling cascades. Multiple studies of epithelial⁻mesenchymal transition (EMT)-related miRNAs have highlighted that miRNAs constitute the intrinsic bistable molecular switches of cell states by forming double negative feedback loops with EMT-inducing transcription factors. This may be important for understanding the reversibility of EMT at the single-cell level, the presence of distinct EMT transition states and the intra- and inter-tumor heterogeneity of cancer cell phenotypes. In the present review, I summarize the connection between TGF-β signaling and the miRNA pathway, placing particular emphasis on the regulation of miRNA expression by TGF-β signaling, the modulation of TGF-β signaling by miRNAs, the miRNA-mediated modulation of EMT and endothelial⁻mesenchymal transition as well as the crosstalk between miRNA and TGF-β pathways in the tumor microenvironment.

    Topics: Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Neoplasms; Signal Transduction; Transcriptome; Transforming Growth Factor beta; Tumor Microenvironment

2018
Long non-coding RNA activated by TGF-β expression in cancer prognosis: A meta-analysis.
    International journal of surgery (London, England), 2018, Volume: 58

    Recently, long non-coding RNA activated by transforming growth factor beta (TGF-β) (lncRNA ATB) was shown to be useful in cancer prognosis, however, its prognostic value in human cancer has been inconsistent. Our study aimed to explore the prognostic role of lncRNA ATB expression in cancer prognosis.. PubMed, Embase, and Cochrane Library databases were thoroughly searched to retrieve studies focusing on the prognostic role of lncRNA ATB expression in cancer, and meta-analysis was performed.. A total of 15 studies were included into this meta-analysis. High lncRNA ATB expression was significantly related to shorter overall survival (OS) (HR = 2.44, 95%CI = 1.98-3.01, P < 0.01), recurrence-free survival (RFS) (HR = 1.85, 95%CI = 1.42-2.40, P < 0.01), disease-free survival (DFS) (HR = 3.61, 95%CI = 2.45-5.33, P < 0.01), and progression-free survival (PFS) (HR = 2.97, 95%CI = 2.12-4.16, P < 0.01) when compared with low lncRNA ATB expression in cancer. Moreover, Patients with high lncRNA ATB expression tended to have worse tumor differentiation (P < 0.01), more advanced clinical stage (P < 0.01), deeper tumor invasion (P < 0.01), earlier distant metastases (P = 0.02), lymph node metastases (P = 0.04), and vascular invasion (P < 0.01) when compared with those with low lncRNA ATB expression.. High lncRNA ATB expression was significantly associated with worse prognosis in cancer. LncRNA ATB expression could be used as a prognostic biomarker for human cancer.

    Topics: Female; Humans; Lymphatic Metastasis; Neoplasms; Prognosis; RNA, Long Noncoding; Transforming Growth Factor beta

2018
Targeting the divergent TGFβ superfamily cytokine MIC-1/GDF15 for therapy of anorexia/cachexia syndromes.
    Current opinion in supportive and palliative care, 2018, Volume: 12, Issue:4

    To review recent finding on MIC-1/GDF15 and re-evaluate it as a potential target for the therapy of anorexia/cachexia syndromes.. MIC-1/GDF15 consistently induces anorexia/cachexia in animal models. Its actions on brainstem feeding centers leads to anorexia, inducing prolonged undernutrition and consequent loss of both lean and fat mass. Epidemiological studies by multiple groups have linked substantially elevated serum levels of this cytokine to anorexia/cachexia syndromes in diverse diseases such as cancer, chronic renal and cardiac failure, and chronic obstructive lung disease. These elevated serum levels are similar to those required to induce this syndrome in animals. Recent identifications of its previously elusive receptor as GFRAL, has enhanced understanding of its biology and suggests that modulating the MIC-1/GDF15-GFRAL pathway may be a therapeutic target for anorexia/cachexia syndrome.. Inhibiting MIC-1/GDF15 or its receptor GFRAL are high-value potential targets for treatment of anorexia/cachexia syndrome in patients whose elevated serum levels may justify its use.

    Topics: Animals; Anorexia; Cachexia; Glial Cell Line-Derived Neurotrophic Factor; Growth Differentiation Factor 15; Humans; Neoplasms; Syndrome; Transforming Growth Factor beta

2018
The immune regulation in cancer by the amino acid metabolizing enzymes ARG and IDO.
    Current opinion in pharmacology, 2017, Volume: 35

    Some enzymes degrading amino acids have evolved in mammals to dampen immune responses and maintain peripheral tolerance. The enzymes metabolizing l-arginine and l-tryptophan are particularly powerful, contributing to restrain immunity towards fetal tissues and establish neonatal tolerance. Solid tumors can hijack these formidable pathways to construct a microenvironment highly unfavorable to anti-tumor T lymphocytes able to recognize them, one of mechanisms for their immune evasion. In this review, we analyze emerging concepts in the cross-talk between cells expressing these enzymes, their immune regulatory functions and pharmacological approaches that can target them to enhance cancer immunotherapy.

    Topics: Animals; Arginase; Dendritic Cells; Humans; Indoleamine-Pyrrole 2,3,-Dioxygenase; Neoplasms; Transforming Growth Factor beta; Tryptophan; Tumor Microenvironment

2017
Transforming growth factor-β: A therapeutic target for cancer.
    Human vaccines & immunotherapeutics, 2017, 08-03, Volume: 13, Issue:8

    Transforming growth factor-β (TGF-β) regulates cell growth and differentiation, apoptosis, cell motility, extracellular matrix production, angiogenesis, and cellular immunity. It has a paradoxical role in cancer. In the early stages it inhibits cellular transformation and prevents cancer progression. In later stages TGF-β plays a key role in promoting tumor progression through mainly 3 mechanisms: facilitating epithelial to mesenchymal transition, stimulating angiogenesis and inducing immunosuppression. As a result of its opposing tumor promoting and tumor suppressive abilities, TGF-β and its pathway has represented potential opportunities for drug development and several therapies targeting the TGF-β pathway have been identified. This review focuses on identifying the mechanisms through which TGF-β is involved in tumorigenesis and current therapeutics that are under development.

    Topics: Animals; Antibodies, Monoclonal; Antineoplastic Agents; Apoptosis; Cell Differentiation; Epithelial-Mesenchymal Transition; Humans; Mice; Neoplasms; Neovascularization, Pathologic; Oligonucleotides, Antisense; Signal Transduction; Transforming Growth Factor beta

2017
Epithelial-mesenchymal transition in cancer metastasis through the lymphatic system.
    Molecular oncology, 2017, Volume: 11, Issue:7

    It was already in the 18th century when the French surgeon LeDran first noted that breast cancer patients with spread of tumor cells to their axillary lymph nodes had a drastically worse prognosis than patients without spread (LeDran et al., ). Since then, metastatic spread of cancer cells to regional lymph nodes has been established as the most important prognostic factor in many types of cancer (Carter et al., ; Elston and Ellis, ). However, despite its clinical importance, lymph metastasis remains an underexplored area of tumor biology. Fundamental questions, such as when, how, and perhaps most importantly, why tumor cells disseminate through the lymphatic system, remain largely unanswered. Accordingly, no treatment strategies exist that specifically target lymph metastasis. The identification of epithelial-mesenchymal transition (EMT) as a mechanism, which allows cancer cells to dedifferentiate and acquire enhanced migratory and invasive properties, has been a game changer in cancer research. Conceptually, EMT provides an explanation for why epithelial cancers with poor differentiation status are generally more aggressive and prone to metastasize than more differentiated cancers. Inflammatory cytokines, such as TGF-β, which are produced and secreted by tumor-infiltrating immune cells, are potent inducers of EMT. Thus, reactivation of EMT also links cancer-related inflammation to invasive and metastatic disease. Recently, we found that breast cancer cells undergoing TGF-β-induced EMT acquire properties of immune cells allowing them to disseminate in a targeted fashion through the lymphatic system similar to activated dendritic cells during inflammation. Here, we review our current understanding of the mechanisms by which cancer cells spread through the lymphatic system and the links to inflammation and the immune system. We also emphasize how imaging techniques have the potential to further expand our knowledge of the mechanisms of lymph metastasis, and how lymph nodes serve as an interface between cancer and the immune system.

    Topics: Animals; Epithelial-Mesenchymal Transition; Humans; Lymph Nodes; Lymphatic Metastasis; Neoplasm Proteins; Neoplasms; Transforming Growth Factor beta

2017
Tumor-Related Exosomes Contribute to Tumor-Promoting Microenvironment: An Immunological Perspective.
    Journal of immunology research, 2017, Volume: 2017

    Exosomes are a kind of cell-released membrane-form structures which contain proteins, lipids, and nucleic acids. These vesicular organelles play a key role in intercellular communication. Numerous experiments demonstrated that tumor-related exosomes (TEXs) can induce immune surveillance in the microenvironment in vivo and in vitro. They can interfere with the maturation of DC cells, impair NK cell activation, induce myeloid-derived suppressor cells, and educate macrophages into protumor phenotype. They can also selectively induce effector T cell apoptosis via Fas/FasL interaction and enhance regulatory T cell proliferation and function by releasing TGF-

    Topics: Animals; Apoptosis; Cell Proliferation; Exosomes; Humans; Immune Tolerance; Immunotherapy; Lymphocyte Activation; Mice; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2017
Targeting TGF-β Signaling in Cancer.
    Trends in cancer, 2017, Volume: 3, Issue:1

    The transforming growth factor (TGF)-β signaling pathway is deregulated in many diseases, including cancer. In healthy cells and early-stage cancer cells, this pathway has tumor-suppressor functions, including cell-cycle arrest and apoptosis. However, its activation in late-stage cancer can promote tumorigenesis, including metastasis and chemoresistance. The dual function and pleiotropic nature of TGF-β signaling make it a challenging target and imply the need for careful therapeutic dosing of TGF-β drugs and patient selection. We review here the rationale for targeting TGF-β signaling in cancer and summarize the clinical status of pharmacological inhibitors. We discuss the direct effects of TGF-β signaling blockade on tumor and stromal cells, as well as biomarkers that can predict the efficacy of TGF-β inhibitors in cancer patients.

    Topics: Animals; Biomarkers, Tumor; Drug Resistance, Neoplasm; Humans; Neoplasms; Precision Medicine; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2017
SOX4: Joining the Master Regulators of Epithelial-to-Mesenchymal Transition?
    Trends in cancer, 2017, Volume: 3, Issue:8

    The epithelial-to-mesenchymal transition (EMT) is an important developmental program exploited by cancer cells to gain mesenchymal features. Transcription factors globally regulating processes during EMT are often referred as 'master regulators' of EMT, and include members of the Snail and ZEB transcription factor families. The SRY-related HMG box (SOX) 4 transcription factor can promote tumorigenesis by endowing cells with migratory and invasive properties, stemness, and resistance to apoptosis, thereby regulating key aspects of the EMT program. We propose here that SOX4 should also be considered as a master regulator of EMT, and we review the molecular mechanisms underlying its function.

    Topics: Antineoplastic Agents; Carcinogenesis; Cell Movement; Cell Transformation, Neoplastic; Disease Progression; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Neoplasm Invasiveness; Neoplasm Recurrence, Local; Neoplasms; Signal Transduction; SOXC Transcription Factors; Transforming Growth Factor beta; Tumor Suppressor Proteins

2017
The roles of ubiquitin modifying enzymes in neoplastic disease.
    Biochimica et biophysica acta. Reviews on cancer, 2017, Volume: 1868, Issue:2

    The initial experiments performed by Rose, Hershko, and Ciechanover describing the identification of a specific degradation signal in short-lived proteins paved the way to the discovery of the ubiquitin mediated regulation of numerous physiological functions required for cellular homeostasis. Since their discovery of ubiquitin and ubiquitin function over 30years ago it has become wholly apparent that ubiquitin and their respective ubiquitin modifying enzymes are key players in tumorigenesis. The human genome encodes approximately 600 putative E3 ligases and 80 deubiquitinating enzymes and in the majority of cases these enzymes exhibit specificity in sustaining either pro-tumorigenic or tumour repressive responses. In this review, we highlight the known oncogenic and tumour suppressive effects of ubiquitin modifying enzymes in cancer relevant pathways with specific focus on PI3K, MAPK, TGFβ, WNT, and YAP pathways. Moreover, we discuss the capacity of targeting DUBs as a novel anticancer therapeutic strategy.

    Topics: Animals; Cell Cycle Proteins; Deubiquitinating Enzymes; Humans; MAP Kinase Signaling System; Neoplasms; Nuclear Proteins; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Smad Proteins; Transcription Factors; Transforming Growth Factor beta; Ubiquitin; Ubiquitin-Protein Ligases; Wnt Signaling Pathway

2017
Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases.
    Immunological reviews, 2017, Volume: 280, Issue:1

    Ionizing radiation is often regarded as an element of danger. But, danger responses on the cellular and molecular level are often beneficial with regard to the induction of anti-tumor immunity and for amelioration of inflammation. We outline how in dependence of radiation dose and fraction, radiation itself-and especially in combination with immune modulators-impacts on the innate and adaptive immune system. Focus is set on radiation-induced changes of the tumor cell phenotype and the cellular microenvironment including immunogenic cancer cell death. Mechanisms how anti-tumor immune responses are triggered by radiotherapy in combination with hyperthermia, inhibition of apoptosis, the adjuvant AnnexinA5, or vaccination with high hydrostatic pressure-killed autologous tumor cells are discussed. Building on this, feasible multimodal radio-immunotherapy concepts are reviewed including overcoming immune suppression by immune checkpoint inhibitors and by targeting TGF-β. Since radiation-induced tissue damage, inflammation, and anti-tumor immune responses are interconnected, the impact of lower doses of radiation on amelioration of inflammation is outlined. Closely meshed immune monitoring concepts based on the liquid biopsy blood are suggested for prognosis and prediction of cancer and non-cancer inflammatory diseases. Finally, challenges and visions for the design of cancer radio-immunotherapies and for treatment of benign inflammatory diseases are given.

    Topics: Animals; Cell Death; Humans; Immune System Diseases; Immunity; Immunomodulation; Inflammation; Molecular Targeted Therapy; Neoplasms; Radiation, Ionizing; Radioimmunotherapy; Transforming Growth Factor beta; Tumor Microenvironment; Vaccination

2017
Proteinase-Activated Receptor 2 May Drive Cancer Progression by Facilitating TGF-β Signaling.
    International journal of molecular sciences, 2017, Nov-22, Volume: 18, Issue:11

    The G protein-coupled receptor proteinase-activated receptor 2 (PAR2) has been implicated in various aspects of cellular physiology including inflammation, obesity and cancer. In cancer, it usually acts as a driver of cancer progression in various tumor types by promoting invasion and metastasis in response to activation by serine proteinases. Recently, we discovered another mode through which PAR2 may enhance tumorigenesis: crosstalk with transforming growth factor-β (TGF-β) signaling to promote TGF-β1-induced cell migration/invasion and invasion-associated gene expression in ductal pancreatic adenocarcinoma (PDAC) cells. In this chapter, we review what is known about the cellular TGF-β responses and signaling pathways affected by PAR2 expression, the signaling activities of PAR2 required for promoting TGF-β signaling, and the potential molecular mechanism(s) that underlie(s) the TGF-β signaling-promoting effect. Since PAR2 is activated through various serine proteinases and biased agonists, it may couple TGF-β signaling to a diverse range of other physiological processes that may or may not predispose cells to cancer development such as local inflammation, systemic coagulation and pathogen infection.

    Topics: Animals; Cell Transformation, Neoplastic; Disease Progression; Humans; Neoplasms; Protein Binding; Protein Serine-Threonine Kinases; Receptor, PAR-2; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2017
Missing Links in Epithelial-Mesenchymal Transition: Long Non-Coding RNAs Enter the Arena.
    Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology, 2017, Volume: 44, Issue:4

    Cancer metastasis occurs through a series of sequential steps, which involves dissemination of tumor cells from a primary site and colonization in distant tissues. To promote the invasion-metastasis cascade, carcinoma cells usually initiate a cell-biological program called epithelial-mesenchymal transition (EMT), which is orchestrated by a set of master regulators, including TGF-β, Snail, ZEB and Twist families. The biological activities of these molecules are tightly regulated by a variety of cell-intrinsic pathways as well as extracellular cues. Recently, accumulating evidence indicates that long non-coding RNAs (lncRNAs) represent some of the most differentially expressed transcripts between primary and metastatic cancers. LncRNAs including MALAT1, HOTAIR, H19, LncRNA-ATB, and LincRNA-ROR have been reported to be involved in the process of EMT, mainly through cross-talking with master regulators of EMT. Thus, understanding the different and precise molecular mechanisms by which functional lncRNAs switch EMT on and off is important for opening up new avenues in lncRNA-directed diagnosis, prognosis, and therapeutic intervention against cancer.

    Topics: Epithelial-Mesenchymal Transition; Humans; Neoplasms; RNA, Long Noncoding; Snail Family Transcription Factors; Transforming Growth Factor beta; Twist-Related Protein 1; Zinc Finger E-box-Binding Homeobox 1

2017
The Potential of Frog Skin-Derived Peptides for Development into Therapeutically-Valuable Immunomodulatory Agents.
    Molecules (Basel, Switzerland), 2017, Dec-13, Volume: 22, Issue:12

    The aim of this article is to review the immunoregulatory actions of frog skin-derived peptides in order to assess their potential as candidates for immunomodulatory or anti-inflammatory therapy. Frog skin peptides with demonstrable immunomodulatory properties have been isolated from skin secretions of a range of species belonging to the families Alytidae, Ascaphidae, Discoglossidae, Leptodactylidae, Pipidae and Ranidae. Their effects upon production of inflammatory and immunoregulatory cytokines by target cells have been evaluated ex vivo and effects upon cytokine expression and immune cell activity have been studied in vivo by flow cytometry after injection into mice. The naturally-occurring peptides and/or their synthetic analogues show complex and variable actions on the production of proinflammatory (TNF-α, IL-1β, IL-12, IL-23, IL-8, IFN-γ and IL-17), pleiotropic (IL-4 and IL-6) and immunosuppressive (IL-10 and TGF-β) cytokines by peripheral and spleen cells, peritoneal cells and/or isolated macrophages. The effects of frenatin 2.1S include enhancement of the activation state and homing capacity of Th1-type lymphocytes and NK cells in the mouse peritoneal cavity, as well as the promotion of their tumoricidal capacities. Overall, the diverse effects of frog skin-derived peptides on the immune system indicate their potential for development into therapeutic agents.

    Topics: Amphibian Proteins; Animals; Anti-Inflammatory Agents; Anura; Gene Expression; Immunologic Factors; Inflammation; Interferon-gamma; Interleukins; Killer Cells, Natural; Mice; Neoplasms; Peptides; Skin; Th1 Cells; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2017
Complex Interplay Between Aging and Cancer: Role of TGF-β Signaling.
    Critical reviews in oncogenesis, 2017, Volume: 22, Issue:3-4

    Although cancer is known to be predominantly a disease of the elderly, it is also thought that aging and cancer may occur either via similar or opposing cellular mechanisms. Studies during the past decades were focused on understanding the molecular events underlying both processes, aiming to ultimately improve the quality of life and lifespan. However, these efforts were traditionally performed or viewed independently without considering the interplay between aging and cancer-promoting mechanisms. Transforming growth factor-beta (TGF-β) signaling is essential for embryonic development and physiological tissue homeostasis. Although the fundamental roles of TGF-β in cancer progression have been investigated extensively, recent reports also provide evidence for its direct or indirect links to aging-related processes such as cell proliferation, senescence, stem cell renewal, DNA damage, inflammation, and telomere length. In this review, we present the latest findings regarding the TGF-β-regulated convergent and divergent mechanisms controlling the balance between aging and cancer. Finally, the implications of these processes in developing strategies to prolong the cancer-free human lifespan are discussed.

    Topics: Aging; Animals; Cellular Senescence; DNA Damage; Humans; Neoplasms; Reactive Oxygen Species; Signal Transduction; Telomere Homeostasis; Transforming Growth Factor beta

2017
TGF-β signalling in tumour associated macrophages.
    Immunobiology, 2017, Volume: 222, Issue:1

    Tumour associated macrophages (TAM) represent an important component of tumour stroma. They develop under the influence of tumour microenvironment where transforming growth factor (TGF)β is frequently present. Activities of TAM regulated by TGFβ stimulate proliferation of tumour cells and lead to tumour immune escape. Despite high importance of TGFβ-induction of TAM activities till now our understanding of the mechanism of this induction is limited. We have previously developed a model of type 2 macrophages (M2) resembling certain properties of TAM. We established that in M2 TGFβRII is regulated on the level of subcellular sorting by glucocorticoids. Further studies revealed that in M2 with high levels of TGFβRII on the surface TGFβ activates not only its canonical Smad2/3-mediated signaling, but also Smad1/5-mediated signaling, what is rather typical for bone morphogenetic protein (BMP) stimulation. Complexity of macrophage populations, however, allows assumption that TGFβ signalling may function in different ways depending on the functional state of the cell. To understand the peculiarities of TGFβ signalling in human TAMs experimental systems using primary cells have to be developed and used together with the modern mathematical modelling approaches.

    Topics: Animals; Bone Morphogenetic Proteins; Gene Expression; Humans; Macrophages; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2017
FoxP3 Expression in Macrophages, Cancer, and B Cells-Is It Real?
    Clinical reviews in allergy & immunology, 2017, Volume: 52, Issue:3

    During the last decade, B regulatory cells are appreciated to have a central role in preventing autoimmunity and maintaining self-tolerance. They are characterized by expressing different phenotypic markers and the production of either IL-10 or TGF-β or both. The recent recognition of Fas ligand expressing B regulatory cells as "killer" cells established their role in maintaining viral persistence by preventing effective antiviral immune responses. The forkhead lineage-transcription factor (FoxP3) was considered for many years to be a highly specific intracellular regulatory marker of CD4+CD25+ T regulatory cells. The possibility of FoxP3 being expressed in B regulatory cells was suggested in many studies. Though controversial, FoxP3 expression was also reported in macrophages and cancer cells. Aiming to avoid artifact staining, many researchers required the usage of FoxP3 messenger RNA (mRNA) and PCR in order to prove a true expression of FoxP3 in these different cells. In addition, most studies' report on that FoxP3 expression in all abovementioned cells is related to their status of activation since naïve (non-activated cells) were found poorly FoxP3 expressing. In this review, we present the existing data on FoxP3 expression in non-T-regulatory cells, but we suggest that further studies are needed to better establish this concept.

    Topics: Animals; B-Lymphocytes, Regulatory; Biomarkers; Forkhead Transcription Factors; Humans; Interleukin-10; Lymphocyte Activation; Macrophages; Neoplasms; Self Tolerance; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2017
TGFΒ-induced transcription in cancer.
    Seminars in cancer biology, 2017, Volume: 42

    The Transforming Growth Factor-beta (TGFβ) pathway mediates a broad spectrum of cellular processes and is involved in several diseases, including cancer. TGFβ has a dual role in tumours, acting as a tumour suppressor in the early phase of tumorigenesis and as a tumour promoter in more advanced stages. In this review, we discuss the effects of TGFβ-driven transcription on all stages of tumour progression, with special focus on lung cancer. Since some TGFβ target genes are specifically involved in promoting metastasis, we speculate that these genes might be good targets to block tumour progression without compromising the tumour suppressor effects of the TGFβ pathway.

    Topics: Cell Transformation, Neoplastic; Genes, Tumor Suppressor; Humans; Neoplasm Staging; Neoplasms; Signal Transduction; Transcription, Genetic; Transforming Growth Factor beta

2017
Garp as a therapeutic target for modulation of T regulatory cell function.
    Expert opinion on therapeutic targets, 2017, Volume: 21, Issue:2

    Topics: Animals; Autoimmunity; Forkhead Transcription Factors; Humans; Immunotherapy; Inflammation; Integrins; Membrane Proteins; Mice; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Transforming Growth Factor beta1

2017
Oxidative stress in cancer and fibrosis: Opportunity for therapeutic intervention with antioxidant compounds, enzymes, and nanoparticles.
    Redox biology, 2017, Volume: 11

    Oxidative stress, mainly contributed by reactive oxygen species (ROS), has been implicated in pathogenesis of several diseases. We review two primary examples; fibrosis and cancer. In fibrosis, ROS promote activation and proliferation of fibroblasts and myofibroblasts, activating TGF-β pathway in an autocrine manner. In cancer, ROS account for its genomic instability, resistance to apoptosis, proliferation, and angiogenesis. Importantly, ROS trigger cancer cell invasion through invadopodia formation as well as extravasation into a distant metastasis site. Use of antioxidant supplements, enzymes, and inhibitors for ROS-generating NADPH oxidases (NOX) is a logical therapeutic intervention for fibrosis and cancer. We review such attempts, progress, and challenges. Lastly, we review how nanoparticles with inherent antioxidant activity can also be a promising therapeutic option, considering their additional feature as a delivery platform for drugs, genes, and imaging agents.

    Topics: Anti-Inflammatory Agents; Antineoplastic Agents; Antioxidants; Cell Proliferation; Fibroblasts; Fibrosis; Humans; Myofibroblasts; NADPH Oxidases; Nanoparticles; Neoplasm Invasiveness; Neoplasms; Neovascularization, Pathologic; Oxidative Stress; Reactive Oxygen Species; Transforming Growth Factor beta

2017
Molecular insights into tumour metastasis: tracing the dominant events.
    The Journal of pathology, 2017, Volume: 241, Issue:5

    Metastasis of malignant cells to vital organs remains the major cause of mortality in many types of cancers. The tumour invasion-metastasis cascade is a stepwise and multistage process whereby tumour cells disseminate from primary sites and spread to colonize distant sites through the systemic haematogenous or lymphatic circulations. The general steps of metastasis may be similar in almost all tumour types, but metastasis to different tissues seems to require distinct sets of regulators and/or an 'educated' microenvironment which may facilitate the infiltration and colonization of tumour cells to specific tissues. Moreover, interactions of tumour cells with stromal cells, endothelial cells, and immune cells that they encounter will also aid them to gain survival advantages, evade immune surveillance, and adapt to the new host microenvironment. Due to the high correlation between tumour metastasis and survival rate of patients, a deeper understanding of the molecular participants and processes involved in metastasis could pave the way towards novel, more effective and targeted approaches to prevent and treat tumour metastasis. In this review, we provide an update on the regulation networks orchestrated by the dominant regulators of different stages throughout the metastatic process including, but not limited to, epithelial-mesenchymal transition in local invasion, resistance to anoikis during migration, and colonization of different distant sites. We also put forward some suggestions and problems concerning the treatment of tumour metastasis that should be solved and/or improved for better therapies in the near future. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

    Topics: Anoikis; Cell Movement; Epithelial-Mesenchymal Transition; Humans; Models, Biological; Neoplasm Metastasis; Neoplasms; Receptors, Notch; Signal Transduction; Stromal Cells; Transforming Growth Factor beta; Wnt Signaling Pathway

2017
TGF-β, Bone Morphogenetic Protein, and Activin Signaling and the Tumor Microenvironment.
    Cold Spring Harbor perspectives in biology, 2017, May-01, Volume: 9, Issue:5

    The cellular and noncellular components surrounding the tumor cells influence many aspects of tumor progression. Transforming growth factor β (TGF-β), bone morphogenetic proteins (BMPs), and activins have been shown to regulate the phenotype and functions of the microenvironment and are attractive targets to attenuate protumorigenic microenvironmental changes. Given the pleiotropic nature of the cytokines involved, a full understanding of their effects on numerous cell types in many contexts is necessary for proper clinical intervention. In this review, we will explore the various effects of TGF-β, BMP, and activin signaling on stromal phenotypes known to associate with cancer progression. We will summarize these findings in the context of their tumor suppressive or promoting effects, as well as the molecular changes that these cytokines induce to influence stromal phenotypes.

    Topics: Activins; Animals; Bone Morphogenetic Proteins; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2017
Modulation of Tumour-Related Signaling Pathways by Natural Pentacyclic Triterpenoids and their Semisynthetic Derivatives.
    Current medicinal chemistry, 2017, Volume: 24, Issue:13

    Pentacyclic triterpenoids are a large class of natural isoprenoids that are widely biosynthesized in higher plants. These compounds are potent anticancer agents that exhibit antiproliferative, antiangiogenic, antiinflammatory and proapoptotic activities. Although their effects on multiple pathways have been reported, unifying mechanisms of action have not yet been established. To date, a huge number of semisynthetic derivatives have been synthesized in different laboratories on the basis of triterpenoid scaffolds, and many have been assayed for their biological activities. The present review focuses on natural triterpenoids of the oleanane-, ursane- and lupane-types and their semisynthetic derivatives. Here, we summarize the diverse cellular and molecular targets of these compounds and the signal pathways involved in the performance of their antitumour actions. Among the most relevant mechanisms involved are cell cycle arrest, apoptosis and autophagy triggered by the effect of triterpenoids on TGF-β and HER cell surface receptors and the downstream PI3KAkt- mTOR and IKK/NF-kB signaling axis, STAT3 pathway and MAPK cascades.

    Topics: Antineoplastic Agents; Apoptosis; Humans; Neoplasms; NF-kappa B; Oleanolic Acid; Signal Transduction; Transforming Growth Factor beta; Triterpenes

2017
Long noncoding RNA activated by TGF-β in human cancers: A meta-analysis.
    Clinica chimica acta; international journal of clinical chemistry, 2017, Volume: 468

    Because long non-coding RNA ATB (activated by TGF-β) is dysregulated in many cancers, we performed a meta-analysis to determine its prognostic potential in malignant tumors.. We searched electronic databases, including PubMed, Medline, OVID, Cochrane Library and Web of Science from inception until November 15, 2016 and identified eight studies with 818 cancer patients for the meta-analysis. We analyzed the hazard ratios (HRs) and 95% confidence intervals (CIs) to determine the relationship between lncRNA-ATB expression and overall survival (OS), recurrence -free survival (RFS), disease-free survival (DFS). We also use RevMan5.3 software to calculate odds ratio (ORs) to assess the association between lncRNA-ATB expression and pathological parameters including lymph node metastasis (LNM), distant metastasis (DM) and tumor stage.. Our analysis showed that increased lncRNA-ATB expression was associated with OS (HR=2.82, 95% CI:1.98-4.00, P<0.00001), DFS (HR=2.75, 95% CI:1.73-4.38, P<0.0001), RFS(HR=3.96, 95% CI:2.30-6.81, P<0.00001), LNM (OR=4.07, 95% CI 1.74-9.53, P=0.001), DM (OR=3.21, 95% CI 1.06-9.72, P=0.04) and high tumor stage (OR=2.81, 95% 1.78-4.43, P<0.0001) in patients with other types of cancers that excluded pancreatic cancer.. Meta-analysis demonstrated that increased lncRNA-ATB expression can be a useful prognostic biomarker in human cancer.

    Topics: Gene Expression Regulation, Neoplastic; Humans; Neoplasms; RNA, Long Noncoding; Transforming Growth Factor beta

2017
Cancer as a contagious disease.
    HLA, 2017, Volume: 89, Issue:4

    Contagious cancers are malignant cells that are physically transferred between individuals as a natural allograft, forming new clonal tumours. These cancers are highly unusual, but have emerged in 2 mammalian species, the dog and the Tasmanian devil, as well as 4 species of bivalve. The transfer of malignant cells in mammals should initiate a robust immune response and although invertebrates have a less complex immune system, these species still have mechanisms that should prevent engraftment and protect against cellular parasitism. Here the naturally occurring contagious cancers are reviewed to determine what features are important and necessary for the emergence and spread of these types of cancer, with a focus on the mammalian contagious cancers and how they successfully cross histocompatibility barriers.

    Topics: Animals; Bivalvia; Cell Proliferation; Disease Transmission, Infectious; Dog Diseases; Dogs; Gene Expression; Immunosuppression Therapy; Interferon-gamma; Killer Cells, Natural; Major Histocompatibility Complex; Marsupialia; Neoplasms; T-Lymphocytes; Transforming Growth Factor beta

2017
[Effects of growth differentiation factor-15 (GDF-15) on neurological systems, cardiovascular diseases, and cancer progression].
    Sheng li xue bao : [Acta physiologica Sinica], 2017, Feb-25, Volume: 69, Issue:1

    Growth differentiation factor-15 (GDF-15) is a member of the transforming growth factor beta superfamily. GDF-15 expression is dramatically upregulated during acute brain injury, cancer, cardiovascular disease, and inflammation, suggesting its potential value as a disease biomarker. It has been suggested that GDF-15 has neurotropic effects in the nervous system. Our studies showed that GDF-15 modulated the expression of neuronal K

    Topics: Animals; Brain Injuries; Calcium Channels; Cardiovascular Diseases; Disease Progression; Growth Differentiation Factor 15; Humans; Inflammation; Mice; Neoplasms; Nervous System; Potassium Channels; Prefrontal Cortex; Transforming Growth Factor beta; Up-Regulation

2017
Tumor growth factor-β is an important factor for immunosuppression and tumorgenesis in Polyoma BK virus infection; a systematic review article.
    Cytokine, 2017, Volume: 95

    Polyoma BK virus (PBK) is a prevalent human specific virus and the cause of several malignancies in human. The main mechanisms used by PBK to induce/stimulate human cancers are yet to be clarified but it has been proposed that PBK may use several mechanisms to induce/stimulate cancers in human including attenuation of immune responses via up-regulation of immunosuppressor molecules. Transforming growth factor beta (TGF-β) is a key multifunctional factor from modulation of immunosurveillance to angiogenesis. The key roles of TGF-β in the progression of Th17 and T regulatory subsets, the most important immune cells involved in development of cancers, have been demonstrated. Thus, this review article aims to describe the mechanisms used by PBK in induction/stimulation of human cancers in TGF-β dependent manner..

    Topics: BK Virus; Carcinogenesis; Humans; Immune Tolerance; Neoplasms; Polyomavirus Infections; Transforming Growth Factor beta; Tumor Virus Infections

2017
TGF-β Family Signaling in Tumor Suppression and Cancer Progression.
    Cold Spring Harbor perspectives in biology, 2017, Dec-01, Volume: 9, Issue:12

    Transforming growth factor-β (TGF-β) induces a pleiotropic pathway that is modulated by the cellular context and its integration with other signaling pathways. In cancer, the pleiotropic reaction to TGF-β leads to a diverse and varied set of gene responses that range from cytostatic and apoptotic tumor-suppressive ones in early stage tumors, to proliferative, invasive, angiogenic, and oncogenic ones in advanced cancer. Here, we review the knowledge accumulated about the molecular mechanisms involved in the dual response to TGF-β in cancer, and how tumor cells evolve to evade the tumor-suppressive responses of this signaling pathway and then hijack the signal, converting it into an oncogenic factor. Only through the detailed study of this complexity can the suitability of the TGF-β pathway as a therapeutic target against cancer be evaluated.

    Topics: Animals; Apoptosis; Cell Differentiation; Cell Movement; Disease Progression; Epigenesis, Genetic; Humans; Killer Cells, Natural; Macrophages; Mice; Mutation; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Phosphorylation; Signal Transduction; T-Lymphocytes; Transforming Growth Factor beta; Tumor Microenvironment

2017
MicroRNAs, TGF-β signaling, and the inflammatory microenvironment in cancer.
    Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine, 2016, Volume: 37, Issue:1

    Inflammatory cells and mediators form a major part of the tumor microenvironment and play important roles in the regulation of cancer initiation, tumor cell proliferation, and metastasis. MicroRNAs (miRNAs) play important roles in several physiological and pathological processes, including the regulation of the inflammatory microenvironment in cancer. Transforming growth factor-β (TGF-β) is an inflammation-related cytokine that functions in both tumor suppression and promotion; however, its underlying molecular mechanisms remain unclear. Recent evidence indicates an association between miRNAs and TGF-β signaling, providing new insight into the nature of the inflammatory microenvironment in cancer. The present review is an overview of the interaction between miRNAs and inflammatory cytokines, with emphasis on the cross talk between TGF-β signaling and miRNAs and their influence on cancer cell behavior. The emerging roles of miRNAs in cancer-related inflammation and the potential to target miRNA signaling pathways for cancer therapy are also discussed.

    Topics: Animals; Cytokines; Gene Expression Regulation, Neoplastic; Humans; Inflammation; Mice; MicroRNAs; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Microenvironment; Up-Regulation

2016
Crosstalk between transforming growth factor-β signaling pathway and long non-coding RNAs in cancer.
    Cancer letters, 2016, Jan-28, Volume: 370, Issue:2

    The transforming growth factor-β (TGF-β) signaling pathway plays an important role in tumorigenesis by exerting either a tumor-suppressing or tumor-promoting effect. Long non-coding RNAs (lncRNAs), a newly discovered class of non-coding RNAs, have been widely studied in recent years and identified as crucial regulators of various biological processes, including cell cycle progression, chromatin remodeling, gene transcription, and posttranscriptional processing. Recent evidence, addressing the crosstalk between the TGF-β signaling pathway and lncRNAs in cancer, found that several members of the TGF-β pathway are targeted by lncRNAs, and the production of hundreds of lncRNAs is induced by TGF-β treatment. This review will summarize the latest progress on the investigation of TGF-β pathway and lncRNA network in regulating cancer development. Further study on the network would provide a better understanding of carcinogenesis and have potentials for the prevention and treatment of malignant diseases.

    Topics: Humans; Neoplasms; RNA, Long Noncoding; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2016
The emerging role of RUNX3 in cancer metastasis (Review).
    Oncology reports, 2016, Volume: 35, Issue:3

    Metastasis remains the major driver of mortality in patients with cancer. The multistep metastatic process starts with the dissemination of tumor cells from a primary site and leading to secondary tumor development in an anatomically distant location. Although significant progress has been made in understanding the molecular characteristics of metastasis, many questions remain regarding the intracellular mechanisms governing transition through the various metastatic stages. The runt-related transcription factor 3 (RUNX3) is a downstream effector of the transforming growth factor-β (TGF-β) signaling pathway, and has critical roles in the regulation of cell death by apoptosis, and in angiogenesis, epithelial-to-mesenchymal transition (EMT), cell migration and invasion. RUNX3 functions as a bona fide initiator of carcinogenesis by linking the Wnt oncogenic and TGF-β tumor suppressive pathways. RUNX3 is frequently inactivated in human cancer cell lines and cancer samples by hemizygous deletion of the Runx3 gene, hypermethylation of the Runx3 promoter, or cytoplasmic sequestration of RUNX3 protein. Inactivation of RUNX3 makes it a putative tumor suppressor in human neoplasia. In the present review, we summarize the proposed roles of RUNX3 in metastasis and, when applicable, highlight the mechanism by which they function.

    Topics: Apoptosis; Carcinogenesis; Cell Movement; Core Binding Factor Alpha 3 Subunit; Epithelial-Mesenchymal Transition; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2016
Cross-talk between platelet and tumor microenvironment: Role of multiligand/RAGE axis in platelet activation.
    Blood reviews, 2016, Volume: 30, Issue:3

    Topics: Amyloid beta-Peptides; Biomarkers, Tumor; Blood Platelets; Cell Communication; HMGB1 Protein; Humans; Leukocyte L1 Antigen Complex; Ligands; Neoplasms; Paracrine Communication; Platelet Activation; Receptor for Advanced Glycation End Products; Transforming Growth Factor beta; Tumor Microenvironment

2016
Cancer Immunotherapy: Selected Targets and Small-Molecule Modulators.
    ChemMedChem, 2016, Mar-04, Volume: 11, Issue:5

    There is a significant amount of excitement in the scientific community around cancer immunotherapy, as this approach has renewed hope for many cancer patients owing to some recent successes in the clinic. Currently available immuno-oncology therapeutics under clinical development and on the market are mostly biologics (antibodies, proteins, engineered cells, and oncolytic viruses). However, modulation of the immune system with small molecules offers several advantages that may be complementary and potentially synergistic to the use of large biologicals. Therefore, the discovery and development of novel small-molecule modulators is a rapidly growing research area for medicinal chemists working in cancer immunotherapy. This review provides a brief introduction into recent trends related to selected targets and pathways for cancer immunotherapy and their small-molecule pharmacological modulators.

    Topics: Adenosine; Humans; Immunotherapy; Kynurenine; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Toll-Like Receptors; Transforming Growth Factor beta

2016
Six family of homeobox genes and related mechanisms in tumorigenesis protocols.
    Tumori, 2016, Jun-02, Volume: 2016, Issue:3

    In recent years, the homeobox gene superfamily has been introduced as a master regulator in downstream target genes related to cell development and proliferation. An indispensable role of this family involved in organogenesis development has been widely demonstrated since expression of Six family led to a distinct increase in development of various organs. These functions of Six family genes are primarily based on structure as well as regulatory role in response to external or internal stimuli. In addition to these roles, mutation or aberrant expression of Six family plays a fundamental role in initiation of carcinogenesis, a multistep process including transformation, proliferation, angiogenesis, migration, and metastasis. This suggests that the Six superfamily members can be considered as novel target molecules to inhibit tumor growth and progression. This review focuses on the structure, function, and mechanisms of the Six family in cancer processes and possible strategies to apply these family members for diagnostic, prognostic, and therapeutic purposes.

    Topics: Animals; Carcinogenesis; Cell Cycle; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Cyclin D; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Genes, Homeobox; Homeodomain Proteins; Humans; MicroRNAs; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Nerve Tissue Proteins; Prognosis; Signal Transduction; Smad Proteins; Trans-Activators; Transforming Growth Factor beta; Up-Regulation

2016
miR-200c: a versatile watchdog in cancer progression, EMT, and drug resistance.
    Journal of molecular medicine (Berlin, Germany), 2016, Volume: 94, Issue:6

    MicroRNAs (miRNAs) are 20-22-nucleotide small endogenous non-coding RNAs which regulate gene expression at post-transcriptional level. In the last two decades, identification of almost 2600 miRNAs in human and their potential to be modulated opened a new avenue to target almost all hallmarks of cancer. miRNAs have been classified as tumor suppressors or oncogenes depending on the phenotype they induce, the targets they modulate, and the tissue where they function. miR-200c, an illustrious tumor suppressor, is one of the highly studied miRNAs in terms of development, stemness, proliferation, epithelial-mesenchymal transition (EMT), therapy resistance, and metastasis. In this review, we first focus on the regulation of miR-200c expression and its role in regulating EMT in a ZEB1/E-cadherin axis-dependent and ZEB1/E-cadherin axis-independent manner. We then describe the role of miR-200c in therapy resistance in terms of multidrug resistance, chemoresistance, targeted therapy resistance, and radiotherapy resistance in various cancer types. We highlight the importance of miR-200c at the intersection of EMT and chemoresistance. Furthermore, we show how miR-200c coordinates several important signaling cascades such as TGF-β signaling, PI3K/Akt signaling, Notch signaling, VEGF signaling, and NF-κB signaling. Finally, we discuss miR-200c as a potential prognostic/diagnostic biomarker in several diseases, but mainly focusing on cancer and its potential application in future therapeutics.

    Topics: Animals; Antigens, CD; Antineoplastic Agents; Cadherins; Cell Transformation, Neoplastic; Disease Progression; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Zinc Finger E-box Binding Homeobox 2; Zinc Finger E-box-Binding Homeobox 1

2016
GARP: a surface molecule of regulatory T cells that is involved in the regulatory function and TGF-β releasing.
    Oncotarget, 2016, Jul-05, Volume: 7, Issue:27

    There are many molecules that define regulatory T cells (Tregs) phenotypically and functionally. Glycoprotein A repetitions predominant (GARP) is a transmembrane protein containing leucine rich repeats. Recently, GARP is found to express highly on the surface of activated Tregs. The combination of GARP and other surface molecules isolates Tregs with higher purity. Besides, GARP is a cell surface molecule of Tregs that maintains their regulatory function and homeosatsis. GARP has also been proved to promote the activation and secretion of transforming growth factor β (TGF-β). Moreover, its potential value in cancer immunotherapy is also discussed in this work.

    Topics: Animals; CD4-Positive T-Lymphocytes; Cell Membrane; Forkhead Transcription Factors; Gene Expression Profiling; Gene Expression Regulation; Glycoproteins; Homeostasis; Humans; Immunotherapy; Lymphocyte Activation; Membrane Proteins; Mice; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Transforming Growth Factor beta1

2016
State-of-the-art of regulatory dendritic cells in cancer.
    Pharmacology & therapeutics, 2016, Volume: 164

    Dendritic cells (DCs) with robust immunosuppressive activity are commonly found in the microenvironment of advanced solid tumors. These innate immune cells are generically termed regulatory DCs and include various subsets such as plasmacytoid, conventional and monocyte-derived/inflammatory populations whose normal function is subverted by tumor-derived signals. This review summarizes recent findings on the nature and function of regulatory DCs, their relationship with other myeloid subsets and unique therapeutic opportunities to abrogate malignant progression through their targeting.

    Topics: Animals; Dendritic Cells; Dinoprostone; Humans; Immune Tolerance; Macrophages; Matrix Attachment Region Binding Proteins; Myelopoiesis; Nanostructures; Neoplasms; Transforming Growth Factor beta

2016
TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology.
    Cold Spring Harbor perspectives in biology, 2016, May-02, Volume: 8, Issue:5

    The transforming growth factor-β (TGF-β) is the prototype of the TGF-β family of growth and differentiation factors, which is encoded by 33 genes in mammals and comprises homo- and heterodimers. This review introduces the reader to the TGF-β family with its complexity of names and biological activities. It also introduces TGF-β as the best-studied factor among the TGF-β family proteins, with its diversity of roles in the control of cell proliferation and differentiation, wound healing and immune system, and its key roles in pathology, for example, skeletal diseases, fibrosis, and cancer.

    Topics: Animals; Cell Differentiation; Cell Proliferation; Disease Progression; Fibrosis; Gene Expression Regulation; Humans; Immune System; Mice; Neoplasms; Phylogeny; Rats; Signal Transduction; Terminology as Topic; Transforming Growth Factor beta; Wound Healing

2016
Regulation of the TGF-β pathway by deubiquitinases in cancer.
    The international journal of biochemistry & cell biology, 2016, Volume: 76

    The transforming growth factor-β (TGF-β) pathway regulates diverse cellular processes. It signals via serine/threonine kinase receptors and intracellular Smad and non-Smad effector proteins. In cancer cells, aberrant TGF-β signalling can lead to loss of growth inhibition and an increase in invasion, epithelial-to-mesenchymal transition (EMT) and metastasis. Therapeutic targeting of the pro-oncogenic TGF-β responses is currently being explored as a potential therapy against certain invasive and metastatic cancer types. The ubiquitin post-translational regulation system is emerging as a key regulatory mechanism for the control of TGF-β pathway components. In this review, we focus on the role of deubiquitinases (DUBs), which counteract the activity of E3 ubiquitin ligases. We will discuss the mechanisms by which specific DUBs control Smad and non-Smad TGF-β signalling routes, and how perturbation of the expression and function of DUBs contributes to misregulation of TGF-β signalling in cancer.

    Topics: Animals; Deubiquitinating Enzymes; Epithelial-Mesenchymal Transition; Humans; Neoplasm Proteins; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2016
Bone Morphogenetic Proteins.
    Cold Spring Harbor perspectives in biology, 2016, Jun-01, Volume: 8, Issue:6

    Bone morphogenetic proteins (BMPs), originally identified as osteoinductive components in extracts derived from bone, are now known to play important roles in a wide array of processes during formation and maintenance of various organs including bone, cartilage, muscle, kidney, and blood vessels. BMPs and the related "growth and differentiation factors" (GDFs) are members of the transforming growth factor β (TGF-β) family, and transduce their signals through type I and type II serine-threonine kinase receptors and their intracellular downstream effectors, including Smad proteins. Furthermore, BMP signals are finely tuned by various agonists and antagonists. Because deregulation of the BMP activity at multiple steps in signal transduction is linked to a wide variety of human diseases, therapeutic use of activators and inhibitors of BMP signaling will provide potential avenues for the treatment of the human disorders that are caused by hypo- and hyperactivation of BMP signals, respectively.

    Topics: Animals; Bone and Bones; Bone Morphogenetic Proteins; Cell Differentiation; DNA-Binding Proteins; Gene Expression Regulation; Humans; Neoplasms; Protein Serine-Threonine Kinases; Signal Transduction; Smad Proteins; Trans-Activators; Transforming Growth Factor beta

2016
The role of integrins in TGFβ activation in the tumour stroma.
    Cell and tissue research, 2016, Volume: 365, Issue:3

    TGFβ1 is the most pleiotropic of all known cytokines and thus, to avoid uncontrolled TGFβ-activated processes, its activity is tightly regulated. Studies in fibrosis have led to the discovery that αv integrins are the major regulators of the local activation of latent TGFβ in our tissues. Since all cells can express one or more types of αv integrins, this raises the possibility that, in the complex milieu of a developing cancer, multiple cell types including both cancer cells and stromal cells activate TGFβ. In normal tissues, TGFβ1 is a tumour suppressor through its ability to suppress epithelial cell division, whereas in cancer, in which tumour cells develop genetic escape mechanisms to become resistant to TGFβ growth suppression, TGFβ signalling creates a tumour-permissive environment by activating fibroblast-to-myofibroblast transition, by promoting angiogenesis, by suppressing immune cell populations and by promoting the secretion of both matrix proteins and proteases. In addition, TGFβ drives epithelial-to-mesenchymal transition (EMT) increasing the potential for metastasis. Since αv integrins activate TGFβ, they almost certainly drive TGFβ-dependent cancer progression. In this review, we discuss the data that are helping to develop this hypothesis and describe the evidence that αv integrins regulate the TGFβ promotion of cancer. Graphical Abstract Mechanisms of integrin-mediated transforming growth factor beta (TGFβ) activation and its effect on stromal processes. 1 Matrix-bound latent LAP-TGFβ1 binds αv integrins expressed by epithelial cells or fibroblasts (LAP latency-associated peptide). TGFβ1 becomes exposed. 2 Active TGFβ1 binds the TGFβ receptor in an autocrine or paracrine fashion. 3 TGFβ1 signalling increases integrin expression, LAP-TGFβ1 secretion and trans-differentiation of fibroblasts into contractile cells that secrete collagens and collagen cross-linking proteins. By contracting the matrix, latent TGFβ1 is stretched making the activation of latent TGFβ1 easier and creating a continuous cycle of TGFβ1 signalling. TGFβ1 promotes cancer progression by promoting angiogenesis, immune suppression and epithelial-to-mesenchymal transition (EMT).

    Topics: Animals; Drug Delivery Systems; Humans; Integrins; Neoplasms; Stromal Cells; Transforming Growth Factor beta; Tumor Microenvironment

2016
Tension in Cancer.
    International journal of molecular sciences, 2016, Nov-16, Volume: 17, Issue:11

    Integrins represent a large family of cell receptors that mediate adhesion to the extracellular matrix (ECM), thereby modulating a variety of cellular functions that are required for proliferation, migration, malignant conversion and invasiveness. During tumorigenesis the conversion of a tumor cell from sessile, stationary phenotype to an invasive phenotype requires the ability of tumor cells to interact with their environment in order to transduce signals from the ECM into the cells. Hence, there is increasing evidence that changes in the composition, topography and tension of tumor matrix can be sensed by integrin receptors, leading to the regulation of intracellular signalling events which subsequently help to fuel cancer progression. The fact that intracellular signals perceived from integrin ligand binding impact on almost all steps of tumor progression, including tumor cell proliferation, survival, metastatic dissemination and colonization of a metastatic niche, renders integrins as ideal candidates for the development of therapeutic agents. In this review we summarize the role of integrins in cancer with the special focus on cancer therapies and the recent progress that has been made in the understanding of "integrin-induced tension in cancer". Finally, we conclude with clinical evidence for the role of integrin-mediated mechanotransduction in the development of therapy-resistant tumors.

    Topics: Animals; Cell Adhesion; Cell Transformation, Neoplastic; Drug Resistance, Neoplasm; Elasticity; Extracellular Matrix; Humans; Integrins; Mechanotransduction, Cellular; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2016
Immunoregulation by members of the TGFβ superfamily.
    Nature reviews. Immunology, 2016, 11-25, Volume: 16, Issue:12

    The transforming growth factor-β (TGFβ) superfamily is encoded by 33 genes and includes TGFβ, bone morphogenetic proteins (BMPs) and activins. Although TGFβ is well recognized as a crucial regulator of immune responses, the immunoregulatory functions of other TGFβ family members are less clear. However, recent evidence suggests that BMPs and activins have important roles in regulating immune responses. In this Review, we briefly outline the signalling pathways of the TGFβ superfamily and discuss new insights into the immunoregulatory functions of BMPs and activins in the context of infection, inflammation and cancer.

    Topics: Activins; Animals; Bone Morphogenetic Proteins; Gene Expression Regulation; Homeostasis; Humans; Immune System; Immunomodulation; Inflammation; Multigene Family; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2016
Dichotomous roles of TGF-β in human cancer.
    Biochemical Society transactions, 2016, 10-15, Volume: 44, Issue:5

    Transforming growth factor-β (TGF-β) mediates numerous biological processes, including embryonic development and the maintenance of cellular homeostasis in a context-dependent manner. Consistent with its central role in maintaining cellular homeostasis, inhibition of TGF-β signaling results in disruption of normal homeostatic processes and subsequent carcinogenesis, defining the TGF-β signaling pathway as a tumor suppressor. However, once carcinogenesis is initiated, the TGF-β signaling pathway promotes cancer progression. This dichotomous function of the TGF-β signaling pathway is mediated through altering effects on both the cancer cells, by inducing apoptosis and inhibiting proliferation, and the tumor microenvironment, by promoting angiogenesis and inhibiting immunosurveillance. Current studies support inhibition of TGF-β signaling either alone, or in conjunction with anti-angiogenic therapy or immunotherapy as a promising strategy for the treatment of human cancers.

    Topics: Apoptosis; Cell Proliferation; Disease Progression; Epithelial-Mesenchymal Transition; Homeostasis; Humans; Models, Biological; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2016
FRA-1 as a driver of tumour heterogeneity: a nexus between oncogenes and embryonic signalling pathways in cancer.
    Oncogene, 2015, Aug-20, Volume: 34, Issue:34

    Tumour heterogeneity is a major factor undermining the success of therapies targeting metastatic cancer. Two major theories are thought to explain the phenomenon of heterogeneity in cancer--clonal evolution and cell plasticity. In this review, we examine a growing body of work implicating the transcription factor FOS-related antigen 1 (FRA-1) as a central node in tumour cell plasticity networks, and discuss mechanisms regulating its activity in cancer cells. We also discuss evidence from the FRA-1 perspective supporting the notion that clonal selection and cell plasticity represent two sides of the same coin. We propose that FRA-1-overexpressing clones featuring high plasticity undergo positive selection during consecutive stages of multistep tumour progression. This model underscores a potential mechanism through which tumour cells retaining elevated levels of plasticity acquire a selective advantage over other clonal populations within a tumour.

    Topics: Animals; Clonal Evolution; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Oncogenes; Proto-Oncogene Proteins c-fos; Signal Transduction; Transforming Growth Factor beta; Wnt Signaling Pathway

2015
Generation, subsets and functions of inducible regulatory T cells.
    Anti-inflammatory & anti-allergy agents in medicinal chemistry, 2015, Volume: 13, Issue:3

    The idea of regulatory T cells (Tregs) lost its popularity during the 1980s and 1990s, since immunologists failed to elucidate how the innate regulation of immunological reactions worked. The entire re-evaluation of the Tregs was supported due to the increasingly influential and state of the art immunological techniques, as cell sorting and also the expanding understanding of the immune system and its functions which aided in attaining a greater insight into the mechanisms of regulation and suppression. Many researchers nowadays have demonstrated that Tregs may well have therapeutic possibilities for the treatment of autoimmune diseases if it was a possibility to isolate and infuse these Tregs into patients, preferably without any harmful side effects. Therefore, modulation of Tregs as well as their generation is being researched since they were proposed as therapeutic interventions in several disease sceneries, nonetheless, sometimes with disastrous consequences. Consequently, a full and complete understanding of the exceptional biology of human Tregs is fundamental for the accurate interpretation of found data, before therapeutic interventions can be undertaken. This literature study gives an overview of the current accessible information on the topic of the characterization, the generation, regulation and functions of the Inducible Treg populations and their subsets in the human immune system.

    Topics: Animals; Autoimmune Diseases; Cytokines; Forkhead Transcription Factors; Humans; Hypersensitivity; Immunity, Innate; Immunity, Mucosal; Immunologic Factors; Immunotherapy; Neoplasms; Phenotype; Signal Transduction; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2015
Targeting the TGFβ pathway for cancer therapy.
    Pharmacology & therapeutics, 2015, Volume: 147

    The TGFβ signaling pathway has pleiotropic functions regulating cell growth, differentiation, apoptosis, motility and invasion, extracellular matrix production, angiogenesis, and immune response. TGFβ signaling deregulation is frequent in tumors and has crucial roles in tumor initiation, development and metastasis. TGFβ signaling inhibition is an emerging strategy for cancer therapy. The role of the TGFβ pathway as a tumor-promoter or suppressor at the cancer cell level is still a matter of debate, due to its differential effects at the early and late stages of carcinogenesis. In contrast, at the microenvironment level, the TGFβ pathway contributes to generate a favorable microenvironment for tumor growth and metastasis throughout all the steps of carcinogenesis. Then, targeting the TGFβ pathway in cancer may be considered primarily as a microenvironment-targeted strategy. In this review, we focus on the TGFβ pathway as a target for cancer therapy. In the first part, we provide a comprehensive overview of the roles played by this pathway and its deregulation in cancer, at the cancer cell and microenvironment levels. We go on to describe the preclinical and clinical results of pharmacological strategies to target the TGFβ pathway, with a highlight on the effects on tumor microenvironment. We then explore the perspectives to optimize TGFβ inhibition therapy in different tumor settings.

    Topics: Animals; Antineoplastic Agents; Clinical Trials as Topic; Drug Delivery Systems; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2015
Role of TGFβ in regulation of the tumor microenvironment and drug delivery (review).
    International journal of oncology, 2015, Volume: 46, Issue:3

    Deregulation of cell signaling homeostasis is a predominant feature of cancer initiation and progression. Transforming growth factor β (TGFβ) is a pleiotropic cytokine, which regulates numerous biological processes of various tissues in an autocrine and paracrine manner. Aberrant activity of TGFβ signaling is well known to play dual roles in cancer, depending on tumor stage and cellular context. The crucial roles of TGFβ in modulating the tumor microenvironment, its contribution to the accumulation of mechanical forces within the solid constituents of a tumor and its effects on the effective delivery of drugs are also becoming increasingly clear. In this review, we discuss the latest advances in the efforts to unravel the effects of TGFβ signaling in various components of the tumor microenvironment and how these influence the generation of forces and the efficacy of drugs. We also report the implications of tumor mechanics in cancer therapy and the potential usage of anti‑TGFβ agents to enhance drug delivery and augment existing therapeutic approaches. These findings provide new insights towards the significance of targeting TGFβ pathway to enhance personalized tumor treatment.

    Topics: Antineoplastic Agents; Drug Repositioning; Humans; Molecular Targeted Therapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2015
Epithelial-mesenchymal transition is regulated at post-transcriptional levels by transforming growth factor-β signaling during tumor progression.
    Cancer science, 2015, Volume: 106, Issue:5

    Transforming growth factor (TGF)-β acts as a tumor suppressor during cancer initiation, but as a tumor promoter during tumor progression. It has become increasingly clear that TGF-β plays fundamental roles in multiple steps of tumor progression, including epithelial-mesenchymal transition (EMT). The EMT, first described by developmental biologists at the beginning of the 1980s, plays crucial roles in appropriate embryonic development, but also functions in adults during wound healing, organ fibrosis, and tumor progression. During EMT, epithelial cells lose their epithelial polarity and acquire mesenchymal phenotypes, endowing them with migratory and invasive properties. Many secreted polypeptides are implicated in this process, and act in a sequential or cooperative manner. TGF-β induces EMT by propagating intracellular signaling pathways and activating transcriptional factors. Here, I discuss new insights into the molecular mechanisms underlying induction of EMT by TGF-β in cooperation with Ras or growth factors, along with the signals that induce EMT through transcriptional and post-transcriptional regulation.

    Topics: Animals; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Neoplasms; Protein Processing, Post-Translational; ras Proteins; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2015
Crosstalk of carcinoembryonic antigen and transforming growth factor-β via their receptors: comparing human and canine cancer.
    Cancer immunology, immunotherapy : CII, 2015, Volume: 64, Issue:5

    There is accumulating evidence that the transforming growth factor beta (TGF-β) and nuclear factor kappa-B (NFκB) pathways are tightly connected and play a key role in malignant transformation in cancer. Immune infiltration by regulatory T- and B-lymphocytes (Tregs, Bregs) has recently gained increased attention for being an important source of TGF-β. There is a plethora of studies examining the pro-tumorigenic functions of carcinoembryonic antigen (CEA), but its receptor CEAR is far less studied. So far, there is a single connecting report that TGF-β also may signal through CEAR. The crosstalk between cancer tissues is further complicated by the expression of CEAR and TGF-β receptors in stromal cells, and implications of TGF-β in epithelial-mesenchymal transition. Furthermore, tumor-infiltrating Tregs and Bregs may directly instruct cancer cells by secreting TGF-β binding to their CEAR. Therefore, both TGF-β and CEA may act synergistically in breast cancer and cause disease progression, and NFκB could be a common crossing point between their signaling. CEAR, TGF-β1-3, TGF-β-R types I-III and NFκB class I and II molecules have an outstanding human-canine sequence identity, and only a canine CEA homolog has not yet been identified. For these reasons, the dog may be a valid translational model patient for investigating the crosstalk of the interconnected CEA and TGF-β networks.

    Topics: Animals; B-Lymphocytes, Regulatory; Carcinoembryonic Antigen; Dog Diseases; Dogs; Epithelial-Mesenchymal Transition; Humans; I-kappa B Kinase; Neoplasms; NF-kappa B; Protein Binding; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2015
Cancer stem cells: the promise and the potential.
    Seminars in oncology, 2015, Volume: 42 Suppl 1

    Despite the advancement of treatment modalities, many cancer patients experience tumor recurrence and metastasis at regional or distant sites. Evolving understanding of tumor biology has led to the hypothesis that tumors may possess a stem cell-like subpopulation known as cancer stem cells (CSCs) that may be involved in driving tumor propagation and pathogenesis. Like normal stem cells (NSCs), CSCs can be identified by markers such as CD133, CD44, and ALDH. CSCs have the ability to self-renew and differentiate into different tumor components through stemness pathways, such as Wnt, TGF-β, STAT, and Hippo-YAP/TAZ, among others. In NSCs, stemness pathways are strictly regulated and control many important biologic processes, including embryogenesis and intestinal crypt cellular regulation. In contrast, stemness pathways in CSCs are significantly dysregulated. Combining current drugs with the targeting of these stemness pathways may significantly improve patient prognosis. The aim of this supplement is to update clinicians on the accumulated evidence characterizing the role of CSCs in tumor initiation, heterogeneity, therapy resistance, and recurrence and metastasis, and the potential for effectively treating patients.

    Topics: Biomarkers, Tumor; Breast Neoplasms; Drug Resistance, Neoplasm; Female; Gastrointestinal Neoplasms; Hematologic Neoplasms; Humans; Hyaluronan Receptors; Molecular Targeted Therapy; Neoplasm Recurrence, Local; Neoplasms; Neoplastic Stem Cells; Prognosis; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment; Wnt Signaling Pathway

2015
Radiation-induced fibrosis: mechanisms and implications for therapy.
    Journal of cancer research and clinical oncology, 2015, Volume: 141, Issue:11

    Radiation-induced fibrosis (RIF) is a long-term side effect of external beam radiation therapy for the treatment of cancer. It results in a multitude of symptoms that significantly impact quality of life. Understanding the mechanisms of RIF-induced changes is essential to developing effective strategies to prevent long-term disability and discomfort following radiation therapy. In this review, we describe the current understanding of the etiology, clinical presentation, pathogenesis, treatment, and directions of future therapy for this condition.. A literature review of publications describing mechanisms or treatments of RIF was performed. Specific databases utilized included PubMed and clinicaltrials.gov, using keywords "Radiation-Induced Fibrosis," "Radiotherapy Complications," "Fibrosis Therapy," and other closely related terms.. RIF is the result of a misguided wound healing response. In addition to causing direct DNA damage, ionizing radiation generates reactive oxygen and nitrogen species that lead to localized inflammation. This inflammatory process ultimately evolves into a fibrotic one characterized by increased collagen deposition, poor vascularity, and scarring. Tumor growth factor beta serves as the primary mediator in this response along with a host of other cytokines and growth factors. Current therapies have largely been directed toward these molecular targets and their associated signaling pathways.. Although RIF is widely prevalent among patients undergoing radiation therapy and significantly impacts quality of life, there is still much to learn about its pathogenesis and mechanisms. Current treatments have stemmed from this understanding, and it is anticipated that further elucidation will be essential for the development of more effective therapies.

    Topics: DNA Damage; Fibrosis; Humans; Inflammation; Neoplasms; Radiation Injuries; Radiotherapy; Transforming Growth Factor beta

2015
[Research progress on epithelial-mesenchymal transition in cancer recurrence and metastasis].
    Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences, 2015, Volume: 44, Issue:2

    Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells lose their morphology and function and gradually transformed into mesenchymal-like cells. It is considered that EMT is the main cause for tumor recurrence and metastasis. Many factors are involved in the regulation of EMT, such as E-cadherin, transforming growth factor-β, Wnt signaling pathway, microRNA and EMT-related transcription factors. This article reviews the research progress on EMT and the involved mechanisms, and thus to provide a new perspective on cancer therapy in the future.

    Topics: Cadherins; Epithelial-Mesenchymal Transition; Humans; MicroRNAs; Neoplasm Metastasis; Neoplasm Recurrence, Local; Neoplasms; Signal Transduction; Transcription Factors; Transforming Growth Factor beta; Wnt Signaling Pathway

2015
Re-evaluating the role of activin-βC in cancer biology.
    Cytokine & growth factor reviews, 2015, Volume: 26, Issue:4

    Transforming growth factor-β (TGF-β) superfamily signaling pathway and its ligands are essential regulators of cellular processes such as proliferation, differentiation, migration, and survival. Alteration of this pathway results in uncontrolled proliferation and cancer progression. This review focuses on a specific member of the TGF-β superfamily: activin-βC. After its initial discovery, activin-βC has been considered non-biologically relevant. Therefore, for years several experimental designs have ignored the potential contribution of this molecule to the final biological outcome. Here we focus on recent advances in the activin field, with a particular emphasis on activin-βC, its antagonistic mechanism, and the physiological relevance of activin-βC actions in reproductive and cancer biology. Covering a novel and previously unexplored function of activin-βC on cancer associated weight loss and muscle metabolism, this review suggests an imminent need to re-evaluate the function of activin-βC in biological systems and advances the understanding of how activin-βC antagonizes the activin signaling pathway. Thus, challenging activin biologists to consider the impact of activin-βC when interpreting their work.

    Topics: Animals; Cell Differentiation; Humans; Inhibin-beta Subunits; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2015
Association between Int7G24A rs334354 polymorphism and cancer risk: a meta-analysis of case-control studies.
    Scientific reports, 2015, Jun-15, Volume: 5

    Accumulating evidences have suggested the potential association between Int7G24A (rs334354) polymorphism and cancer risk. However, results from epidemiological studies are controversial. We thus conducted this meta-analysis to clarify the association. Relevant studies were identified on electronic databases according to the inclusion criteria. A total of 13 case-control studies containing 4092 cases and 5909 controls were included in our meta-analysis. Odds ratios (ORs) with 95% confidence intervals (CIs) were applied to assess the association. The results of the overall population had suggested that Int7G24A polymorphism had an increased risk for cancer, reaching significant levels in the 2 genetic models (allele model, OR = 1.25, 95% CI 1.09-1.42, P = 0.001; dominant model, OR = 1.24, 95% CI 1.06-1.46, P < 0.008). Besides, significant association was found among Asian population (allele model, OR = 1.27, 95% CI 1.11-1.45, P < 0.001; dominant model, OR = 1.28, 95% CI 1.11-1.49, P < 0.001), whereas there was non-significant relationship detected among Caucasian population (allele model, OR = 1.08, 95% CI 0.92-1.26, P = 0.352; dominant model, OR = 1.05, 95% CI 0.87-1.26, P = 0.639). The present meta-analysis had suggested that Int7G24A polymorphism of gene TGFBR1 involved in the transforming growth factor beta (TGF-β) signaling pathway had a significantly increased risk for cancer development.

    Topics: Case-Control Studies; Gene Expression; Genetic Association Studies; Genetic Predisposition to Disease; Humans; Introns; Models, Genetic; Mutation; Neoplasms; Polymorphism, Genetic; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Risk; Signal Transduction; Transforming Growth Factor beta

2015
Transforming Growth Factor-Beta and Oxidative Stress Interplay: Implications in Tumorigenesis and Cancer Progression.
    Oxidative medicine and cellular longevity, 2015, Volume: 2015

    Transforming growth factor-beta (TGF-β) and oxidative stress/Reactive Oxygen Species (ROS) both have pivotal roles in health and disease. In this review we are analyzing the interplay between TGF-β and ROS in tumorigenesis and cancer progression. They have contradictory roles in cancer progression since both can have antitumor effects, through the induction of cell death, senescence and cell cycle arrest, and protumor effects by contributing to cancer cell spreading, proliferation, survival, and metastasis. TGF-β can control ROS production directly or by downregulating antioxidative systems. Meanwhile, ROS can influence TGF-β signaling and increase its expression as well as its activation from the latent complex. This way, both are building a strong interplay which can be taken as an advantage by cancer cells in order to increment their malignancy. In addition, both TGF-β and ROS are able to induce cell senescence, which in one way protects damaged cells from neoplastic transformation but also may collaborate in cancer progression. The mutual collaboration of TGF-β and ROS in tumorigenesis is highly complex, and, due to their differential roles in tumor progression, careful consideration should be taken when thinking of combinatorial targeting in cancer therapies.

    Topics: Cellular Senescence; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Oxidative Stress; Oxidoreductases; Reactive Oxygen Species; Signal Transduction; Transforming Growth Factor beta

2015
The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence.
    Nature reviews. Cancer, 2015, Volume: 15, Issue:7

    Radiotherapy plays a central part in curing cancer. For decades, most research on improving treatment outcomes has focused on modulating radiation-induced biological effects on cancer cells. Recently, we have better understood that components within the tumour microenvironment have pivotal roles in determining treatment outcomes. In this Review, we describe vascular, stromal and immunological changes that are induced in the tumour microenvironment by irradiation and discuss how these changes may promote radioresistance and tumour recurrence. We also highlight how this knowledge is guiding the development of new treatment paradigms in which biologically targeted agents will be combined with radiotherapy.

    Topics: Animals; Cell Hypoxia; Fibroblasts; Humans; Immune Tolerance; Neoplasm Recurrence, Local; Neoplasms; Radiation Tolerance; Transforming Growth Factor beta; Tumor Microenvironment; Vascular Endothelial Growth Factor A

2015
RUNX2 and the PI3K/AKT axis reciprocal activation as a driving force for tumor progression.
    Molecular cancer, 2015, Jul-25, Volume: 14

    From the first reported role of the transcription factor RUNX2 in osteoblast and chondrocyte differentiation and migration to its involvement in promigratory/proinvasive behavior of breast, prostate, and thyroid cancer cells, osteosarcoma, or melanoma cells, RUNX2 currently emerges as a key player in metastasis. In this review, we address the interaction of RUNX2 with the PI3K/AKT signaling pathway, one of the critical axes controlling cancer growth and metastasis. AKT, either by directly phosphorylating/activating RUNX2 or phosphorylating/inactivating regulators of RUNX2 stability or activity, contributes to RUNX2 transcriptional activity. Reciprocally, the activation of the PI3K/AKT pathway by RUNX2 regulation of its different components has been described in non-transformed and transformed cells. This mutual activation in the context of cancer cells exhibiting constitutive AKT activation and high levels of RUNX2 might constitute a major driving force in tumor progression and aggressiveness.

    Topics: Animals; Cell Transformation, Neoplastic; Core Binding Factor Alpha 1 Subunit; Disease Progression; Gene Expression Regulation, Neoplastic; Humans; Mitogen-Activated Protein Kinases; Neoplasms; Phosphatidylinositol 3-Kinases; Protein Binding; Proto-Oncogene Proteins c-akt; ras Proteins; Signal Transduction; Transforming Growth Factor beta

2015
[Advances of transforming growth factor-β inhibitors].
    Yao xue xue bao = Acta pharmaceutica Sinica, 2015, Volume: 50, Issue:4

    Transforming growth factor-β is an important cytokine with various bioactivities, including embryonic development, wound healing, chemotaxis and cell cycle regulation. Epithelial-mesenchymal transition (EMT) is the main pathway of tumor cell to obtain the ability of invasion and metastasis. The TGF-β is the key factor known to induce EMT in cancer cells and plays an important role in the process. In recent years, some progress has been obtained. Some TGF-β inhibitors have approved in the market or in clinical trials. TGF-β inhibitors can play an important role on the treatment of tumors, glaucoma, liver and kidney fibrosis disease and scar repair. Novel TGF-β inhibitors reported in recent years were reviewed in this article.

    Topics: Epithelial-Mesenchymal Transition; Humans; Neoplasms; Transforming Growth Factor beta; Wound Healing

2015
Association Between BMP-2 and Carcinogenicity.
    Spine, 2015, Volume: 40, Issue:23

    Literature review.. To evaluate the association between recombinant human bone morphogenetic protein-2 (rhBMP-2) and malignancy.. The use of rhBMP-2 in spine surgery has been the topic of much debate as studies assessing the association between rhBMP-2 and malignancy have come to conflicting conclusions.. A systematic review of the literature was performed using the PubMed-National Library of Medicine/National Institute of Health databases. Only non-clinical studies directly addressing BMP-2 and cancer were included. Articles were categorized by study type (animal, in vitro cell line/human/animal), primary malignancy, cancer attributes, and whether BMP-2 was pro-malignancy or not.. A total of 4,131 articles were reviewed. Of those, 515 articles made reference to both BMP-2 and cancer, 99 of which were found to directly examine the role of BMP-2 in cancer. Seventy-five studies were in vitro and 24 were animal studies. Forty-three studies concluded that BMP-2 enhanced cancer function, whereas 18 studies found that BMP-2 suppressed malignancy. Thirty-six studies did not examine whether BMP-2 enhanced or suppressed cancer function. Fifteen studies demonstrated BMP-2 dose dependence (9 enhancement, 6 suppression) and one study demonstrated no dose dependence. Nine studies demonstrated BMP-2 time dependence (6 enhancement, 3 suppression). However, no study demonstrated that BMP-2 caused cancer de novo.. Currently, conflicting data exist with regard to the effect of exogenous BMP-2 on cancer. The majority of studies addressed the role of BMP-2 in prostate (17%), breast (17%), and lung (15%) cancers. Most were in vitro studies (75%) and examined cancer invasiveness and metastatic potential (37%). Of 99 studies, there was no demonstration of BMP-2 causing cancer de novo. However, 43% of studies suggested that BMP-2 enhances tumor function, motivating more definitive research on the topic that also includes clinically meaningful dose- and time-dependence.. 2.

    Topics: Bone Morphogenetic Protein 2; Humans; Neoplasms; Recombinant Proteins; Spinal Fusion; Spine; Transforming Growth Factor beta

2015
Roles of Signaling Pathways in the Epithelial-Mesenchymal Transition in Cancer.
    Asian Pacific journal of cancer prevention : APJCP, 2015, Volume: 16, Issue:15

    The epithelial-mesenchymal transition (EMT) is a cellular process though which an epithelial phenotype can be converted into a phenotype of mesenchymal cells. Under physiological conditions EMT is important for embryogenesis, organ development, wound repair and tissue remodeling. However, EMT may also be activated under pathologic conditions, especially in carcinogenesis and metastatic progression. Major signaling pathways involved in EMT include transforming growth factor β(TGF-β), Wnt, Notch, Hedgehog and other signaling pathways. These pathways are related to several transcription factors, including Twist, Smads and zinc finger proteins snail and slug. These interact with each other to provide crosstalk between the relevant signaling pathways. This review lays emphasis on studying the relationship between EMT and signaling pathways in carcinogenesis and metastatic progression.

    Topics: Animals; Carcinogenesis; Epithelial-Mesenchymal Transition; Hedgehog Proteins; Humans; Neoplasm Metastasis; Neoplasms; Receptors, Notch; Signal Transduction; Transforming Growth Factor beta; Wnt Signaling Pathway

2015
TGF-beta signaling in cancer treatment.
    Current pharmaceutical design, 2014, Volume: 20, Issue:17

    The transforming growth factor-beta (TGF-β ) belongs to a superfamily of cytokines that act on protein kinase receptors at the plasma membrane to induce a plethora of biological signals that regulate cell growth and death, differentiation, immune response, angiogenesis and inflammation. Dysregulation of its pathway contributes to a broad variety of pathologies, including cancer. TGF-β is an important regulatory tumor suppressor factor in epithelial cells, where it early inhibits proliferation and induces apoptosis. However, tumor cells develop mechanisms to overcome the TGF-β -induced suppressor effects. Once this occurs, cells may respond to this cytokine inducing other effects that contribute to tumor progression. Indeed, TGF-β induces epithelial-mesenchymal transition (EMT), a process that is favored in tumor cells and facilitates migration and invasion. Furthermore, TGF-β mediates production of mitogenic growth factors, which stimulate tumor proliferation and survival. Finally, TGF-β is a well known immunosuppressor and pro-angiogenic factor. Many studies have identified the overexpression of TGF-β 1 in various types of human cancer, which correlates with tumor progression, metastasis, angiogenesis and poor prognostic outcome. For these reasons, different strategies to block TGF-β pathway in cancer have been developed and they can be classified in: (1) blocking antibodies and ligand traps; (2) antisense oligos; (3) TβRII and/or ALK5 inhibitors; (4) immune response-based strategies; (5) other inhibitors of the TGF-β pathway. In this review we will overview the two faces of TGF-β signaling in the regulation of tumorigenesis and we will dissect how targeting the TGF-β pathway may contribute to fight against cancer.

    Topics: Antineoplastic Agents; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Humans; Molecular Targeted Therapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2014
Th9 cells, new players in adaptive immunity.
    Trends in immunology, 2014, Volume: 35, Issue:2

    Upon antigen-specific stimulation, naïve CD4⁺ T cells have the potential to differentiate into various T helper (Th) cell subsets. Earlier models of Th cell differentiation focused on IFN-γ-producing Th1 cells and IL-4-secreting Th2 cells. The discovery of additional CD4⁺ Th cell subsets has extended our understanding of Th cell differentiation beyond this dichotomy. Among these is the recently described Th9 cell subset, which preferentially produces interleukin (IL)-9. Here, we review the latest developments in Th9 cell development and differentiation, focusing on contributing environmental signals, and discuss potential physiological and pathophysiological functions of these cells. We describe the challenges inherent to unambiguously defining roles for Th9 cells using the available experimental animal models, and suggest new experimental models to address these concerns.

    Topics: Adaptive Immunity; Animals; Humans; Hypersensitivity; Interferon Regulatory Factors; Interleukin-9; Mice; Models, Immunological; Neoplasms; Receptors, Antigen, T-Cell; Receptors, Interleukin-2; Receptors, Interleukin-4; Signal Transduction; T-Lymphocyte Subsets; T-Lymphocytes, Helper-Inducer; Transforming Growth Factor beta

2014
Neuropilin 1: function and therapeutic potential in cancer.
    Cancer immunology, immunotherapy : CII, 2014, Volume: 63, Issue:2

    Neuropilin 1 (NRP1) is a transmembrane glycoprotein that acts as a co-receptor for a number of extracellular ligands including class III/IV semaphorins, certain isoforms of vascular endothelial growth factor and transforming growth factor beta. An exact understanding of the role of NRP1 in the immune system has been obscured by the differences in NRP1 expression observed between mice and humans. In mice, NRP1 is selectively expressed on thymic-derived Tregs and greatly enhances immunosuppressive function. In humans, NRP1 is expressed on plasmacytoid dendritic cells (pDCs) where it aids in priming immune responses and on a subset of T regulatory cells (Tregs) isolated from secondary lymph nodes. Preliminary studies that show NRP1 expression on T cells confers enhanced immunosuppressive activity. However, the mechanism by which this activity is mediated remains unclear. NRP1 expression has also been identified on activated T cells and Tregs isolated from inflammatory microenvironments, suggesting NRP1 might represent a novel T cell activation marker. Of clinical interest, NRP1 may enhance Treg tumour infiltration and a decrease in NRP1+ Tregs correlates with successful chemotherapy, suggesting a specific role for NRP1 in cancer pathology. As a therapeutic target, NRP1 allows simultaneous targeting of NRP1-expressing tumour vasculature, NRP1+ Tregs and pDCs. With the development of anti-NRP1 monoclonal antibodies and cell-penetrating peptides, NRP1 represents a promising new target for cancer therapies. This paper reviews current knowledge on the role and function of NRP1 in Tregs and pDCs, both in physiological and cancer settings, as well as its potential as a therapeutic target in cancer.

    Topics: Animals; Dendritic Cells; Humans; Lymphocyte Activation; Neoplasms; Neuropilin-1; Proto-Oncogene Proteins c-met; Semaphorin-3A; Semaphorins; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2014
The microRNA networks of TGFβ signaling in cancer.
    Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine, 2014, Volume: 35, Issue:4

    In metazoans, the transforming growth factor β (TGFβ) signaling regulates a host of activities ranging from embryonic development to tissue homeostasis. The normal as well as tumor cells respond to this cytokine signaling pathway in a highly context-dependent manner. It acts as a potent tumor suppressor initially by inducing cell cycle arrest and apoptosis. But advanced tumors often misuse TGFβ signaling for tumor progression by selectively disabling the tumor suppressor arm and using other properties of TGFβ signaling such as induction of angiogenesis, epithelial to mesenchymal transition, and metastases. This dual role of TGFβ in cancer remained a mystery until recently. But recent advances in the field of microRNA provided a deeper understanding about this dual nature of TGFβ signaling in cancers. In the present review, we present an account of the role of microRNAs in deregulating TGFβ signaling and modulating cancer cell behavior during tumor initiation and cancer progression. This review also includes a discussion on the recent advances in the deregulation of TGFβ signaling in carcinogenesis.

    Topics: Animals; Humans; MicroRNAs; Neoplasm Invasiveness; Neoplasms; Neovascularization, Pathologic; PTEN Phosphohydrolase; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2014
Regulation of the anti-tumour immune response by cancer-associated fibroblasts.
    Seminars in cancer biology, 2014, Volume: 25

    The microenvironment of established tumours is often immunosuppressed, and this allows tumours to grow and disseminate without being eliminated by the patient's immune system. The recent FDA approval of immunotherapies such as ipilimumab and sipuleucel-T that directly activate the adaptive and innate immune responses has triggered interest in developing other novel anti-cancer approaches that modulate the immune system. Understanding how the different constituents of the tumour microenvironment influence the immune system is thus crucial and is expected to generate a plethora of factors that can be targeted to boost immunity and trigger long lasting anti-tumour efficacy. Cancer associated fibroblasts (CAFs) are a crucial component of the tumour microenvironment. Through secretion of multiple growth factors, cytokines and proteases, CAFs are known to be key effectors for tumour progression and can promote cancer cell growth, invasiveness and angiogenesis. However, recent publications have also linked CAF biology to innate and adaptive immune cell recruitment and regulation. Here, we review recent findings on how CAFs can influence the immune status of tumours through direct and indirect interaction with immune cells and other key components of the tumour microenvironment.

    Topics: Adaptive Immunity; Animals; Fibroblasts; Humans; Immunity, Cellular; Immunity, Innate; Immunotherapy; Inflammation; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2014
TGF-beta in CAF-mediated tumor growth and metastasis.
    Seminars in cancer biology, 2014, Volume: 25

    TGF-beta signaling is one of the major pathways controlling cell and tissue behavior not only in homeostasis but also in disease. During tumorigenesis TGF-beta orchestrated processes are key due to its dual role as tumor suppressor and tumor promoter. Important functions of this pathway have been described in a context-dependent manner both in epithelial cancer cells and in the tumor microenvironment during tumor progression. Carcinoma-associated fibroblasts (CAFs) are one of the most abundant stromal cell types in virtually all solid tumors. CAFs favor malignant progression by providing cancer cells with proliferative, migratory, survival and invasive capacities. A complex network of signaling pathways underlying their tumor-promoting properties is beginning to take shape. In this review, we examine current evidence on the emerging mechanisms involving TGF-beta in CAF-mediated cancer progression, and discuss their potential as therapeutic targets.

    Topics: Animals; Fibroblasts; Humans; Molecular Targeted Therapy; Neoplasm Metastasis; Neoplasms; Paracrine Communication; Signal Transduction; Transforming Growth Factor beta; Tumor Burden; Tumor Microenvironment

2014
TGF-β: duality of function between tumor prevention and carcinogenesis.
    Journal of the National Cancer Institute, 2014, Volume: 106, Issue:2

    Several mechanisms underlying tumor progression have remained elusive, particularly in relation to transforming growth factor beta (TGF-β). Although TGF-β initially inhibits epithelial growth, it appears to promote the progression of advanced tumors. Defects in normal TGF-β pathways partially explain this paradox, which can lead to a cascade of downstream events that drive multiple oncogenic pathways, manifesting as several key features of tumorigenesis (uncontrolled proliferation, loss of apoptosis, epithelial-to-mesenchymal transition, sustained angiogenesis, evasion of immune surveillance, and metastasis). Understanding the mechanisms of TGF-β dysregulation will likely reveal novel points of convergence between TGF-β and other pathways that can be specifically targeted for therapy.

    Topics: Animals; Anticarcinogenic Agents; Apoptosis; Cadherins; Carcinogenesis; Carcinogens; Cell Proliferation; Cell Transformation, Neoplastic; Disease Progression; Humans; Neoplasms; Neovascularization, Pathologic; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta; Up-Regulation

2014
Transforming growth factor-beta and matrix metalloproteinases: functional interactions in tumor stroma-infiltrating myeloid cells.
    TheScientificWorldJournal, 2014, Volume: 2014

    Transforming growth factor-beta (TGF-β) is a pleiotropic factor with several different roles in health and disease. In tumorigenesis, it may act as a protumorigenic factor and have a profound impact on the regulation of the immune system response. Matrix metalloproteinases (MMPs) are a family that comprises more than 25 members, which have recently been proposed as important regulators acting in tumor stroma by regulating the response of noncellular and cellular microenvironment. Tumor stroma consists of several types of resident cells and infiltrating cells derived from bone marrow, which together play crucial roles in the promotion of tumor growth and metastasis. In cancer cells, TGF-β regulates MMPs expression, while MMPs, produced by either cancer cells or residents' stroma cells, activate latent TGF-β in the extracellular matrix, together facilitating the enhancement of tumor progression. In this review we will focus on the compartment of myeloid stroma cells, such as tumor-associated macrophages, neutrophils, and dendritic and mast cells, which are potently regulated by TGF-β and produce large amounts of MMPs. Their interplay and mutual implications in the generation of pro-tumorigenic cancer microenvironment will be analyzed.

    Topics: Animals; Cell Communication; Gene Expression Regulation, Enzymologic; Humans; Metalloproteases; Myeloid Cells; Neoplasm Proteins; Neoplasms; Stromal Cells; Transforming Growth Factor beta; Tumor Microenvironment

2014
Regulated intramembrane proteolysis of the TGFβ type I receptor conveys oncogenic signals.
    Future oncology (London, England), 2014, Volume: 10, Issue:11

    Cancer cells produce high levels of TGFβ, a multipotent cytokine. Binding of TGFβ to its cell surface receptors, the transmembrane serine/threonine kinases TβRII and TβRI, causes phosphorylation and activation of intracellular latent Smad transcription factors. Nuclear Smads act in concert with specific transcription factors to reprogram epithelial cells to become invasive mesenchymal cells. TGFβ also propagates non-canonical signals, so it is crucial to have a better understanding of the underlying molecular mechanisms which favor this pathway. Here we highlight our recent discovery that TGFβ promotes the proteolytic cleavage of TβRI in cancer cells, resulting in the liberation and nuclear translocation of its intracellular domain, acting as co-regulator to transcribe pro-invasive genes. This newly identified oncogenic TGFβ pathway resembles the Notch signaling pathway. We discuss our findings in relation to Notch and provide a short overview of other growth factors that transduce signals via nuclear translocation of their cell surface receptors.

    Topics: Cell Membrane; Cell Nucleus; Cell Transformation, Neoplastic; Disease Progression; Humans; Neoplasms; Protein Serine-Threonine Kinases; Proteolysis; Receptor, Transforming Growth Factor-beta Type I; Receptors, Cytoplasmic and Nuclear; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2014
[SMAD family proteins: the current knowledge on their expression and potential role in neoplastic diseases].
    Postepy higieny i medycyny doswiadczalnej (Online), 2014, Mar-20, Volume: 68

    Transforming growth factor beta (TGF-β) plays a crucial role and takes part in many processes in the human body both in physiology and pathology. This cytokine is involved in angiogenesis, regulates apoptosis and stimulates divisions of cells, such as hepatocytes, lymphocytes or hematopoietic cells. SMAD proteins family is a unique group of particles responsible for transducting the signal induced by TGF-β into the nucleus. This molecules, after receiving a signal from activated TGF-β, act on transcription factors in the nucleus, leading directly to the expression of the corresponding genes. According to current knowledge, disturbances in the functioning of SMAD proteins are present in a number of diseases. The reduced expression was observed, for example in cardiovascular diseases such as primary pulmonary hypertension or myocardial infarction, autoimmune diseases for instance systemic lupus erythematosus and multiple sclerosis, Alzheimer's disease or osteoporosis. The latest clinical data showed the presence of mutations in SMAD proteins in cancerogenesis. Mutation of SMAD-4 protein can be detected in half of the patients with pancreatic cancer, 20% of patients with colorectal cancer and 10% of patients with lung cancer. However, mutation in SMAD-2 protein was observed in 7% of both patients with colorectal cancer and lung cancer. On the basis of numerous works, SMAD protein expression would be valuable prognostic factor in some of neoplastic diseases.

    Topics: Apoptosis; Colorectal Neoplasms; Gene Expression Regulation, Neoplastic; Humans; Lung Neoplasms; Mutation; Neoplasms; Neovascularization, Pathologic; Pancreatic Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2014
IGF binding proteins in cancer: mechanistic and clinical insights.
    Nature reviews. Cancer, 2014, Volume: 14, Issue:5

    The six members of the family of insulin-like growth factor (IGF) binding proteins (IGFBPs) were originally characterized as passive reservoirs of circulating IGFs, but they are now understood to have many actions beyond their endocrine role in IGF transport. IGFBPs also function in the pericellular and intracellular compartments to regulate cell growth and survival - they interact with many proteins, in addition to their canonical ligands IGF-I and IGF-II. Intranuclear roles of IGFBPs in transcriptional regulation, induction of apoptosis and DNA damage repair point to their intimate involvement in tumour development, progression and resistance to treatment. Tissue or circulating IGFBPs might also be useful as prognostic biomarkers.

    Topics: Animals; Apoptosis; Biological Availability; Biomarkers; Cell Nucleus; Cell Proliferation; Cell Survival; DNA Damage; Humans; Insulin-Like Growth Factor Binding Protein 1; Insulin-Like Growth Factor Binding Protein 2; Insulin-Like Growth Factor Binding Protein 3; Insulin-Like Growth Factor Binding Proteins; Ligands; Neoplasms; Prognosis; Protein Structure, Tertiary; Somatomedins; Transcription, Genetic; Transforming Growth Factor beta

2014
Novel patents and cancer therapies for transforming growth factor-beta and urokinase type plasminogen activator: potential use of their interplay in tumorigenesis.
    Recent patents on anti-cancer drug discovery, 2014, Volume: 9, Issue:3

    Transforming growth factor beta (TGF-β) plays different roles in health and disease. TGF-β has been assumed as a dual factor in tumor growth, since it can repress epithelial tumor development in early stages, while it acts as a tumor promoter in the late stages of tumor progression. The cancer cells, during cancerogenesis, acquire migration and invasion capacities and finally they metastasize. The urokinase type plasminogen activator (uPA) system, comprised of uPA, the cell surface receptor (uPAR) and plasminogen-plasmin, is involved in the proteolytic degradation of the extracellular matrix and it also regulates several critical cellular events by its capacity to trigger the activation of intracellular signaling pathways. This enables the cancer cell survival, its dissemination, and enhancement of cell malignancy during tumor progression. The expression of both uPA and uPAR is finely regulated in normal development, but their expression is deregulated in cancer. TGF-β regulates uPA expression in cancer cells while uPA, by conversion of plasminogen to active form, plasmin, may release TGF-β from its latent state. Thus, these pathways cross-regulate each other by mutual feedback contributing to tumor progression. Here, we review the specific roles and the interplay between TGF-β and uPA system in cancer cells, the current cancer therapies and the novel patents focused mainly on uPA and TGF-beta ligands and their cell surface receptors respectively. Finally, with regard to the mutual activity of uPA and TGF-β in tumorigenesis, the aim of this review is to expose the potentiality of TGF-β and uPA systems as becoming combinatorial targets for therapies and patents.

    Topics: Antineoplastic Agents; Carcinogenesis; Humans; Neoplasms; Patents as Topic; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator

2014
[Hypoxia, epithelia-mesenchymal transition and cancer].
    Zhonghua bing li xue za zhi = Chinese journal of pathology, 2014, Volume: 43, Issue:3

    Topics: Cell Hypoxia; Epithelial-Mesenchymal Transition; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Neoplasms; Receptors, Notch; Signal Transduction; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta; Twist-Related Protein 1; Wnt Proteins

2014
SMAD7: a timer of tumor progression targeting TGF-β signaling.
    Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine, 2014, Volume: 35, Issue:9

    In the context of cancer, transforming growth factor β (TGF-β) is a cell growth suppressor; however, it is also a critical inducer of invasion and metastasis. SMAD is the important mediator of TGF-β signaling pathway, which includes receptor-regulated SMADs (R-SMADs), common-mediator SMADs (co-SMADs), and inhibitory SMADs (I-SMADs). I-SMADs block the activation of R-SMADs and co-SMADs and thus play important roles especially in the SMAD-dependent signaling. SMAD7 belongs to the I-SMADs. As an inhibitor of TGF-β signaling, SMAD7 is overexpressed in numerous cancer types and its abundance is positively correlated to the malignancy. Emerging evidence has revealed the switch-in-role of SMAD7 in cancer, from a TGF-β inhibiting protein at the early stages that facilitates proliferation to an enhancer of invasion at the late stages. This role change may be accompanied or elicited by the tumor microenvironment and/or somatic mutation. Hence, current knowledge suggests a tumor-favorable timer nature of SMAD7 in cancer progression. In this review, we summarized the advances and recent findings of SMAD7 and TGF-β signaling in cancer, followed by specific discussion on the possible factors that account for the functional changes of SMAD7.

    Topics: Animals; Disease Progression; Humans; Models, Biological; Neoplasms; Signal Transduction; Smad7 Protein; Time Factors; Transforming Growth Factor beta

2014
TGF- β: an important mediator of allergic disease and a molecule with dual activity in cancer development.
    Journal of immunology research, 2014, Volume: 2014

    The transforming growth factor- β (TGF- β ) superfamily is a family of structurally related proteins that includes TGF- β , activins/inhibins, and bone morphogenic proteins (BMPs). Members of the TGF- β superfamily regulate cellular functions such as proliferation, apoptosis, differentiation, and migration and thus play key roles in organismal development. TGF- β is involved in several human diseases, including autoimmune disorders and vascular diseases. Activation of the TGF- β receptor induces phosphorylation of serine/threonine residues and triggers phosphorylation of intracellular effectors (Smads). Once activated, Smad proteins translocate to the nucleus and induce transcription of their target genes, regulating various processes and cellular functions. Recently, there has been an attempt to correlate the effect of TGF- β with various pathological entities such as allergic diseases and cancer, yielding a new area of research known as "allergooncology," which investigates the mechanisms by which allergic diseases may influence the progression of certain cancers. This knowledge could generate new therapeutic strategies aimed at correcting the pathologies in which TGF- β is involved. Here, we review recent studies that suggest an important role for TGF- β in both allergic disease and cancer progression.

    Topics: Animals; Cell Transformation, Neoplastic; Humans; Hypersensitivity; Multigene Family; Neoplasms; Protein Binding; Protein Biosynthesis; Protein Isoforms; Protein Multimerization; Proteolysis; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2014
TGFBR1 and cancer susceptibility.
    Transactions of the American Clinical and Climatological Association, 2014, Volume: 125

    Transforming growth factor beta (TGF-β) is a potent inhibitor of cell growth. TGFBR1 6A is a polymorphism consisting of a 9-base pair in-frame deletion within exon 1 of the type I TGF-β receptor (TGFBR1), which results in a receptor with decreased TGF-β signaling capability. The discovery of an association between TGFBR1*6A and cancer susceptibility led to the hypothesis that hypomorphic variants of the TGF-β signaling pathway may predispose to the development of cancer. This hypothesis was tested in vivo with the development of a mouse model of Tgfbr1 haploinsufficiency. Tgfbr1 (+/-) mice developed twice as many intestinal tumors as Tgfbr1 (+/+). Tgfbr1 haploinsufficiency was also associated with early onset adenocarcinoma and increased tumor cell proliferation. A case control study identified two haplotypes associated with constitutively decreased TGFBR1 and substantially increased colorectal cancer risk indicating that TGFBR1 may act as a potent modifier of cancer risk.

    Topics: Animals; Biomarkers, Tumor; Cell Transformation, Neoplastic; Genetic Predisposition to Disease; Humans; Mice, Knockout; Neoplasms; Phenotype; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Risk Factors; Signal Transduction; Transforming Growth Factor beta

2014
The multifaceted role of the embryonic gene Cripto-1 in cancer, stem cells and epithelial-mesenchymal transition.
    Seminars in cancer biology, 2014, Volume: 29

    Cripto-1 (CR-1)/Teratocarcinoma-derived growth factor1 (TDGF-1) is a cell surface glycosylphosphatidylinositol (GPI)-linked glycoprotein that can function either in cis (autocrine) or in trans (paracrine). The cell membrane cis form is found in lipid rafts and endosomes while the trans acting form lacking the GPI anchor is soluble. As a member of the epidermal growth factor (EGF)/Cripto-1-FRL-1-Cryptic (CFC) family, CR-1 functions as an obligatory co-receptor for the transforming growth factor-β (TGF-β) family members, Nodal and growth and differentiation factors 1 and 3 (GDF1/3) by activating Alk4/Alk7 signaling pathways that involve Smads 2, 3 and 4. In addition, CR-1 can activate non-Smad-dependent signaling elements such as PI3K, Akt and MAPK. Both of these pathways depend upon the 78kDa glucose regulated protein (GRP78). Finally, CR-1 can facilitate signaling through the canonical Wnt/β-catenin and Notch/Cbf-1 pathways by functioning as a chaperone protein for LRP5/6 and Notch, respectively. CR-1 is essential for early embryonic development and maintains embryonic stem cell pluripotentiality. CR-1 performs an essential role in the etiology and progression of several types of human tumors where it is expressed in a population of cancer stem cells (CSCs) and facilitates epithelial-mesenchymal transition (EMT). In this context, CR-1 can significantly enhance tumor cell migration, invasion and angiogenesis. Collectively, these facts suggest that CR-1 may be an attractive target in the diagnosis, prognosis and therapy of several types of human cancer.

    Topics: Activin Receptors, Type I; beta Catenin; Cell Membrane; Endoplasmic Reticulum Chaperone BiP; Epithelial-Mesenchymal Transition; GPI-Linked Proteins; Heat-Shock Proteins; Humans; Intercellular Signaling Peptides and Proteins; Membrane Proteins; Neoplasm Invasiveness; Neoplasm Proteins; Neoplasms; Neoplastic Stem Cells; Neovascularization, Pathologic; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Receptors, Notch; Smad2 Protein; Smad3 Protein; Smad4 Protein; TGF-beta Superfamily Proteins; Transforming Growth Factor beta; Wnt Proteins; Wnt Signaling Pathway

2014
TGFβ in T cell biology and tumor immunity: Angel or devil?
    Cytokine & growth factor reviews, 2014, Volume: 25, Issue:4

    The evolutionally conserved transforming growth factor β (TGFβ) affects multiple cell types in the immune system by either stimulating or inhibiting their differentiation and function. Studies using transgenic mice with ablation of TGFβ or its receptor have revealed the biological significance of TGFβ signaling in the control of T cells. However, it is now clear that TGFβ is more than an immunosuppressive cytokine. Disruption of TGFβ signaling pathway also leads to impaired generation of certain T cell populations. Therefore, in the normal physiological state, TGFβ actively maintains T cell homeostasis and regulates T cell function. However, in the tumor microenvironment, TGFβ creates an immunosuppressive milieu that inhibits antitumor immunity. Here, we review recent advances in our understanding of the roles of TGFβ in the regulation of T cells and tumor immunity.

    Topics: Animals; Cell Differentiation; Dendritic Cells; Humans; Macrophages; Mice; Mice, Transgenic; Neoplasms; Neutrophils; Receptors, Transforming Growth Factor beta; Signal Transduction; T-Lymphocytes; Transforming Growth Factor beta; Tumor Escape; Tumor Microenvironment

2014
Signaling pathway cooperation in TGF-β-induced epithelial-mesenchymal transition.
    Current opinion in cell biology, 2014, Volume: 31

    Transdifferentiation of epithelial cells into cells with mesenchymal properties and appearance, that is, epithelial-mesenchymal transition (EMT), is essential during development, and occurs in pathological contexts, such as in fibrosis and cancer progression. Although EMT can be induced by many extracellular ligands, TGF-β and TGF-β-related proteins have emerged as major inducers of this transdifferentiation process in development and cancer. Additionally, it is increasingly apparent that signaling pathways cooperate in the execution of EMT. This update summarizes the current knowledge of the coordination of TGF-β-induced Smad and non-Smad signaling pathways in EMT, and the remarkable ability of Smads to cooperate with other transcription-directed signaling pathways in the control of gene reprogramming during EMT.

    Topics: Animals; Embryonic Development; Epithelial Cells; Epithelial-Mesenchymal Transition; Fibrosis; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2014
Hey bHLH transcription factors.
    Current topics in developmental biology, 2014, Volume: 110

    Hey bHLH transcription factors are direct targets of canonical Notch signaling. The three mammalian Hey proteins are closely related to Hes proteins and they primarily repress target genes by either directly binding to core promoters or by inhibiting other transcriptional activators. Individual candidate gene approaches and systematic screens identified a number of Hey target genes, which often encode other transcription factors involved in various developmental processes. Here, we review data on interaction partners and target genes and conclude with a model for Hey target gene regulation. Furthermore, we discuss how expression of Hey proteins affects processes like cell fate decisions and differentiation, e.g., in cardiovascular, skeletal, and neural development or oncogenesis and how this relates to the observed developmental defects and phenotypes observed in various knockout mice.

    Topics: Animals; Basic Helix-Loop-Helix Transcription Factors; Cardiovascular System; Cell Cycle Proteins; Cell Differentiation; COUP Transcription Factor II; Gene Expression Regulation; Gene Expression Regulation, Neoplastic; Heart Diseases; Humans; Mice, Knockout; Muscle Development; Muscle, Smooth, Vascular; Neoplasms; Nuclear Receptor Coactivator 2; Receptors, Notch; Repressor Proteins; Transforming Growth Factor beta

2014
The miRNA23b-regulated signaling network as a key to cancer development--implications for translational research and therapeutics.
    Journal of molecular medicine (Berlin, Germany), 2014, Volume: 92, Issue:11

    A growing body of evidence indicates that microRNA23b (miR23b) is pleiotropic-it plays important roles in regulating physiological functions of cells, in regulating differentiation of cells and in regulating cellular immune responses. Our review of the literature showed that dysregulation of miR23b expression is implicated in the disruption of these cellular mechanisms and development of diseases such as cancer. MiR23b dysregulation appears to do this by modulating the expression level of candidate gene products involved in a network of signaling pathways including TGF-beta and Notch pathways that govern malignant properties of cancer cells such as motility and invasiveness. More recently, miR23b regulation of gene expression has also been associated with cancer stem cells and chemoresistance. Our review covers miR23b's role in immunity, endothelial function, differentiation, and cancer as well as its potential for translation into future cancer diagnostics and therapeutics.

    Topics: Animals; Cell Differentiation; Endothelial Cells; Female; Gene Expression Regulation; Homeostasis; Humans; Immune System; Male; Mice; MicroRNAs; Neoplasms; Neoplastic Stem Cells; Protein Biosynthesis; Receptors, Notch; Signal Transduction; Transforming Growth Factor beta; Translational Research, Biomedical

2014
Extracellular matrix as a contextual determinant of transforming growth factor-β signaling in epithelial-mesenchymal transition and in cancer.
    Cell adhesion & migration, 2014, Volume: 8, Issue:6

    Extracellular matrix (ECM) provides both structural support and contextual information to cells within tissues and organs. The combination of biochemical and biomechanical signals from the ECM modulates responses to extracellular signals toward differentiation, proliferation, or apoptosis; alterations in the ECM are necessary for development and remodeling processes, but aberrations in the composition and organization of ECM are associated with disease pathology and can predispose to development of cancer. The primary cell surface sensors of the ECM are the integrins, which provide the physical connection between the ECM and the cytoskeleton and also convey biochemical information about the composition of the ECM. Transforming growth factor-β (TGF-β) is an extracellular signaling molecule that is a powerful controller of a variety of cellular functions, and that has been found to induce very different outcomes according to cell type and cellular context. It is becoming clear that ECM-mediated signaling through integrins is reciprocally influenced by TGF-β: integrin expression, activation, and responses are affected by cellular exposure to TGF-β, and TGF-β activation and cellular responses are in turn controlled by signaling from the ECM through integrins. Epithelial-mesenchymal transition (EMT), a physiological process that is activated by TGF-β in normal development and in cancer, is also affected by the composition and structure of the ECM. Here, we will outline how signaling from the ECM controls the contextual response to TGF-β, and how this response is selectively modulated during disease, with an emphasis on recent findings, current challenges, and future opportunities.

    Topics: Cell Differentiation; Cytoskeleton; Epithelial Cells; Epithelial-Mesenchymal Transition; Extracellular Matrix; Humans; Integrins; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2014
Genome-wide association and longitudinal analyses reveal genetic loci linking pubertal height growth, pubertal timing and childhood adiposity.
    Human molecular genetics, 2013, Jul-01, Volume: 22, Issue:13

    The pubertal height growth spurt is a distinctive feature of childhood growth reflecting both the central onset of puberty and local growth factors. Although little is known about the underlying genetics, growth variability during puberty correlates with adult risks for hormone-dependent cancer and adverse cardiometabolic health. The only gene so far associated with pubertal height growth, LIN28B, pleiotropically influences childhood growth, puberty and cancer progression, pointing to shared underlying mechanisms. To discover genetic loci influencing pubertal height and growth and to place them in context of overall growth and maturation, we performed genome-wide association meta-analyses in 18 737 European samples utilizing longitudinally collected height measurements. We found significant associations (P < 1.67 × 10(-8)) at 10 loci, including LIN28B. Five loci associated with pubertal timing, all impacting multiple aspects of growth. In particular, a novel variant correlated with expression of MAPK3, and associated both with increased prepubertal growth and earlier menarche. Another variant near ADCY3-POMC associated with increased body mass index, reduced pubertal growth and earlier puberty. Whereas epidemiological correlations suggest that early puberty marks a pathway from rapid prepubertal growth to reduced final height and adult obesity, our study shows that individual loci associating with pubertal growth have variable longitudinal growth patterns that may differ from epidemiological observations. Overall, this study uncovers part of the complex genetic architecture linking pubertal height growth, the timing of puberty and childhood obesity and provides new information to pinpoint processes linking these traits.

    Topics: Adiposity; Adolescent; Age Factors; Body Height; Body Mass Index; Child; Female; Follow-Up Studies; Gene Expression; Genetic Linkage; Genome-Wide Association Study; Humans; Male; Menarche; Mitogen-Activated Protein Kinase 3; Neoplasms; Phenotype; Puberty; Quantitative Trait Loci; Signal Transduction; Transforming Growth Factor beta; Young Adult

2013
Association of SMAD7 rs12953717 polymorphism with cancer: a meta-analysis.
    PloS one, 2013, Volume: 8, Issue:3

    Accumulating evidence has suggested that Mothers against decapentaplegic homolog 7 (SMAD7) rs12953717 polymorphism might be related to cancer risk. However, epidemiologic findings have been inconsistent. We therefore performed a meta-analysis to clarify the association between the SMAD7 rs12953717 polymorphism and cancer risk.. A comprehensive search was conducted to identify all eligible studies of SMAD7 rs12953717 polymorphism and cancer risk. We used odds ratios (ORs) to assess the strength of the association, and 95% confidence intervals (CIs) to give a sense of the precision of the estimate. Heterogeneity, publication bias, and sensitivity analysis were also explored.. A total of 14 case-control studies, including 16928 cases and 14781 controls, were included in the present meta-analysis. The overall results showed that the variant genotypes were associated with a significantly increased risk of all cancer types (homozygote comparison, OR = 1.23, 95%CI = 1.10-1.38, P<0.01; heterozygote comparison, OR = 1.12, 95%CI = 1.02-1.22, P = 0.02; recessive model, OR = 1.17, 95%CI = 1.07-1.29, P<0.01; dominant model, OR = 1.15, 95%CI = 1.06-1.25, P<0.01; allelic model, OR = 1.12, 95%CI = 1.06-1.18, P<0.01). Further sensitivity analysis confirmed the significant association. In the subgroup analysis by ethnicity, SMAD7 rs12953717 polymorphism was significantly associated with cancer risk in both Caucasians and Asians. In the subgroup analysis by cancer types, SMAD7 rs12953717 polymorphism was significantly associated with colorectal cancer.. Our investigations demonstrate that rs12953717 polymorphism is associated with the susceptibility of cancer. Large-scale and well-designed case-control studies are necessary to validate the risk identified in the present meta-analysis.

    Topics: Alleles; Case-Control Studies; Genetic Predisposition to Disease; Heterozygote; Homozygote; Humans; Models, Genetic; Neoplasms; Odds Ratio; Polymorphism, Single Nucleotide; Sensitivity and Specificity; Smad7 Protein; Transforming Growth Factor beta

2013
The relevance of the TGF-β Paradox to EMT-MET programs.
    Cancer letters, 2013, Nov-28, Volume: 341, Issue:1

    The role of transforming growth factor-β (TGF-β) during tumorigenesis is complex and paradoxical, reflecting its ability to function as a tumor suppressor in normal and early-stage cancers, and as a tumor promoter in their late-stage counterparts. The switch in TGF-β function is known as the "TGF-β Paradox," whose manifestations are intimately linked to the initiation of epithelial-mesenchymal transition (EMT) programs in developing and progressing carcinomas. Indeed, as carcinoma cells emerge from EMT programs stimulated by TGF-β, they readily display a variety of acquired phenotypes that provide a selective advantage to growing carcinomas, including (i) enhanced cell migration and invasion; (ii) heightened resistance to cytotoxic agents, targeted chemotherapeutic, and radiation treatments; and (iv) boosted expansion of cancer-initiating and stem-like cell populations that underlie tumor metastasis and disease recurrence. At present, the molecular, cellular, and microenvironmental mechanisms that enable post-EMT and metastatic carcinoma cells to hijack the oncogenic activities of TGF-β remain incompletely understood. Additionally, the molecular mechanisms that counter EMT programs and limit the aggressiveness of late-stage carcinomas, events that transpire via mesenchymal-epithelial transition (MET) reactions, also need to be further elucidated. Here we review recent advances that provide new insights into how TGF-β promotes EMT programs in late-stage carcinoma cells, as well as how these events are balanced by MET programs during the development and metastatic progression of human carcinomas.

    Topics: Alternative Splicing; Animals; Bone Morphogenetic Proteins; Cell Hypoxia; DNA-Binding Proteins; Epithelial-Mesenchymal Transition; Fibroblasts; Heat-Shock Proteins; Humans; Integrins; MicroRNAs; Neoplasms; Neoplastic Stem Cells; Proto-Oncogene Proteins c-abl; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2013
Mechanisms that link the oncogenic epithelial-mesenchymal transition to suppression of anoikis.
    Journal of cell science, 2013, Jan-01, Volume: 126, Issue:Pt 1

    The oncogenic epithelial-mesenchymal transition (EMT) contributes to tumor progression in various context-dependent ways, including increased metastatic potential, expansion of cancer stem cell subpopulations, chemo-resistance and disease recurrence. One of the hallmarks of EMT is resistance of tumor cells to anoikis. This resistance contributes to metastasis and is a defining property not only of EMT but also of cancer stem cells. Here, we review the mechanistic coupling between EMT and resistance to anoikis. The discussion focuses on several key aspects. First, we provide an update on new pathways that lead from the loss of E-cadherin to anoikis resistance. We then discuss the relevance of transcription factors that are crucial in wound healing in the context of oncogenic EMT. Next, we explore the consequences of the breakdown of cell-polarity complexes upon anoikis sensitivity, through the Hippo, Wnt and transforming growth factor β (TGF-β) pathways, emphasizing points of crossregulation. Finally, we summarize the direct regulation of cell survival genes through EMT-inducing transcription factors, and the roles of the tyrosine kinases focal adhesion kinase (FAK) and TrkB neurotrophin receptor in EMT-related regulation of anoikis. Emerging from these studies are unifying principles that will lead to improvements in cancer therapy by reprogramming sensitivity of anoikis.

    Topics: Animals; Anoikis; Epithelial-Mesenchymal Transition; Focal Adhesion Protein-Tyrosine Kinases; Humans; Neoplasms; Transforming Growth Factor beta

2013
MicroRNAs play a central role in molecular dysfunctions linking inflammation with cancer.
    Immunological reviews, 2013, Volume: 253, Issue:1

    It is now largely admitted that a pro-inflammatory environment may curtail anti-tumor immunity and favor cancer initiation and progression. The discovery that small non-coding regulatory RNAs, namely microRNAs (miRNAs), regulate all aspects of cell proliferation, differentiation, and function has shed a new light on regulatory mechanisms linking inflammation and cancer. Thus, miRNAs such as miR-21, miR-125b, miR-155, miR-196, and miR-210 that are critical for the immune response or hypoxia are often overexpressed in cancers and leukemias. Given the high number of their target transcripts, their deregulation may have a number of deleterious consequences, depending on the cellular context. In this review, we focus on how the factors encoded by transcripts targeted by these five miRNAs, be they transcription factors, tumor-suppressors, or regulators of different signaling pathways, can deregulate the immune response and favor pro-tumor immunity. Furthermore, we expose how the misdirected action of the main regulators of these miRNAs, such as nuclear factor κB (NF-κB), activator protein-1 (AP-1), and signal transduction and activators of transcription (STAT) transcription factors, or AKT and transforming growth factor β (TGFβ) signaling pathways, can contribute to decrease anti-tumor immunity and enhance cell proliferation and oncogenesis. We conclude by briefly discussing about how these discoveries may possibly lead to the development of new miRNA-based cancer therapies.

    Topics: Animals; Biological Therapy; Cell Proliferation; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; Immunity; Inflammation; MicroRNAs; Neoplasms; Signal Transduction; Transcription Factors; Transforming Growth Factor beta; Tumor Escape

2013
The role of endosomal signaling triggered by metastatic growth factors in tumor progression.
    Cellular signalling, 2013, Volume: 25, Issue:7

    Within tumor microenvironment, a lot of growth factors such as hepatocyte growth factor and epidermal growth factor may induce similar signal cascade downstream of receptor tyrosine kinase (RTK) and trigger tumor metastasis synergistically. In the past decades, the intimate relationship of RTK-mediated receptor endocytosis with signal transduction was well established. In general, most RTK undergoes clathrin-dependent endocytosis and/or clathrin-independent endocytosis. The internalized receptors may sustain the signaling within early endosome, recycling to plasma membrane for subsequent ligand engagement or sorting to late endosomes/lysosome for receptor degradation. Moreover, receptor endocytosis influences signal transduction in a temporal and spatial manner for periodical and polarized cellular processes such as cell migration. The endosomal signalings triggered by various metastatic factors are quite similar in some critical points, which are essential for triggering cell migration and tumor progression. There are common regulators for receptor endocytosis including dynamin, Rab4, Rab5, Rab11 and Cbl. Moreover, many critical regulators within the RTK signal pathway such as Grb2, p38, PKC and Src were also modulators of endocytosis. In the future, these may constitute a new category of targets for prevention of tumor metastasis.

    Topics: Disease Progression; Endocytosis; Endosomes; Epidermal Growth Factor; Fibroblast Growth Factors; Hepatocyte Growth Factor; Humans; Neoplasm Metastasis; Neoplasms; Platelet-Derived Growth Factor; Signal Transduction; Transforming Growth Factor beta

2013
Natural products and transforming growth factor-beta (TGF-β) signaling in cancer development and progression.
    Current cancer drug targets, 2013, Volume: 13, Issue:5

    Actions of many herbal medicine products for cancer treatment are linked to an altered production of TGF-β in the target cells. An altered TGF-β production in the target cells will have profound effects on the patients. Therefore, it is important that we review the pros and cons of these products on cancer development and progression in terms of TGF-β signaling. It has been well established that TGF-β is growth inhibitory to benign cells or early stages of cancer cells but it is tumor promoting and metastatic for advanced malignancies. Further, many dietary components can alter gene-specific DNA methylation levels in systemic and in target tissues. Since TGF-β signaling in cancer is closely linked to the DNA methylation profiles, we also review the effect of dietary components on DNA methylation. In light of this knowledge, it is important to note that many natural products that can induce TGF-β production in the target cells may be beneficial in preventing cancer development but may be harmful for cancer patients, especially when they harbor advanced stage cancer. A discussion of the effect of herbal natural products on cancer can be divided into three categories. The first category of herbal medicine products will be those related to the induction of cancer as far as TGF-β is concerned. Since TGF-β is growth inhibitory and pro-apoptosis to benign cells, any herbal medication that can induce the production of TGF-β in the target cells will be beneficial to the patients. However, such herbal medicine may not necessarily be beneficial for patients with established and advanced cancer. The second category of herbal products will inhibit TGF-β signaling and will reduce TGF-β mediated growth promotion and metastasis in advanced cancers. For patients with established and advanced cancer, agents that can inhibit the production of TGF-β may also inhibit cancer growth and metastasis. Finally, the third category of herbal products has no impact on TGF-β signaling, such as lycopene.

    Topics: Animals; Antineoplastic Agents; Biological Products; Disease Progression; Humans; Neoplasms; Phytotherapy; Signal Transduction; Transforming Growth Factor beta

2013
Targeting TGF-β signaling in cancer.
    Expert opinion on therapeutic targets, 2013, Volume: 17, Issue:7

    The transforming growth factor-β (TGF-β) signaling pathway has a pivotal role in tumor suppression and yet, paradoxically, in tumor promotion. Functional context dependent insights into the TGF-β pathway are crucial in developing TGF-β-based therapeutics for cancer.. This review discusses the molecular mechanism of the TGF-β pathway and describes the different ways of tumor suppression by TGF-β. It is then explained how tumors can evade these effects and how TGF-β contributes to further growing and spreading of some of the tumors. In the last part of the review, the data on targeting TGF-β pathway for cancer treatment is assessed. This review focuses on anti-TGF-β based treatment and other options targeting activated pathways in tumors where the TGF-β tumor suppressor pathway is lost. Pre-clinical as well up to date results of the most recent clinical trials are given.. Targeting the TGF-β pathway can be a promising direction in cancer treatment. However, several challenges still exist, the most important are differentiating between the carcinogenic effects of TGF-β and its other physiological roles, and delineating the tumor suppressive versus the tumor promoting roles of TGF-β in each specific tumor. Future studies are needed in order to find safer and more effective TGF-β-based drugs.

    Topics: Animals; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2013
What are the molecules involved in regulatory T-cells induction by dendritic cells in cancer?
    Clinical & developmental immunology, 2013, Volume: 2013

    Dendritic cells (DCs) are essential for the maintenance of homeostasis in the organism, and they do that by modulating lymphocyte priming, expansion, and response patterns according to signals they receive from the environment. The induction of suppressive lymphocytes by DCs is essential to hinder the development of autoimmune diseases but can be reverted against homeostasis when in the context of neoplasia. In this setting, the induction of suppressive or regulatory T cells contributes to the establishment of a state of tolerance towards the tumor, allowing it to grow unchecked by an otherwise functional immune system. Besides affecting its local environment, tumor also has been described as potent sources of anti-inflammatory/suppressive factors, which may act systemically, generating defects in the differentiation and maturation of immune cells, far beyond the immediate vicinity of the tumor mass. Cytokines, as IL-10 and TGF-beta, as well as cell surface molecules like PD-L1 and ICOS seem to be significantly involved in the redirection of DCs towards tolerance induction, and recent data suggest that tumor cells may, indeed, modulate distinct DCs subpopulations through the involvement of these molecules. It is to be expected that the identification of such molecules should provide molecular targets for more effective immunotherapeutic approaches to cancer.

    Topics: B7-H1 Antigen; Cell Communication; Dendritic Cells; Gene Expression Regulation; Humans; Immune Tolerance; Inducible T-Cell Co-Stimulator Protein; Interleukin-10; Lymphocyte Activation; Neoplasms; Signal Transduction; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Microenvironment

2013
Safety and effectiveness of recombinant human bone morphogenetic protein-2 for spinal fusion: a meta-analysis of individual-participant data.
    Annals of internal medicine, 2013, Jun-18, Volume: 158, Issue:12

    Recombinant human bone morphogenetic protein-2 (rhBMP-2) is widely used to promote fusion in spinal surgery, but its safety has been questioned.. To evaluate the effectiveness and safety of rhBMP-2.. Individual-participant data obtained from the sponsor or investigators and data extracted from study publications identified by systematic bibliographic searches through June 2012.. Randomized, controlled trials of rhBMP-2 versus iliac crest bone graft (ICBG) in spinal fusion surgery for degenerative disc disease and related conditions and observational studies in similar populations for investigation of adverse events.. Individual-participant data from 11 eligible of 17 provided trials sponsored by Medtronic (Minneapolis, Minnesota) (n = 1302) and 1 of 2 other eligible trials (n = 106) were included. Additional aggregate adverse event data were extracted from 35 published observational studies.. Primary outcomes were pain (assessed with the Oswestry Disability Index [ODI] or Short Form-36), fusion, and adverse events. At 24 months, ODI scores were 3.5% lower (better) with rhBMP-2 than with ICBG (95% CI, 0.5% to 6.5%) and radiographic fusion was 12% higher (CI, 2% to 23%). At or shortly after surgery, pain was more common with rhBMP-2 (odds ratio, 1.78 [CI, 1.06 to 2.95]). Cancer was more common after rhBMP-2 (relative risk, 1.98 [CI, 0.86 to 4.54]), but the small number of events precluded definite conclusions.. The observational studies were diverse and at risk of bias.. At 24 months, rhBMP-2 increases fusion rates, reduces pain by a clinically insignificant amount, and increases early postsurgical pain compared with ICBG. Evidence of increased cancer incidence is inconclusive.. Yale University Open Data Access Project.

    Topics: Bone Morphogenetic Protein 2; Disability Evaluation; Humans; Ilium; Incidence; Intervertebral Disc Degeneration; Neoplasms; Pain, Postoperative; Recombinant Proteins; Spinal Fusion; Time Factors; Transforming Growth Factor beta; Treatment Outcome

2013
Effectiveness and harms of recombinant human bone morphogenetic protein-2 in spine fusion: a systematic review and meta-analysis.
    Annals of internal medicine, 2013, Jun-18, Volume: 158, Issue:12

    Recombinant human bone morphogenetic protein-2 (rhBMP-2) is used as a bone graft substitute in spinal fusion, which unites (fuses) bones in the spine. The accuracy and completeness of journal publications of industry-sponsored trials on the effectiveness and harms of rhBMP-2 has been called into question.. To independently assess the effectiveness and harms of rhBMP-2 in spinal fusion and reporting bias in industry-sponsored journal publications.. Individual-patient data (IPD) from 17 industry-sponsored studies; related internal documents; and searches of MEDLINE (1996 to August 2012), other databases, and reference lists.. Randomized, controlled trials (RCTs) and cohort studies of rhBMP-2 versus any control and uncontrolled studies of harms.. Effectiveness outcomes in IPD were recalculated using consistent definitions. Study characteristics and results were abstracted by 1 investigator and confirmed by another. Two investigators independently assessed quality using predefined criteria.. Thirteen RCTs and 31 cohort studies were included. For lumbar spine fusion, rhBMP-2 and iliac crest bone graft were similar in overall success, fusion, and other effectiveness measures and in risk for any adverse event, although rates were high across interventions (77% to 93% at 24 months from surgery). For anterior lumbar interbody fusion, rhBMP-2 was associated with nonsignificantly increased risk for retrograde ejaculation and urogenital problems. For anterior cervical spine fusion, rhBMP-2 was associated with increased risk for wound complications and dysphagia. At 24 months, the cancer risk was increased with rhBMP-2 (risk ratio, 3.45 [95% CI, 1.98 to 6.00]), but event rates were low and cancer was heterogeneous. Early journal publications misrepresented the effectiveness and harms through selective reporting, duplicate publication, and underreporting.. Outcome assessment was not blinded, and ascertainment of harms in trials was poor. No trials were truly independent of industry sponsorship.. In spinal fusion, rhBMP-2 has no proven clinical advantage over bone graft and may be associated with important harms, making it difficult to identify clear indications for rhBMP-2. Earlier disclosure of all relevant data would have better informed clinicians and the public than the initial published trial reports did.. Yale University and Medtronic.

    Topics: Bone Morphogenetic Protein 2; Conflict of Interest; Drug Industry; Humans; Ilium; Incidence; Intervertebral Disc Degeneration; Neoplasms; Off-Label Use; Publication Bias; Publishing; Recombinant Proteins; Spinal Fusion; Transforming Growth Factor beta; Treatment Outcome

2013
The multiple facets of the TGF-β family cytokine growth/differentiation factor-15/macrophage inhibitory cytokine-1.
    Cytokine & growth factor reviews, 2013, Volume: 24, Issue:4

    GDF-15 (also MIC-1, NAG-1, PLAB, PTGFB) is a member of the TGF-β superfamily, which is widely distributed in mammalian tissues and has been shown to play multiple roles in various pathologies, including inflammation, cancer, cardiovascular diseases, and obesity. GDF-15 serum levels are a highly reliable predictor of disease progression. Both the anti-tumorigenic potential of GDF-15 and its capacity to promote metastasis have been documented for a large variety of cancers, yet its opposing functions, which are typical for members of the TGF-β superfamily, have only partly been resolved on the molecular level. Knowledge on physiological functions in the non-diseased organism is scarce. In the nervous system GDF-15 knockout analyses have revealed that GDF-15 is essential for the postnatal maintenance of various neuron populations. When applied exogenously GDF-15 is a powerful factor for promoting survival of developing and lesioned neurons in vitro and in vivo. Receptor activation by GDF-15 has only been partially resolved.

    Topics: Animals; Cardiovascular Diseases; Gene Expression Regulation; Growth Differentiation Factor 15; Humans; Inflammation; Mice; Neoplasms; Obesity; Signal Transduction; Tissue Distribution; Transforming Growth Factor beta

2013
Smad phospho-isoforms direct context-dependent TGF-β signaling.
    Cytokine & growth factor reviews, 2013, Volume: 24, Issue:4

    Better understanding of TGF-β signaling has deepened our appreciation of normal epithelial cell homeostasis and its dysfunction in such human disorders as cancer and fibrosis. Smad proteins, which convey signals from TGF-β receptors to the nucleus, possess intermediate linker regions connecting Mad homology domains. Membrane-bound, cytoplasmic, and nuclear protein kinases differentially phosphorylate Smad2 and Smad3 to create C-tail (C), the linker (L), or dually (L/C) phosphorylated (p, phospho-) isoforms. According to domain-specific phosphorylation, distinct transcriptional responses, and selective metabolism, Smad phospho-isoform pathways can be grouped into 4 types: cytostatic pSmad3C signaling, mitogenic pSmad3L (Ser-213) signaling, invasive/fibrogenic pSmad2L (Ser-245/250/255)/C or pSmad3L (Ser-204)/C signaling, and mitogenic/migratory pSmad2/3L (Thr-220/179)/C signaling. We outline how responses to TGF-β change through the multiple Smad phospho-isoforms as normal epithelial cells mature from stem cells through progenitors to differentiated cells, and further reflect upon how constitutive Ras-activating mutants favor the Smad phospho-isoform pathway promoting tumor progression. Finally, clinical analyses of reversible Smad phospho-isoform signaling during human carcinogenesis could assess effectiveness of interventions aimed at reducing human cancer risk. Spatiotemporally separate, functionally different Smad phospho-isoforms have been identified in specific cells and tissues, answering long-standing questions about context-dependent TGF-β signaling.

    Topics: Animals; Cell Differentiation; Cell Transformation, Neoplastic; Epithelial Cells; Humans; Mice; Neoplasms; Phosphorylation; Protein Isoforms; Protein Kinases; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2013
[TGF-β signaling pathways in cancers].
    Postepy higieny i medycyny doswiadczalnej (Online), 2013, Sep-25, Volume: 67

    TGF-β is a multifunctional cytokine involved in growth, cell differentiation and maintenanceof tissue homeostasis. In addition, TGF-β plays a key role in the pathogenesis of many diseases, including cancer. TGF-β-induced signaling pathways have either tumor-suppression or tumor-promoting effects in a cancer-type-specific and stage-dependent manner. TGF-β at an early stage of cancer development induces signaling pathways involved in inhibitionof cell proliferation, induction of differentiation, apoptosis or autophagy, suppression of angiogenesis and inflammation. At a later stage of disease, TGF-β exerts metastasis-promoting activity associated with epithelial-to-mesenchymal transition, modulation of cancer microenvironment and extracellular matrix components, inflammation and immune suppression. Furthermore, the TGF-β pathways play a pivotal role in the maintenance of stem cell-like properties of tumor cells. The pleiotropic action of TGF-β during tumorigenesis depends on interactions with different signaling pathways, including Hedgehog, WNT, PI3K--AKT, NOTCH, INF-γ, TNF-α, and RAS-ERK.

    Topics: Apoptosis; Autophagy; Cell Transformation, Neoplastic; Cytokines; Epithelial-Mesenchymal Transition; Extracellular Matrix; Humans; Inflammation; Neoplasms; Neovascularization, Pathologic; Phosphatidylinositol 3-Kinases; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor-alpha

2013
The roles of TGFβ in the tumour microenvironment.
    Nature reviews. Cancer, 2013, Volume: 13, Issue:11

    The influence of the microenvironment on tumour progression is becoming clearer. In this Review we address the role of an essential signalling pathway, that of transforming growth factor-β, in the regulation of components of the tumour microenvironment and how this contributes to tumour progression.

    Topics: Adaptive Immunity; Animals; Cell Movement; Disease Progression; Epigenesis, Genetic; Fibroblasts; Gene Expression Regulation, Neoplastic; Humans; Immunity, Innate; Mice; Neoplasms; Phenotype; Signal Transduction; Stromal Cells; Transforming Growth Factor beta; Tumor Microenvironment

2013
[Crosstalk between canonical TGF-β/Smad and Wnt/β-catenin signaling pathway].
    Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences, 2013, Volume: 42, Issue:5

    TGF-β signaling pathway plays a central role in the signaling networks that control the growth, differentiation of the cell, and the initiation of fibrosis and cancer. Wnt signaling pathway is critical for the embryonic development and the invasion and migration of cancer cells. TGF-β signaling and Wnt signaling, both of which play an important role in regulating embryonic development, fibrotic disease and tumor progression, have a close relationship. Researches find several typical cross points between these two signaling systems, such as Smad, Axin, Dvl and β-catenin. In this review, we focus on the crosstalk between TGF-β signaling and Wnt signaling through these typical factors, intending to better understand the process of fibrosis and tumor progression.

    Topics: Fibrosis; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Wnt Proteins; Wnt Signaling Pathway

2013
The dual role of Smad7 in the control of cancer growth and metastasis.
    International journal of molecular sciences, 2013, Dec-05, Volume: 14, Issue:12

    Smad7 was initially identified as an inhibitor of Transforming growth factor (TGF)-β due mainly to its ability to bind TGF-β receptor type I and prevent TGF-β-associated Smad signaling. More recently, it has been demonstrated that Smad7 can interact with other intracellular proteins and regulate also TGF-β-independent signaling pathways thus making a valid contribution to the neoplastic processes in various organs. In particular, data emerging from experimental studies indicate that Smad7 may differently modulate the course of various tumors depending on the context analyzed. These observations, together with the demonstration that Smad7 expression is deregulated in many cancers, suggest that therapeutic interventions around Smad7 can help interfere with the development/progression of human cancers. In this article we review and discuss the available data supporting the role of Smad7 in the modulation of cancer growth and progression.

    Topics: Humans; Neoplasm Metastasis; Neoplasms; Signal Transduction; Smad7 Protein; Transforming Growth Factor beta

2013
The conditional role of inflammation in pregnancy and cancer.
    Clinical nutrition (Edinburgh, Scotland), 2013, Volume: 32, Issue:3

    Cancer growth is characterized by proliferation of tumor cells in conjunction with invasion of all different immune cells that also invade healing wounds. This inflammatory response is necessary for cell proliferation but a second purpose of the inflammatory process is so that a low Th1/Th2 ratio is present with overexpression of IL-10, TGF-β and IFN-γ. Down regulation of NO activity also shifts the balance between M1 and M2 macrophages. Both aspects allow the antigenous nature of the tumor to escape anti-tumor effects of the host. Support for this view comes from observations in pregnancy in which the placenta exhibits identical immune responses and downregulation of NO production to allow trophoblast cells to invade the uterine tissues without being rejected. Cell proliferation requires a metabolic set-up in which the organism produces adequate substrate for growth. This also bears the characteristics of a systemic inflammatory response delivering a similar substrate mix required for cancer and fetal growth. This arrangement is clearly beneficial in pregnancy and therefore supports the view that cancer growth is facilitated by the organism: the cancerous tumor elicits an immunological response opposing anti-tumor effects and induces the host to produce building blocks for growth.

    Topics: Cell Proliferation; Down-Regulation; Female; Humans; Inflammation; Interferon-gamma; Interleukin-10; Neoplasms; Nitric Oxide; Pregnancy; Th1-Th2 Balance; Transforming Growth Factor beta; Trophoblasts

2013
Cutl1: a potential target for cancer therapy.
    Cellular signalling, 2013, Volume: 25, Issue:1

    CDP, a key transcription regulator encoded by Cutl1 gene, has been demonstrated to be involved in repressing or promoting expression of target genes through its specific DNA-binding, meanwhile, the activity of CDP was influenced by some types of modifications including transcriptional, posttranscriptional, translational and posttranslational modifications. In this review, we systematically analyzed the role of CDP in normal development and tumor progression, and then emphasized its interactors and downstream molecules. Eventually, we concluded that Cut1 could promote cancer progression and its down-regulating expression will be a promising strategy for cancer therapy.

    Topics: Disease Progression; Homeodomain Proteins; Humans; Neoplasms; Nuclear Proteins; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Repressor Proteins; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2013
RUNX family: Regulation and diversification of roles through interacting proteins.
    International journal of cancer, 2013, Mar-15, Volume: 132, Issue:6

    The Runt-related transcription factors (RUNX) belong to an ancient family of metazoan genes involved in developmental processes. Through multiple protein-interacting partners, RUNX proteins have been implicated in diverse signaling pathways and cellular processes. The frequent inactivation of RUNX genes in cancer indicates crucial roles for RUNX in tumor suppression. This review discusses the abilities of RUNX proteins, in particular RUNX3, to integrate oncogenic signals or environmental cues and to initiate appropriate tumor suppressive responses.

    Topics: Animals; Cell Cycle; Core Binding Factor Alpha 2 Subunit; Core Binding Factor Alpha 3 Subunit; Core Binding Factor alpha Subunits; Humans; Mutation; Neoplasms; Protein Processing, Post-Translational; Protein Serine-Threonine Kinases; Serine-Threonine Kinase 3; Signal Transduction; Transcription, Genetic; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Wnt Signaling Pathway

2013
TGF-β signaling and epithelial-mesenchymal transition in cancer progression.
    Current opinion in oncology, 2013, Volume: 25, Issue:1

    TGF-β acts as a potent driver of cancer progression through the induction of epithelial-mesenchymal transition (EMT), in which epithelial cells acquire mesenchymal phenotype, leading to enhanced motility and invasion. Recent reports highlight the fundamental roles of TGF-β-induced EMT in multiple aspects of cancer progression. In this review, we focus on the novel insights into the roles of TGF-β-induced EMT in cancer progression and the underlying mechanisms that enable TGF-β to activate this epithelial plasticity response at transcription, translation, and posttranslational levels.. Smad-mediated transcription regulation is known to activate TGF-β-induced EMT. More recently, novel mechanisms of epigenetic control, alternative splicing, miRNAs, translation control, and posttranslational modifications have been shown to play key roles in the control of EMT. In addition to initiating carcinoma cell invasion, TGF-β-induced EMT can guide cancer cells to de-differentiate and gain cancer stem-cell-like properties. EMT also allows the generation of stromal cells that support and instruct cancer progression.. The differentiation plasticity of epithelial cells that mediates TGF-β-induced EMT and reversion from mesenchymal to epithelial phenotype are increasingly seen as integral aspects of cancer progression that contribute to survival and dissemination of cancer cells. Further mechanistic insights under physiological conditions may lead to new therapeutic or prognostic strategies in cancer treatment.

    Topics: Animals; Cell Transformation, Neoplastic; Disease Progression; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Neoplastic Stem Cells; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2013
IDO⁺ DCs and signalling pathways.
    Current cancer drug targets, 2013, Volume: 13, Issue:3

    Dendritic cells (DCs) have traditionally been viewed as constituting an 'information management' system that functions solely to integrate a diverse array of incoming signals, in order to induce immune reactivity. In recent years, however, there has been a shift towards viewing these cells as key regulators in the orchestration of immunological tolerance, with increasing recognition that they are capable of suppressing T-cell responses depending on signalling processes and localised biochemical conditions. Indoleamine 2,3-dioxygenase (IDO) competent (IDO⁺) DCs are a subset of human DCs which are programmed to a tolerogenic state and play a vital role in establishing and maintaining a tumour-suppressing milieu. The expression of IDO in these DCs represents a key mechanism responsible for inducing the tolerogenic state. However, the mechanisms by which IDO becomes dysregulated in this subset of DCs have not yet been described. In this review, the function of IDO⁺ DCs within the cancer-tolerogenic milieu, as well as the signals responsible for expression of IDO in this subset, will be discussed.

    Topics: Animals; Antineoplastic Agents; Cell Transformation, Neoplastic; CTLA-4 Antigen; Dendritic Cells; Enzyme Inhibitors; Humans; Immune Tolerance; Immunologic Surveillance; Indoleamine-Pyrrole 2,3,-Dioxygenase; Neoplasm Proteins; Neoplasms; NF-kappa B; Protein Isoforms; Signal Transduction; STAT Transcription Factors; Transforming Growth Factor beta; Tumor Microenvironment

2013
Fibroblast-mediated drug resistance in cancer.
    Biochemical pharmacology, 2013, Apr-15, Volume: 85, Issue:8

    Tumor progression relies upon the dynamic interaction of cancer cells with host fibroblasts, endothelial cells, immune cells and components of the extracellular matrix, collectively known as the tumor microenvironment. Despite this, relatively little is known about how normal host cells dictate the response of tumors to anti-cancer therapies. Emerging data suggests that host factors play a critical role in determining risks for tumor progression and decreased therapeutic responses. In particular, recent findings have provided evidence that the tumor microenvironment provides a protective niche that allows minor populations of cancer cells to escape from the cytotoxic effects of radiation, chemotherapy and targeted therapies. In this review we will outline the mechanisms by which tumor cells and host fibroblasts co-operate to drive tumor initiation and progression. In particular, we will focus upon the mechanisms by which tumor cells exposed to targeted therapies co-opt the host leading to therapeutic escape and resistance. We will end by discussing the idea that long-term responses to targeted anticancer therapies will only be achieved through strategies that target both the tumor and host.

    Topics: Animals; Disease Progression; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Fibroblasts; Humans; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment; Wound Healing

2013
Role of Smads in TGFβ signaling.
    Cell and tissue research, 2012, Volume: 347, Issue:1

    Transforming growth factor-β (TGFβ) is the prototype for a large family of pleiotropic factors that signal via heterotetrameric complexes of type I and type II serine/threonine kinase receptors. Important intracellular mediators of TGFβ signaling are members of the Smad family. Smad2 and 3 are activated by C-terminal receptor-mediated phosphorylation, whereafter they form complexes with Smad4 and are translocated to the nucleus where they, in cooperation with other transcription factors, co-activators and co-repressors, regulate the transcription of specific genes. Smads have key roles in exerting TGFβ-induced programs leading to cell growth arrest and epithelial-mesenchymal transition. The activity and stability of Smad molecules are carefully regulated by a plethora of post-translational modifications, including phosphorylation, ubiquitination, sumoylation, acetylation and poly(ADP)-ribosylation. The Smad function has been shown to be perturbed in certain diseases such as cancer.

    Topics: Active Transport, Cell Nucleus; Animals; Epithelial-Mesenchymal Transition; Humans; MicroRNAs; Neoplasms; Protein Isoforms; Protein Processing, Post-Translational; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2012
Deconstructing the mechanisms and consequences of TGF-β-induced EMT during cancer progression.
    Cell and tissue research, 2012, Volume: 347, Issue:1

    Transforming growth factor-β (TGF-β) is a potent pleiotropic cytokine that regulates mammalian development, differentiation, and homeostasis in essentially all cell types and tissues. TGF-β normally exerts anticancer activities by prohibiting cell proliferation and by creating cell microenvironments that inhibit cell motility, invasion, and metastasis. However, accumulating evidence indicates that the process of tumorigenesis, particularly that associated with metastatic progression, confers TGF-β with oncogenic activities, a functional switch known as the "TGF-β paradox." The molecular determinants governing the TGF-β paradox are complex and represent an intense area of investigation by researchers in academic and industrial settings. Recent findings link genetic and epigenetic events in mediating the acquisition of oncogenic activity by TGF-β, as do aberrant alterations within tumor microenvironments. These events coalesce to enable TGF-β to direct metastatic progression via the stimulation of epithelial-mesenchymal transition (EMT), which permits carcinoma cells to abandon polarized epithelial phenotypes in favor of apolar mesenchymal-like phenotypes. Attempts to deconstruct the EMT process induced by TGF-β have identified numerous signaling molecules, transcription factors, and microRNAs operant in mediating the initiation and resolution of this complex transdifferentiation event. In addition to its ability to enhance carcinoma cell invasion and metastasis, EMT also endows transitioned cells with stem-like properties, including the acquisition of self-renewal and tumor-initiating capabilities coupled to chemoresistance. Here, we review recent findings that delineate the pathophysiological mechanisms whereby EMT stimulated by TGF-β promotes metastatic progression and disease recurrence in human carcinomas.

    Topics: Animals; Disease Progression; Epithelial-Mesenchymal Transition; Gene Expression Regulation; Humans; MicroRNAs; Neoplasm Metastasis; Neoplasms; Neoplastic Stem Cells; Signal Transduction; Transforming Growth Factor beta

2012
Regulation of endothelial cell plasticity by TGF-β.
    Cell and tissue research, 2012, Volume: 347, Issue:1

    Recent evidence has demonstrated that endothelial cells can have a remarkable plasticity. By a process called Endothelial-to-Mesenchymal Transition (EndMT) endothelial cells convert to a more mesenchymal cell type that can give rise to cells such as fibroblasts, but also bone cells. EndMT is essential during embryonic development and tissue regeneration. Interestingly, it also plays a role in pathological conditions like fibrosis of organs such as the heart and kidney. In addition, EndMT contributes to the generation of cancer associated fibroblasts that are known to influence the tumor-microenvironment favorable for the tumor cells. EndMT is a form of the more widely known and studied Epithelial-to-Mesenchymal Transition (EMT). Like EMT, EndMT can be induced by transforming growth factor (TGF)-β. Indeed many studies have pointed to the important role of TGF-β receptor/Smad signaling and downstream targets, such as Snail transcriptional repressor in EndMT. By selective targeting of TGF-β receptor signaling pathological EndMT may be inhibited for the therapeutic benefit of patients with cancer and fibrosis.

    Topics: Animals; Biomarkers; Cell Differentiation; Cell Transdifferentiation; Endothelial Cells; Fibrosis; Humans; Mesoderm; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2012
aV integrins and TGF-β-induced EMT: a circle of regulation.
    Journal of cellular and molecular medicine, 2012, Volume: 16, Issue:3

    Transforming growth factor-β (TGF-β) has roles in embryonic development, the prevention of inappropriate inflammation and tumour suppression. However, TGF-β signalling also regulates pathological epithelial-to-mesenchymal transition (EMT), inducing or progressing a number of diseases ranging from inflammatory disorders, to fibrosis and cancer. However, TGF-β signalling does not proceed linearly but rather induces a complex network of cascades that mutually influence each other and cross-talk with other pathways to successfully induce EMT. Particularly, there is substantial evidence for cross-talk between αV integrins and TGF-β during EMT, and anti-integrin therapeutics are under development as treatments for TGF-β-related disorders. However, TGF-β's complex signalling network makes the development of therapeutics to block TGF-β-mediated pathology challenging. Moreover, despite our current understanding of integrins and TGF-β function during EMT, the precise mechanism of their role during physiological versus pathological EMT is not fully understood. This review focuses on the circle of regulation between αV integrin and TGF-β signalling during TGF-β induced EMT, which pose as a significant driver to many known TGF-β-mediated disorders.

    Topics: Animals; Cell Adhesion; Epithelial-Mesenchymal Transition; Extracellular Matrix; Fibrosis; Gene Expression Regulation; Genes, Regulator; Humans; Inflammation; Integrin alphaV; Neoplasms; Protein Isoforms; Signal Transduction; Transforming Growth Factor beta

2012
Regulatory T cell: a protection for tumour cells.
    Journal of cellular and molecular medicine, 2012, Volume: 16, Issue:3

    Characterized by immunosuppression regulatory T cells (Tregs) play a key role in maintaining immune tolerance. A growing number of tumours have been found with Tregs accumulating in microenvironment and patients with high density of Tregs in tumour stroma get a worse prognosis, which suggests that Tregs may inhibit anti-tumour immunity in stroma, resulting in a poor prognosis. In this paper, we demonstrate the accumulation of Tregs in tumour stroma and the possible suppressive mechanisms. We also state the immunotherapy that has being used in animal and clinical trials.

    Topics: CD8-Positive T-Lymphocytes; Dendritic Cells; Humans; Immune Tolerance; Immunotherapy; Interleukin-10; Killer Cells, Natural; Neoplasm Metastasis; Neoplasms; Signal Transduction; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Escape; Tumor Microenvironment

2012
WWP1: a versatile ubiquitin E3 ligase in signaling and diseases.
    Cellular and molecular life sciences : CMLS, 2012, Volume: 69, Issue:9

    WW domain-containing E3 ubiquitin protein ligase 1 (WWP1) is a multifunction protein containing an N-terminal C2 domain, four tandem WW domains for substrate binding, and a C-terminal catalytic HECT domain for ubiquitin transferring. WWP1 has been suggested to function as the E3 ligase for several PY motif-containing proteins, such as Smad2, KLF5, p63, ErbB4/HER4, RUNX2, JunB, RNF11, SPG20, and Gag, as well as several non-PY motif containing proteins, such as TβR1, Smad4, KLF2, and EPS15. WWP1 regulates a variety of cellular biological processes including protein trafficking and degradation, signaling, transcription, and viral budding. WWP1 has been implicated in several diseases, such as cancers, infectious diseases, neurological diseases, and aging. In this review article, we extensively summarize the current knowledge of WWP1 with special emphasis on the roles and action of mechanism of WWP1 in signaling and human diseases.

    Topics: Aging; ErbB Receptors; Gene Expression; Humans; Infections; Kruppel-Like Transcription Factors; Neoplasms; Nervous System Diseases; Protein Structure, Tertiary; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Ubiquitin-Protein Ligases; Ubiquitination

2012
Tumor-promoting functions of transforming growth factor-β in progression of cancer.
    Upsala journal of medical sciences, 2012, Volume: 117, Issue:2

    Transforming growth factor-β (TGF-β) elicits both tumor-suppressive and tumor-promoting functions during cancer progression. Here, we describe the tumor-promoting functions of TGF-β and how these functions play a role in cancer progression. Normal epithelial cells undergo epithelial-mesenchymal transition (EMT) through the action of TGF-β, while treatment with TGF-β and fibroblast growth factor (FGF)-2 results in transdifferentiation into activated fibroblastic cells that are highly migratory, thereby facilitating cancer invasion and metastasis. TGF-β also induces EMT in tumor cells, which can be regulated by oncogenic and anti-oncogenic signals. In addition to EMT promotion, invasion and metastasis of cancer are facilitated by TGF-β through other mechanisms, such as regulation of cell survival, angiogenesis, and vascular integrity, and interaction with the tumor microenvironment. TGF-β also plays a critical role in regulating the cancer-initiating properties of certain types of cells, including glioma-initiating cells. These findings thus may be useful for establishing treatment strategies for advanced cancer by inhibiting TGF-β signaling.

    Topics: Animals; Cell Differentiation; Disease Progression; Epithelial-Mesenchymal Transition; Fibroblast Growth Factor 2; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2012
LIM-domain proteins in transforming growth factor β-induced epithelial-to-mesenchymal transition and myofibroblast differentiation.
    Cellular signalling, 2012, Volume: 24, Issue:4

    Epithelial to mesenchymal transition (EMT) is a process during which junctions of the cell-cell contacts are dissolved, actin cytoskeleton is deformed, apical-basolateral cell polarity is lost and cell motility is increased. EMT is needed during normal embryonal development and wound healing, but may also lead to pathogenic transformation and formation of myofibroblasts. Transforming growth factor β (TGFβ) is a multifunctional cytokine promoting EMT and myofibroblast differentiation, and its dysregulation is involved in pathological disorders like cancer and fibrosis. Lin11, Isl-1 and Mec-3 (LIM) domain proteins are associated with actin cytoskeleton and linked to regulation of cell growth, damage signaling, cell fate determination and signal transduction. LIM-domain proteins generally do not bind DNA, but are more likely to function via protein-protein interactions. Despite being a disparate group of proteins, similarities in their functions are observed. In this review we will discuss the role of LIM-domain proteins in TGFβ-signaling pathway and in EMT-driven processes. LIM-domain proteins regulate TGFβ-induced actin cytoskeleton reorganization, motility and adhesion, but also dissolution of cell-cell junctions during EMT. Finally, the role of LIM-domain proteins in myofibroblasts found in fibrotic foci and tumor stroma will be discussed.

    Topics: Animals; Cell Adhesion; Cell Dedifferentiation; Cell Differentiation; Cell Movement; Epithelial Cells; Epithelial-Mesenchymal Transition; Gene Expression Regulation; Humans; Idiopathic Pulmonary Fibrosis; Intercellular Junctions; LIM Domain Proteins; Mice; Mice, Knockout; Myofibroblasts; Neoplasms; Phosphorylation; Protein Binding; Signal Transduction; Transforming Growth Factor beta; Wound Healing

2012
Alterations in the Smad pathway in human cancers.
    Frontiers in bioscience (Landmark edition), 2012, 01-01, Volume: 17, Issue:4

    Members of the TGF-beta superfamily exhibit various biological activities, and perturbations of their signaling are linked to certain clinical disorders including cancer. The role of TGF-beta signaling as a tumor suppressor pathway is best illustrated by the presence of inactivating mutations in genes encoding TGF-beta receptors and Smads in human carcinomas. This perspective is further supported by studies of tumor development in mouse models after modulation of receptors and Smads. TGF-beta also controls processes such as cell invasion, immune regulation, and microenvironment alterations that cancer cells may exploit to their advantage for their progression. Consequently, the output of a TGF-beta response is highly situation dependent, across different tissues, and also in cancer in general. Understanding the mechanisms of TGF-beta superfamily signaling is thus important for the development of new ways to treat various types of cancer. This review focuses on recent advances in understanding the Smad dependent TGF-beta pathway as it relates to human carcinogenesis.

    Topics: Animals; Humans; Mice; Models, Molecular; Neoplasms; Smad Proteins; Transforming Growth Factor beta; Up-Regulation

2012
TGF-beta antiproliferative effects in tumor suppression.
    Frontiers in bioscience (Scholar edition), 2012, 01-01, Volume: 4, Issue:2

    The TGF-beta signaling pathway controls multiple functions of cancer cells and the surrounding stromal tissue. Some TGF-beta actions suppress cancer formation, while others contribute to tumor progression. Evidence supporting a tumor suppressive role for the TGF-beta/Smad signaling axis is presented here. These data are compiled from cell culture studies, animal models, analyses of human tumors, and investigations of polymorphisms of TGF-beta pathway components and their associated cancer risk. Therapeutic strategies for cancer treatment involving either restoring or potentiating TGF-beta tumor suppressive activities, or blocking TGF-beta tumor promoting functions are considered.

    Topics: Animals; Cell Growth Processes; Humans; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2012
A portrait of Transforming Growth Factor β superfamily signalling: Background matters.
    The international journal of biochemistry & cell biology, 2012, Volume: 44, Issue:3

    Ligands of the Transforming Growth Factor β superfamily like Transforming Growth Factor β and Bone Morphogenetic Proteins govern developmental processes and regulate adult homeostasis by controlling cellular proliferation, survival, differentiation and migration. Aberrant signalling activity is associated with human disorders such as cancer, cardiovascular, musculoskeletal, or fibrotic disease. Upon binding to specific sets of cognate cell surface receptors, family members induce highly similar pathways which include canonical SMAD dependent signalling as well as pathways without direct involvement of SMAD proteins, which activate signalling molecules like mitogen-activated protein kinases or small GTPases. The diverse ligand functionalities are achieved through regulation and modulation of the pathways at all levels, resulting in a highly quantitative and context sensitive signal integration reflecting the cellular state and background. Strategies to target Transforming Growth Factor β or Bone Morphogenetic Protein pathways have been developed on the basis of our current understanding and have proven a highly beneficial potential.

    Topics: Animals; Bone Morphogenetic Proteins; Cardiovascular Diseases; Cellular Microenvironment; Gene Expression Regulation; GTP Phosphohydrolases; Humans; Mitogen-Activated Protein Kinases; Musculoskeletal Diseases; Neoplasms; Receptor Cross-Talk; Signal Transduction; Smad Proteins; Transcriptional Activation; Transforming Growth Factor beta

2012
Cripto/GRP78 modulation of the TGF-β pathway in development and oncogenesis.
    FEBS letters, 2012, Jul-04, Volume: 586, Issue:14

    Cripto is a small, GPI-anchored signaling protein that regulates cellular survival, proliferation, differentiation and migration during normal developmental processes and tumorigenesis. Cripto functions as an obligatory co-receptor for the TGF-β ligands Nodal, GDF1 and GDF3 but attenuates signaling of others such as activin-A, activin-B and TGF-β1. Soluble, secreted forms of Cripto also activate Src, ras/raf/MAPK and PI3K/Akt pathways via a mechanism that remains largely obscure. This review describes the biological roles and signaling mechanisms of Cripto, highlighting our identification of the 78 kDa glucose regulated protein (GRP78) as a cell surface receptor/co-factor required for Cripto signaling via both TGF-β and Src/MAPK/PI3K pathways. We discuss emerging evidence indicating that Cripto/GRP78 signaling regulates normal somatic stem cells and their tumorigenic counterparts.

    Topics: Animals; Cell Membrane; Endoplasmic Reticulum Chaperone BiP; Gene Expression Regulation, Developmental; Gene Expression Regulation, Neoplastic; GPI-Linked Proteins; Heat-Shock Proteins; Humans; Intercellular Signaling Peptides and Proteins; Mice; Models, Biological; Neoplasm Proteins; Neoplasms; Protein Structure, Tertiary; Signal Transduction; Stem Cells; Transforming Growth Factor beta

2012
Inhibition of TGFβ signaling and its implications in anticancer treatments.
    Experimental oncology, 2012, Volume: 34, Issue:1

    The transforming growth factor-β (TGFβ) is a potent regulator of tumorigenesis. In cancer, two distinctive behaviors of TGFβ have been reported as a tumor suppressor at early stage of the disease, and as a tumor promoter at later stages. The past decades, the dualistic role of TGFβ has garnered a lot of attention. As a result, cancer researchers' has been tasked to elucidate how TGFβ signaling may lead to metastatic dissemination, how to tackle carcinogenesis and which therapeutic strategies should be adopted. Consequently, TGFβ signaling pathways have been considered as appropriate targets for cancer therapy. The TGFβ therapeutic strategies have emerged at three levels: ligand, ligand-receptor interaction and intracellular signaling level. Promising inhibitors of TGFβ signaling have entered clinical trials and shown encouraging results. Here we review the three strategies of TGFβ signa-ling inhibition and theirs applications in treatment of cancer.

    Topics: Animals; Antineoplastic Agents; Humans; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2012
Induction of epithelial-mesenchymal transition by transforming growth factor β.
    Seminars in cancer biology, 2012, Volume: 22, Issue:5-6

    Transforming growth factor β (TGFβ) is implicated in human malignancy. Tumors may escape the tumor suppressor activity of TGFβ by mutating some of its signaling components. Carcinoma and stromal cells produce high amounts of TGFβ which promotes epithelial-mesenchymal transition (EMT), tumor cell invasiveness and tumor angiogenesis, while suppressing immune responses against the tumor. Thus, TGFβ has tumor suppressive as well as tumor promoting effects supporting metastasis. TGFβ elicits the EMT response by activating complementary signaling cascades that mobilize embryonic transcription factors that reprogram the epithelial cell so that it acquires both progenitor-like, pro-motility and mesenchymal features. Such nuclear reprogramming of carcinoma cells involves epigenetic and transcriptional regulation, the activity of miRNAs, and modulation of RNA splicing and mRNA translation, leading to the expression of key intracellular and membrane proteins together with a large pool of secreted factors that mediate and account for the phenotypic changes that accompany EMT.

    Topics: Animals; Epithelial-Mesenchymal Transition; Extracellular Matrix; Gene Regulatory Networks; Humans; Membrane Proteins; MicroRNAs; Neoplasm Metastasis; Neoplasms; Neoplastic Stem Cells; Protein Biosynthesis; RNA Splicing; Signal Transduction; Transforming Growth Factor beta

2012
Releasing the brake: targeting Cbl-b to enhance lymphocyte effector functions.
    Clinical & developmental immunology, 2012, Volume: 2012

    The E3 ubiquitin ligase Cbl-b is an established nonredundant negative regulator of T-cell activation. Cbl-b fine-tunes the activation threshold of T cells and uncouples T cells from their vital need of a costimulatory signal to mount a productive immune response. Accordingly, mice deficient in cblb are prone to autoimmunity and reject tumors. The latter has been described to be mediated via CD8(+) T cells, which are hyperactive and more abundant in shrinking tumors of cblb-deficient animals. This might at least also in part be mediated by resistance of cblb-deficient T cells to negative cues exerted by tumor-associated immuno-suppressive factors, such as TGF-β and regulatory T cells (Treg). Experiments using cblb-deficient T cells either alone or in combination with vaccines validate the therapeutic concept of enhancing the efficacy of adoptively transferred lymphocytes to treat malignant tumors. This paper summarizes the current knowledge about the negative regulatory role of Cbl-b in T-cell activation and its potential therapeutic implications for cancer immunotherapy.

    Topics: Adoptive Transfer; Animals; Cancer Vaccines; CD8-Positive T-Lymphocytes; Humans; Immunotherapy; Lymphocyte Activation; Mice; Mice, Knockout; Neoplasms; Proto-Oncogene Proteins c-cbl; Signal Transduction; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2012
Transforming growth factor (TGF)-β expression and activation mechanisms as potential targets for anti-tumor therapy and tumor imaging.
    Pharmacology & therapeutics, 2012, Volume: 135, Issue:2

    Cancer remains one of the leading causes of death in the developed countries and cancer mortality is expected to rise globally. Despite encouraging developments regarding targeted drugs, the most prevalent cancer mortality remains metastatic disease. Therefore, drugs that target cancer progression, invasion and metastasis are clearly needed. One of the most interesting targets in this setting is transforming growth factor β (TGF-β). TGF-β can promote tumor growth, invasion and metastasis. However, TGF-β also has a physiological, opposing role: maintaining tissue homeostasis and suppression of tumor progression. The window of effective TGF-β targeting is therefore evidently small, which poses a clear challenge in selecting patients at the right time. Despite this complexity, several TGF-β inhibitors are currently in clinical development, modulating TGF-β production, activation or signaling. Still, specificity and long term toxicity remain unclear, emphasizing the importance of careful monitoring of clinical trials. Development and application of these drugs in the clinic require adequate insight and evaluation methods for the role of TGF-β during tumor invasion and metastasis. In this review, presently available methods for clinical evaluation will be discussed, such as an ex vivo stimulation assay, TGF-β response signature and molecular imaging techniques. Future clinical trials incorporating the validation of these evaluation methods will show which method will be most predictive and suitable for clinical application.

    Topics: Animals; Diagnostic Imaging; Humans; Neoplasms; Transforming Growth Factor beta

2012
Interaction between regulatory T cells and cancer stem cells.
    International journal of cancer, 2012, Oct-01, Volume: 131, Issue:7

    The concept that cancer stem cells (CSCs)/tumor stem cells/cancer-initiating cells are one of the key centers to cure neoplastic disease has drawn an increasing attention recent years. Because of their high resistance and potential to initiate tumors, CSCs are considered a critical factor associated with tumor relapse. Regulatory T(T(reg)) cells, a group of immune cells with tumor-promoting effect, exert their function through inhibition of effector T cells and regulation of the tumor microenvironment by producing a series of soluble factors. However, the interactions between T(reg)s and CSCs are less understood. The mechanisms of how T(reg)s, as tumor-promoting cells, manipulate CSCs remain obscure. In this review, we elucidate their interactions.

    Topics: Animals; Cell Communication; Epithelial-Mesenchymal Transition; Forkhead Transcription Factors; Humans; Macrophages; Neoplasms; Neoplastic Stem Cells; Neovascularization, Pathologic; T-Lymphocytes, Regulatory; Th17 Cells; Transforming Growth Factor beta

2012
Crosstalk between TGF-β and hedgehog signaling in cancer.
    FEBS letters, 2012, Jul-04, Volume: 586, Issue:14

    Hedgehog (HH) and TGF-β signals control various aspects of embryonic development and cancer progression. While their canonical signal transduction cascades have been well characterized, there is increasing evidence that these pathways are able to exert overlapping activities that challenge efficient therapeutic targeting. We herein review the current knowledge on HH signaling and summarize the recent findings on the crosstalks between the HH and TGF-β pathways in cancer.

    Topics: Animals; Epithelial-Mesenchymal Transition; Fibrosis; Hedgehog Proteins; Humans; Kruppel-Like Transcription Factors; Ligands; Mice; Models, Biological; Neoplasms; Nuclear Proteins; Oncogene Proteins; Rats; RNA Processing, Post-Transcriptional; Signal Transduction; Trans-Activators; Transforming Growth Factor beta; Zinc Finger Protein GLI1; Zinc Finger Protein Gli2

2012
TGF-β-induced epithelial-mesenchymal transition: a link between cancer and inflammation.
    Seminars in cancer biology, 2012, Volume: 22, Issue:5-6

    Metastatic spread of tumor cells to vital organs is the major cause of death in cancer. Accumulating data support an important role of infiltrating immune cells in promoting carcinoma progression into metastatic disease. Tumor-infiltrating immune cells produce and secrete cytokines, growth factors and proteases that re-activate latent developmental processes including epithelial-mesenchymal transition (EMT). EMT provides tumor cells with invasive, migratory and stem cell properties allowing them to disseminate and propagate at distant sites. Induction of EMT requires two criteria to be fulfilled: (i) cells are competent to undergo EMT (ii) an EMT-permissive microenvironment exists. The cytokine TGF-β, which is expressed by tumor-infiltrating immune cells, stands out as a master regulator of the pro-invasive tumor microenvironment. TGF-β cooperates with stem cell pathways, such as Wnt and Ras signaling, to induce EMT. In addition, TGF-β contributes to an EMT-permissive microenvironment by switching the phenotypes of tumor-infiltrating immune cells, which thereby mount pro-invasive and pro-metastatic immune responses. In this review, we discuss the role of TGF-β-induced EMT as a link between cancer and inflammation in the context of questions, which from our point of view are key to answer in order to understand the functionality of EMT in tumors.

    Topics: Animals; Cell Movement; Cytokines; Epithelial-Mesenchymal Transition; Humans; Inflammation; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2012
Context-dependent action of transforming growth factor β family members on normal and cancer stem cells.
    Current pharmaceutical design, 2012, Volume: 18, Issue:27

    The transforming growth factor β (TGFβ) family embraces many growth factors including the Activins and bone morphogenetic proteins (BMPs). The pathways mediated by these growth factors are implicated in many fundamental biological processes such as early embryonic development, organ morphogenesis and adult tissue homeostasis and in a large number of pathologies including cancer. The action of these pathways is often contextual, which means that different cell types present different physiological responses to these ligands or that the response of one cell type to a certain ligand differs depending on the presence of other signaling proteins that stimulate the target cell together with TGFβ/BMP. The latter usually reflects developmental stage or progression to a specific pathological stage. Not only diverse growth factors and cytokines can influence the response of tissues to TGFβ/BMP, but a single cell type may also show drastically different physiological outcomes to TGFβ or Activin signaling as compared to BMP signaling. This review describes differential physiological outcomes of TGFβ and BMP signaling in normal embryonic or adult stem cells and eventually in cancer stem cells and the process of epithelial-mesenchymal transition. We also summarize evidence on the mechanistic antagonism between TGFβ and BMP signaling as established in vascular differentiation and the progression of tissue fibrosis and cancer. The article ends by discussing possible advantages that the acquired knowledge of these signaling mechanisms offers to new regimes of cancer therapy and the ever-lasting problem of drug resistance elicited by tumor initiating cells.

    Topics: Activins; Adult; Adult Stem Cells; Animals; Antineoplastic Agents; Bone Morphogenetic Proteins; Disease Progression; Drug Resistance, Neoplasm; Embryonic Stem Cells; Humans; Neoplasms; Neoplastic Stem Cells; Signal Transduction; Transforming Growth Factor beta

2012
TGF-beta: a master switch in tumor immunity.
    Current pharmaceutical design, 2012, Volume: 18, Issue:27

    The capacity of the immune system to distinguish foreign from self-antigen, and to subsequently eliminate the threat of disease without injuring the host is crucial for survival. It also serves to defend against tumor formation and progression via a process termed cancer immunosurveillance. Innate and adaptive immune cell types and effector molecules collectively function as extrinsic tumorsuppressor mechanisms. However, tumors may escape immunesurveillance through a variety of mechanisms that create a local microenvironment that is unfavorable for effective tumor immunity. Transforming growth factor β (TGF-β) has pleiotropic effects on the immune system, and is recognized as one of the most potent immunosuppressive agents in facilitating oncogenesis. The TGF-β pathway promotes cancer progression by concomitantly enhancing tumor metastases while inhibiting the protective host immunity. In this review, we discuss mechanisms through which TGF-β interferes with the development of an anti-tumor immunity and potential means through which to circumvent its activity in order to define more effective cancer immunotherapies.

    Topics: Adaptive Immunity; Animals; Disease Progression; Humans; Immunity, Innate; Immunologic Surveillance; Neoplasm Metastasis; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2012
Regulation of EMT by TGFβ in cancer.
    FEBS letters, 2012, Jul-04, Volume: 586, Issue:14

    Transforming growth factor-β (TGFβ) suppresses tumor formation since it inhibits cell growth and promotes apoptosis. However, in advanced cancers TGFβ elicits tumor promoting effects through its ability to induce epithelial-mesenchymal transition (EMT) which enhances invasiveness and metastasis; in addition, TGFβ exerts tumor promoting effects on non-malignant cells of the tumor, including suppression of immune surveillance and stimulation of angiogenesis. TGFβ promotes EMT by transcriptional and posttranscriptional regulation of a group of transcription factors that suppresses epithelial features, such as expression of components of cell junctions and polarity complexes, and enhances mesenchymal features, such as production of matrix molecules and several cytokines and growth factors that stimulate cell migration. The EMT program has certain similarities with the stem cell program. Inducers and effectors of EMT are interesting targets for the development of improved diagnosis, prognosis and therapy of cancer.

    Topics: Animals; Cell Adhesion; Cell Nucleus; Epigenesis, Genetic; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Inflammation; MAP Kinase Signaling System; Mice; Models, Biological; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neoplastic Stem Cells; Platelet-Derived Growth Factor; RNA Processing, Post-Transcriptional; RNA Splicing; Signal Transduction; Transcription, Genetic; Transforming Growth Factor beta

2012
Molecular pathways: targeting the TGF-β pathway for cancer therapy.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2012, Sep-01, Volume: 18, Issue:17

    TGF-β is a ubiquitous cytokine that plays an active role in many cellular processes. Nearly every cell type has the ability to secrete TGF-β, as well as the ability to respond to TGF-β via the presence of TGF-β receptors on the cell surface. Consequently, gain or loss of function of the TGF-β pathway and its components are known to lead to a variety of diseases, including cancer. In epithelial cells, TGF-β functions as a tumor suppressor, where it inhibits proliferation, induces apoptosis, and mediates differentiation. Conversely, in other contexts, TGF-β promotes tumor progression through increasing tumor cell invasion and metastasis. Thus, TGF-β can have opposing roles, likely dependent, in part, on whether the cancer is early or late stage. The effects of TGF-β on tumor suppression and promotion are not limited to the tumor cell itself; rather, these effects can also be mediated through the stroma and the immune system. The dichotomous role of TGF-β in cancer highlights our need to understand the contextual effects of this cytokine to better guide patient selection for the use of anti-TGF-β therapies currently in clinical trials.

    Topics: Apoptosis; Cell Proliferation; Clinical Trials as Topic; Epithelial Cells; Female; Genes, Tumor Suppressor; Humans; Metabolic Networks and Pathways; Molecular Targeted Therapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Translational Research, Biomedical

2012
TGF-β signalling and its role in cancer progression and metastasis.
    Cancer metastasis reviews, 2012, Volume: 31, Issue:3-4

    The transforming growth factor-β (TGF-β) system signals via protein kinase receptors and SMAD mediators to regulate a large number of biological processes. Alterations of the TGF-β signalling pathway are implicated in human cancer. Prior to tumour initiation and early during progression, TGF-β acts as a tumour suppressor; however, at later stages, it is often a tumour promoter. Knowledge about the mechanisms involved in TGF-β signal transduction has allowed a better understanding of cancer progression, invasion, metastasis and epithelial-to-mesenchymal transition. Furthermore, several molecular targets with great potential in therapeutic interventions have been identified. This review discusses the TGF-β signalling pathway, its involvement in cancer and current therapeutic approaches.

    Topics: Animals; Disease Progression; Epithelial-Mesenchymal Transition; Humans; Mutation; Neoplasm Metastasis; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2012
Targeting of TAK1 in inflammatory disorders and cancer.
    Trends in pharmacological sciences, 2012, Volume: 33, Issue:10

    The transcription factors nuclear factor-κB (NF-κB) and activating protein-1 (AP-1) are critical regulators of stress responses, immunity, inflammation and cancer. A large variety of cellular stimuli utilize these signaling pathways through a common upstream kinase transforming growth factor-β-activated kinase 1 (TAK1). TAK1 was originally identified as a mitogen-activated kinase kinase kinase (MAP3K) activated by transforming growth factor-β (TGF-β); however, it has been characterized as a key regulator in inflammatory and immune signaling pathways. In addition, microbial proteins and components of host cell signaling scramble for the TAK1 complex in innate immunity. This review highlights the recent advances in the activation mechanisms and physiological functions of TAK1. Research targeting TAK1 raises the potential for new therapeutic options for inflammatory disorders, including cancer.

    Topics: Animals; Humans; Immunity, Innate; Inflammation; MAP Kinase Kinase Kinases; Neoplasms; NF-kappa B; Signal Transduction; Transforming Growth Factor beta

2012
Complexities of TGF-β targeted cancer therapy.
    International journal of biological sciences, 2012, Volume: 8, Issue:7

    Many advanced tumors produce excessive amounts of Transforming Growth Factor-β (TGF-β) which, in normal epithelial cells, is a potent growth inhibitor. However, in oncogenically activated cells, the homeostatic action of TGF-β is often diverted along alternative pathways. Hence, TGF-β signaling elicits protective or tumor suppressive effects during the early growth-sensitive stages of tumorigenesis. However, later in tumor development when carcinoma cells become refractory to TGF-β-mediated growth inhibition, the tumor cell responds by stimulating pathways with tumor progressing effects. At late stages of malignancy, tumor progression is driven by TGF-β overload. The tumor microenvironment is a target of TGF-β action that stimulates tumor progression via pro-tumorigenic effects on vascular, immune, and fibroblastic cells. Bone is one of the richest sources of TGF-β in the body and a common site for dissemination of breast cancer metastases. Osteoclastic degradation of bone matrix, which accompanies establishment and growth of metastases, triggers further release of bone-derived TGF-β. This leads to a vicious positive feedback of tumor progression, driven by ever increasing levels of TGF-β released from both the tumor and bone matrix. It is for this reason, that pharmaceutical companies have developed therapeutic agents that block TGF-β signaling. Nonetheless, the choice of drug design and dosing strategy can affect the efficacy of TGF-β therapeutics. This review will describe pre-clinical and clinical data of four major classes of TGF-β inhibitor, namely i) ligand traps, ii) antisense oligonucleotides, iii) receptor kinase inhibitors and iv) peptide aptamers. Long term dosing strategies with TGF-β inhibitors may be ill-advised, since this class of drug has potentially highly pleiotropic activity, and development of drug resistance might potentiate tumor progression. Current paradigms for the use of TGF-β inhibitors in oncology have therefore moved towards the use of combinatorial therapies and short term dosing, with considerable promise for the clinic.

    Topics: Animals; Antineoplastic Agents; Humans; Neoplasms; Transforming Growth Factor beta

2012
Retinoid and TGF-β families: crosstalk in development, neoplasia, immunity, and tissue repair.
    Seminars in nephrology, 2012, Volume: 32, Issue:3

    Transforming growth factor-β (TGF-β) isoforms are profibrotic cytokines, par excellence, and have complex multifunctional effects on many systems, depending on the biologic setting. Retinoids are vitamin A derivatives that also have diverse effects in development, physiology, and disease. The interactions between these classes of molecules are, not surprisingly, highly complex and are dependent on the tissue, cellular, and molecular settings.

    Topics: Embryonic Development; Fetal Development; Humans; Neoplasms; Receptors, Retinoic Acid; Retinoid X Receptors; Retinoids; Signal Transduction; Transforming Growth Factor beta; Tretinoin; Vitamin A; Wound Healing

2012
[Role of CEACAM in neutrophil activation].
    Postepy higieny i medycyny doswiadczalnej (Online), 2012, Aug-24, Volume: 66

     Neutrophils express many surface adhesion molecules, including CEACAM1, CEACAM3, CEACAM4, CEACAM6 and CEACAM8 glycoproteins, which play an important role in biological functions of neutrophils such as adhesion, phagocytosis, oxidative burst and degranulation. CEACAM3 activates neutrophils and initiates phagocytosis as a result of binding to bacterial Opa protein. In addition, CEACAM1 and CEACAM6 can delay apoptosis. All neutrophil CEACAMs, except for CEACAM3, can stimulate adhesion of neutrophils to endothelium. One CEACAM family member, CEA, which is not expressed by neutrophils, displays strong chemotactic activity, and probably can prime and/or activate neutrophils to adhesion. Induction of CEACAM signaling can be initiated by dimerization of CEACAMs and/or phosphorylation of their cytoplasmic domains. CEACAM signaling is often associated with an increase in the cytoplasmic calcium level.

    Topics: Antigens, CD; Cadherins; Cell Adhesion Molecules; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Neutrophil Activation; Neutrophils; Phagocytosis; Signal Transduction; Transforming Growth Factor beta

2012
TGF-β - an excellent servant but a bad master.
    Journal of translational medicine, 2012, Sep-03, Volume: 10

    The transforming growth factor (TGF-β) family of growth factors controls an immense number of cellular responses and figures prominently in development and homeostasis of most human tissues. Work over the past decades has revealed significant insight into the TGF-β signal transduction network, such as activation of serine/threonine receptors through ligand binding, activation of SMAD proteins through phosphorylation, regulation of target genes expression in association with DNA-binding partners and regulation of SMAD activity and degradation. Disruption of the TGF-β pathway has been implicated in many human diseases, including solid and hematopoietic tumors. As a potent inhibitor of cell proliferation, TGF-β acts as a tumor suppressor; however in tumor cells, TGF-β looses anti-proliferative response and become an oncogenic factor. This article reviews current understanding of TGF-β signaling and different mechanisms that lead to its impairment in various solid tumors and hematological malignancies.

    Topics: Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2012
TGFβ signalling in context.
    Nature reviews. Molecular cell biology, 2012, Volume: 13, Issue:10

    The basic elements of the transforming growth factor-β (TGFβ) pathway were revealed more than a decade ago. Since then, the concept of how the TGFβ signal travels from the membrane to the nucleus has been enriched with additional findings, and its multifunctional nature and medical relevance have relentlessly come to light. However, an old mystery has endured: how does the context determine the cellular response to TGFβ? Solving this question is key to understanding TGFβ biology and its many malfunctions. Recent progress is pointing at answers.

    Topics: Animals; Cell Membrane; Cell Nucleus; Chromatin; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Phosphorylation; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2012
[The role of TGF-β-related signal transduction pathways in pathogenesis of epithelial-mesenchymal transition as a key element in cancer development and progression].
    Postepy higieny i medycyny doswiadczalnej (Online), 2012, Sep-07, Volume: 66

    Epithelial-mesenchymal transition (EMT) is a biological process that drives polarized, immotile epithelial cells to undergo multiple biochemical changes to acquire a mesenchymal cell phenotype. The characteristic features of EMT are cell apolarity, loss of cellular adhesion, reduced expression of E-cadherin and increased migratory capacity, as well as invasiveness. EMT is a physiological process that is essential for normal embryonic development. Additionally, abnormal activation of EMT contributes to some human pathologies such as tissue fibrosis, cancer cell invasion and metastasis. In both situations, the basic molecular mechanisms are similar, but lead to different effects depending on cell type and biological conditions of the environment. TGF-β is a multifunctional cytokine that controls proliferation, differentiation and other functions in many cell types. It has been found that neoplastic development converts TGF-β into an oncogenic cytokine. It activates various molecular processes, which are engaged in EMT initiation. All that makes TGF-β a key regulator of EMT.  

    Topics: Animals; Cadherins; Cell Adhesion; Cell Differentiation; Disease Progression; Epithelial-Mesenchymal Transition; Humans; Mesenchymal Stem Cells; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Precancerous Conditions; Signal Transduction; Transforming Growth Factor beta

2012
Controversies on the role of Th17 in cancer: a TGF-β-dependent immunosuppressive activity?
    Trends in molecular medicine, 2012, Volume: 18, Issue:12

    The immune system has important roles in limiting the spread of cancer and shaping the tumor microenvironment. Although the contributions of T helper 17 (Th17) cells (a subtype of CD4(+) T lymphocytes) to autoimmunity and allergy response are well known, their roles in cancer remain ambiguous. Despite adoptive transfer studies indicating that mouse Th17 cells support anticancer immunity, the Th17 cells that naturally infiltrate experimental tumors appear to have a tumor-promoting effect. These contradictory properties can be related to the high degree of plasticity inherent in Th17 cells and their capacity to differentiate into tumoricidal Th1-like cells. Mouse Th17 cells induced by transforming growth factor-β (TGF-β) express CD39 and CD73 ectonucleotidases on their surfaces, which leads to adenosine release and suppression of T cell immunity. Here, we discuss how TGF-β acts as a molecular switch controlling the immunoregulatory properties of Th17 cells.

    Topics: 5'-Nucleotidase; Animals; Antigen-Presenting Cells; Antigens, CD; Apyrase; Cell Differentiation; Immune Tolerance; Immunity, Cellular; Mice; Models, Immunological; Neoplasms; Neovascularization, Pathologic; Th1 Cells; Th17 Cells; Transforming Growth Factor beta

2012
Advances in the biology of bone metastasis: how the skeleton affects tumor behavior.
    Bone, 2011, Volume: 48, Issue:1

    It is increasingly evident that the microenvironment of bone can influence the cancer phenotype in many ways that favor growth in bone. The ability of cancer cells to adhere to bone matrix and to promote osteoclast formation are key requirements for the establishment and growth of bone metastases. Several cytokine products of breast cancers (e.g. PTHrP, IL-11, IL-8) have been shown to act upon host cells of the bone microenvironment to promote osteoclast formation, allowing for excessive bone resorption. The increased release of matrix-derived growth factors, especially TGF-β, acts back upon the tumor to facilitate further tumor expansion and enhance cytokine production, and also upon osteoblasts to suppress bone formation. This provides a self-perpetuating cycle of bone loss and tumor growth within the skeleton. Other contributing factors favoring tumor metastasis and colonization in bone include the unique structure and stiffness of skeletal tissue, along with the diverse cellular composition of the marrow environment (e.g. bone cells, stromal fibroblasts, immune cells), any of which can contribute to the phenotypic changes that can take place in metastatic deposits that favor their survival. Additionally, it is also apparent that breast cancer cells begin to express different bone specific proteins as well as proteins important for normal breast development and lactation that allow them to grow in bone and stimulate bone destruction. Taken together, these continually emerging areas of study suggest new potential pathways important in the pathogenesis of bone metastasis and potential areas for targeting therapeutics.

    Topics: Bone and Bones; Bone Neoplasms; Bone Resorption; Breast Neoplasms; Cytokines; Female; Humans; Interleukin-11; Interleukin-8; Neoplasms; Osteoblasts; Osteoclasts; Osteogenesis; Parathyroid Hormone-Related Protein; Transforming Growth Factor beta

2011
Gene regulation by SMAR1: Role in cellular homeostasis and cancer.
    Biochimica et biophysica acta, 2011, Volume: 1815, Issue:1

    Changes in the composition of nuclear matrix associated proteins contribute to alterations in nuclear structure, one of the major phenotypes of malignant cancer cells. The malignancy-induced changes in this structure lead to alterations in chromatin folding, the fidelity of genome replication and gene expression programs. The nuclear matrix forms a scaffold upon which the chromatin is organized into periodic loop domains called matrix attachment regions (MAR) by binding to various MAR binding proteins (MARBPs). Aberrant expression of MARBPs modulates the chromatin organization and disrupt transcriptional network that leads to oncogenesis. Dysregulation of nuclear matrix associated MARBPs has been reported in different types of cancers. Some of these proteins have tumor specific expression and are therefore considered as promising diagnostic or prognostic markers in few cancers. SMAR1 (scaffold/matrix attachment region binding protein 1), is one such nuclear matrix associated protein whose expression is drastically reduced in higher grades of breast cancer. SMAR1 gene is located on human chromosome 16q24.3 locus, the loss of heterozygosity (LOH) of which has been reported in several types of cancers. This review elaborates on the multiple roles of nuclear matrix associated protein SMAR1 in regulating various cellular target genes involved in cell growth, apoptosis and tumorigenesis.

    Topics: Animals; Apoptosis; Cell Cycle; Cell Cycle Proteins; DNA-Binding Proteins; Gene Expression Regulation; Genes, bcl-1; Homeostasis; Humans; Neoplasm Invasiveness; Neoplasms; Nuclear Proteins; Signal Transduction; Transforming Growth Factor beta

2011
Interaction of p53 with tumor suppressive and oncogenic signaling pathways to control cellular reactive oxygen species production.
    Antioxidants & redox signaling, 2011, Sep-15, Volume: 15, Issue:6

    p53 is a crucial transcription factor with tumor suppressive properties that elicits its function through specific target genes. It constitutes a pivotal system that integrates information received by many signaling pathways and subsequently orchestrates cell fate decisions, namely, growth-arrest, senescence, or apoptosis. Reactive oxygen species (ROS) production in cells can play a key role in signal transduction, being able to trigger different processes as cell death or cell proliferation. Sustained oxidative stress can induce genomic instability and collaborates with cancer development, whereas acute enhancement of high ROS levels leads to toxic oxidative cell damage and cell death. Here, it has been considered p53 broad potential contribution through its ability to regulate selected key cancer signaling pathways, where ROS participate as inductors or effectors of the final biological outcome. Further, we have discussed how p53 could play a role in preventing potentially harmful oxidative state and cell proliferation by pro-oncogenic pathways such as PI3K/AKT/mTOR and WNT/β-catenin or under hypoxia state. In addition, we have considered potential mechanisms by which p53 could collaborate with signal transduction pathways such as transforming growth factor-β (TGF-β) and stress-activated protein kinases (SAPK) that produce ROS, to stop or eliminate uncontrolled proliferating cells.

    Topics: Apoptosis; Genomic Instability; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Mitogen-Activated Protein Kinases; Neoplasms; Oncogenes; Reactive Oxygen Species; Signal Transduction; TOR Serine-Threonine Kinases; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Wnt Proteins

2011
Transforming growth factor-β and the hallmarks of cancer.
    Cellular signalling, 2011, Volume: 23, Issue:6

    Tumorigenesis is in many respects a process of dysregulated cellular evolution that drives malignant cells to acquire six phenotypic hallmarks of cancer, including their ability to proliferate and replicate autonomously, to resist cytostatic and apoptotic signals, and to induce tissue invasion, metastasis, and angiogenesis. Transforming growth factor-β (TGF-β) is a potent pleiotropic cytokine that functions as a formidable barrier to the development of cancer hallmarks in normal cells and tissues. Paradoxically, tumorigenesis counteracts the tumor suppressing activities of TGF-β, thus enabling TGF-β to stimulate cancer invasion and metastasis. Fundamental gaps exist in our knowledge of how malignant cells overcome the cytostatic actions of TGF-β, and of how TGF-β stimulates the acquisition of cancer hallmarks by developing and progressing human cancers. Here we review the molecular and cellular mechanisms that underlie the ability of TGF-β to mediate tumor suppression in normal cells, and conversely, to facilitate cancer progression and disease dissemination in malignant cells.

    Topics: Cell Cycle; Cell Movement; Cell Proliferation; Cell Survival; Epithelial-Mesenchymal Transition; Humans; Intracellular Signaling Peptides and Proteins; Mutation, Missense; Neoplasm Invasiveness; Neoplasms; Neovascularization, Pathologic; Phenotype; Signal Transduction; Transforming Growth Factor beta

2011
The dynamic roles of TGF-β in cancer.
    The Journal of pathology, 2011, Volume: 223, Issue:2

    The transforming growth factor-β (TGF-β) signalling pathway plays a critical and dual role in the progression of human cancer. During the early phase of tumour progression, TGF-β acts as a tumour suppressor, exemplified by deletions or mutations in the core components of the TGF-β signalling pathway. On the contrary, TGF-β also promotes processes that support tumour progression such as tumour cell invasion, dissemination, and immune evasion. Consequently, the functional outcome of the TGF-β response is strongly context-dependent including cell, tissue, and cancer type. In this review, we describe the molecular signalling pathways employed by TGF-β in cancer and how these, when perturbed, may lead to the development of cancer. Concomitantly with our increased appreciation of the molecular mechanisms that govern TGF-β signalling, the potential to therapeutically target specific oncogenic sub-arms of the TGF-β pathway increases. Indeed, clinical trials with systemic TGF-β signalling inhibitors for treatment of cancer patients have been initiated. However, considering the important role of TGF-β in cardiovascular and many other tissues, careful screening of patients is warranted to minimize unwanted on-target side effects.

    Topics: Disease Progression; Epithelial-Mesenchymal Transition; Humans; Neoplasm Metastasis; Neoplasm Proteins; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
TGF-beta-dependent and -independent roles of STRAP in cancer.
    Frontiers in bioscience (Landmark edition), 2011, 01-01, Volume: 16, Issue:1

    The serine-threonine kinase receptor-associated protein (STRAP) was initially identified as a putative inhibitor of the canonical TGF-beta signaling pathway. Because the Smad-dependent TGF-beta pathway negatively regulates cellular growth, early functional studies suggested that STRAP behaves as an oncogene. Indeed, a correlation between STRAP overexpression and various cancers has been identified. With the emergence of new studies on the biological function of STRAP, it is becoming clear that STRAP regulates several distinct cellular processes and modulates multiple signaling pathways. While STRAP itself does not possess enzymatic activity, it appears that STRAP influences biological processes through associations with cellular proteins. In this review, we will describe the TGF-beta-dependent and -independent functions of STRAP and provide a context for the significance of STRAP activity in the development of cancer.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Calmodulin-Binding Proteins; Epithelial-Mesenchymal Transition; Humans; MAP Kinase Kinase Kinase 5; Mice; Neoplasms; NM23 Nucleoside Diphosphate Kinases; Phosphatidylinositol 3-Kinases; Protein Serine-Threonine Kinases; Proteins; Pyruvate Dehydrogenase Acetyl-Transferring Kinase; RNA-Binding Protein EWS; RNA-Binding Proteins; RNA, Messenger; Signal Transduction; Smad Proteins; Smad Proteins, Inhibitory; Transforming Growth Factor beta; Tumor Cells, Cultured

2011
Growth Factors and their receptors in cancer metastases.
    Frontiers in bioscience (Landmark edition), 2011, 01-01, Volume: 16, Issue:2

    Metastatic, rather than primary tumours are responsible for ninety percent cancer deaths. Despite significant advances in the understanding of molecular and cellular mechanisms in tumour metastases, there are limitations in preventive treatment of metastatic tumours. Much evidence arising from laboratory and clinical studies suggests that growth factors and their receptors are implicated in cancer metastases development. We review the origin and production of growth factors and their receptors in all stages of cancer metastases including epithelial-mesenchymal transition, cancer cell invasion and migration, survival within the circulation, seeding at distant organs and metastatic tumour angiogenesis. The functions of growth factors and their receptors are also discussed. This review presents the efforts made in understanding this challenge to aid in the development of new treatment strategies for cancer metastases.

    Topics: Angiopoietins; Animals; Apoptosis; Cell Movement; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; ErbB Receptors; Glucose-6-Phosphate Isomerase; Hepatocyte Growth Factor; Humans; Insulin-Like Growth Factor I; Intercellular Signaling Peptides and Proteins; Interleukin-8; Multienzyme Complexes; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasm Seeding; Neoplasms; Neoplastic Cells, Circulating; Neovascularization, Pathologic; Phosphodiesterase I; Phosphoric Diester Hydrolases; Pyrophosphatases; Receptor, IGF Type 1; Receptors, Growth Factor; Ribonuclease, Pancreatic; Smad Proteins; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2011
Epithelial and mesenchymal phenotypic switchings modulate cell motility in metastasis.
    Frontiers in bioscience (Landmark edition), 2011, 01-01, Volume: 16, Issue:3

    The most ominous stage of cancer progression is metastasis, or the dissemination of carcinoma cells from the primary site into distant organs. Metastases are often resistant to current extirpative therapies and even the newest biological agents cure only a small subset of patients. Therefore a greater understanding of tumor biology that integrates properties intrinsic to carcinomas with tissue environmental modulators of behavior is needed. In no aspect of tumor progression is this more evident than the acquisition of cell motility that is critical for both escape from the primary tumor and colonization. In this overview, we discuss how this behavior is modified by carcinoma cell phenotypic plasticity that is evidenced by reversible switching between epithelial and mesenchymal phenotypes. The presence or absence of intercellular adhesions mediate these switches and dictate the receptivity towards signals from the extracellular milieu. These signals, which include soluble growth factors, cytokines, and extracellular matrix embedded with matrikines and matricryptines will be discussed in depth. Finally, we will describe a new mode of discerning the balance between epithelioid and mesenchymal movement.

    Topics: Cadherins; Cell Adhesion; Cell Movement; Cell Transformation, Neoplastic; Cytokines; Desmosomes; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Extracellular Matrix Proteins; Gap Junctions; Hepatocyte Growth Factor; Humans; Insulin-Like Growth Factor I; Integrins; Neoplasm Metastasis; Neoplasms; Phenotype; Signal Transduction; Tight Junctions; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2011
Inhibitory Smad7: emerging roles in health and disease.
    Current molecular pharmacology, 2011, Volume: 4, Issue:2

    Smad7 is an inhibitory Smad protein that blocks Transforming Growth Factor-beta (TGF-β) signaling through a negative feedback loop, also capable of mediating the crosstalk between TGF-β and other signaling pathways. Smad7 mRNA and protein levels are upregulated after TGF-β signaling; subsequently, Smad7 protein binds TGF-β type I receptor blocking R-Smad phosphorylation and eventually TGF-β signaling. Because of this inhibitory function, Smad7 can antagonize diverse cellular processes regulated by TGF-β such as cell proliferation, differentiation, apoptosis, adhesion and migration. Smad7 induction by different cytokines, besides TGF-β, is also critical for crosstalk/integration of a variety of signaling pathways, and relevant in the pathology of some diseases. Thus, Smad7 plays a key role in the control of various physiological events, and even in some pathological processes including fibrosis and cancer. This review highlights the main known functions of Smad7 with a particular focus on the relevance that alterations of Smad7 function may have in homeostasis, also describing some Smad7 emerging roles in the development of several human diseases that identify this protein as a potential therapeutic target.

    Topics: Animals; Fibrosis; Homeostasis; Humans; Neoplasms; Protein Processing, Post-Translational; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad7 Protein; Transforming Growth Factor beta

2011
Switching TGFβ from a tumor suppressor to a tumor promoter.
    Current opinion in genetics & development, 2011, Volume: 21, Issue:1

    TGFβ acts as a potent tumor suppressor and tumor promoter in a context dependent manner. Tumor suppressive functions include inhibition of cell proliferation, induction of apoptosis and regulation of autophagy. As tumors develop they switch their response to TGFβ and utilise this factor as a potent promoter of cell motility, invasion, metastasis and tumor stem cell maintenance. These multifactorial tumor influencing actions of TGFβ involve regulation of an increasing number of signal transduction pathways employing a diverse range of signaling molecules. Understanding the molecular mechanisms of how tumor cells respond to TGFβ and switch their response to this cytokine during disease progression is vital for both the development and the informed use of potentially powerful TGFβ targeted therapeutics.

    Topics: Animals; Antineoplastic Agents; Disease Progression; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2011
The immunoregulatory mechanisms of carcinoma for its survival and development.
    Journal of experimental & clinical cancer research : CR, 2011, Jan-21, Volume: 30

    The immune system in patients detects and eliminates tumor cells, but tumors still progress persistently. The mechanisms by which tumor cells survive under the pressure of immune surveillance are not fully understood. This review is to present the evidence from clinical studies, showing a significant correlation of clinicopathological features of carcinoma with: (1) the loss of classical human leukocyte antigen class I, (2) the up-regulation of non-classical human leukocyte antigen class I, pro-apoptotic Fas ligand and receptor-binding cancer antigen expressed on SiSo cells I, and (3) the formation of immunosuppressive microenvironment by up-regulation of transforming growth factor-beta, Galectin-1, inhibitory ligand B7s, indoleamine 2,3-dioxygenase and arginase, as well as by recruitment of tumor-induced myeloid-derived suppressor cells and regulatory T cells. All of these factors may together protect carcinoma cells from the immune-cytotoxicity.

    Topics: Animals; Antigens, CD; Antigens, Neoplasm; Arginase; Fas Ligand Protein; Galectin 1; Humans; Immune Tolerance; Indoleamine-Pyrrole 2,3,-Dioxygenase; Lymphocyte Activation; Lymphocytes, Tumor-Infiltrating; Neoplasms; Severe Combined Immunodeficiency; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta

2011
[Involvement of LMO4 in tumorigenesis associated epithelial-mesenchymal transition].
    Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences, 2011, Volume: 40, Issue:1

    LMO4 is a novel member of the LIM-only (LMO) subfamily of LIM domain-containing transcription factors, so named because they are composed almost entirely of two tandem LIM domains. This subgroup of LIM proteins has 4 members: LMO-1, LMO-2, LMO-3 and LMO-4. They all play important roles in the normal mammalian development, functioning as an important regulator of cell proliferation. LMO4 is highly expressed in the epithelial compartments at locations of active epithelial-mesenchymal interactions, and can interact with some signaling pathways involved in epithelial-mesenchymal signaling. Thus the disregulation of LMO4 expression may be involved in tumorigenesis. In this paper, we will at first expound LMO4 in detail, based on which the possible mechanisms for its interaction with TGF-β signaling and the roles of this cross-talk between them in the vital process of cell will be introduced. All of those will add to our understanding of tumorigenesis and contribute to the search of new targets for the treatment of cancer.

    Topics: Adaptor Proteins, Signal Transducing; Epithelial-Mesenchymal Transition; Homeodomain Proteins; Humans; LIM Domain Proteins; Neoplasms; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2011
[Encounter of cancer cells with bone. Epithelial-mesenchymal transition in cancer].
    Clinical calcium, 2011, Volume: 21, Issue:3

    Acquisition of invasive phenotypes of cancer cells is one of the key steps to promote metastasis. It has been reported that fibroblastic cells are involved in enhancement of proliferation and invasion of cancer cells. Thus, it is important to know the molecular mechanisms of EMT (epithelial-mesenchymal transition) for developing diagnosis and cancer therapy. Since TGF-βis a key mediator of EMT and frequently expressed in various tumors, it may regulate not only the EMT of cancer cells as they acquire metastatic properties, but also the EMT of normal epithelial cells that are adjacent to tumors. In this review, we will discuss the EMT induced by TGF-β.

    Topics: Epithelial-Mesenchymal Transition; Humans; Molecular Targeted Therapy; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Protein Splicing; RNA-Binding Proteins; Signal Transduction; Transforming Growth Factor beta

2011
[Encounter of cancer cells with bone. The significance of cancer stem cells and epithelial-mesenchymal transition in tumor invasion and metastasis].
    Clinical calcium, 2011, Volume: 21, Issue:3

    Cancer stem cells (CSCs) , which are subset of tumor cells resistant to radiation and chemotherapy, are associated with malignant characteristics of tumor and possess both self-renewal ability and pluripotency for tumor formation. In the process of generating non-CSCs from CSCs, gene mutations and epigenetic changes are induced in those cells, resulting in composition of tumor tissue with heterogeneous cell population. CSCs have been recognized as the source of metastatic foci. Epithelial-mesenchymal transition (EMT) is a change in cellular phenotype characterized by the loss of cell-to-cell adhesions and the gain of migratory behaviors,which has been shown to be a critical factor for initiating cancer invasion and metastasis. However, some recent studies suggest that EMT is not essential requirement for tumor invasion and metastasis. Herein, we discuss the biological significance of CSCs and EMT in tumor invasion and metastasis.

    Topics: Cell Adhesion; Cell Movement; Epigenesis, Genetic; Epithelial-Mesenchymal Transition; Humans; Mutation; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neoplastic Stem Cells; Transforming Growth Factor beta

2011
Epithelial-mesenchymal transition: implications in cancer progression and metastasis.
    Current pharmaceutical biotechnology, 2011, Volume: 12, Issue:11

    During the past few years, Epithelial-Mesenchymal Transition (EMT) has emerged as one of the most hot spots in clinical research. Its existence in human tumors can form the basis for explaining characteristics of cancer progression and metastasis, as well as certain cases of drug resistance and relapses after treatment. These cellular responses are tightly regulated by intracellular signaling pathways evoked by humoral factors that include growth factors, chemokines and cytokines. Indeed, several gene regulatory programs known to promote EMT during development have recently been discovered to play key roles in cancer progression. A deeper understanding of the cellular and molecular basis of these different programs should aid in both the development of better diagnosis methods, as well as of specific treatments for invasive cancer. In this review we set out to summarize recent novel insights into the molecular players underlying EMT and its relation with cancer progression and metastasis.

    Topics: Animals; Disease Progression; Epithelial-Mesenchymal Transition; Humans; Matrix Metalloproteinases; MicroRNAs; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Receptor Protein-Tyrosine Kinases; Signal Transduction; Transforming Growth Factor beta

2011
Non-cell-autonomous tumor suppression: oncogene-provoked apoptosis promotes tumor cell senescence via stromal crosstalk.
    Journal of molecular medicine (Berlin, Germany), 2011, Volume: 89, Issue:9

    Activated oncogenes evoke cellular fail-safe programs such as apoptosis, senescence, or autophagy to protect the organism from the expansion of damaged and potentially harmful cells. Non-cell-autonomous interactions between tumor cells and nonmalignant bystander cells add to cell-autonomous modes of tumor suppression during tumor development and progression. In particular, the role of stroma or host immune cells converting tumor-generated signals into a response that feeds back to the tumor cell population has been experimentally underappreciated. Using the Eμ-myc transgenic mouse lymphoma model, we elucidated how constitutive Myc signaling indirectly promotes cellular senescence via cytokines that were released by nonmalignant cells in response to oncogene-evoked cell-autonomous effects. Specifically, Myc primarily promotes apoptosis in a subset of the tumor cell population, leading to the attraction of macrophages, which subsequently engulf the apoptotic remainders. Phagocytosis-activated macrophages, in turn, exhibit strongly increased secretion of various cytokines, among them transforming growth factor beta to an extent that is capable of inducing cellular senescence in surrounding malignant cells. Our findings, recapitulated in human aggressive B-cell lymphomas, unveil that apoptosis and senescence are not simply two context-dependent cell-autonomous choices of stress responses, but rather cooperate via extracellular mediators-namely cells of the innate immune system-to profoundly limit tumorigenesis in vivo. A deeper mechanistic understanding of the organismic interconnection between different fail-safe programs will help to identify cellular components of the tumor stroma and their signal mediators that are readily available to impose a second line of host defense against cancer cells. This will open new perspectives for the development of antineoplastic therapies, whose targets not only encompass tumor but also stroma cell populations.

    Topics: Animals; Apoptosis; Cellular Senescence; Humans; Immune System; Macrophages; Neoplasms; Oncogene Proteins; Stromal Cells; Transforming Growth Factor beta; Tumor Microenvironment

2011
TGF-β/TGF-β receptor system and its role in physiological and pathological conditions.
    Clinical science (London, England : 1979), 2011, Volume: 121, Issue:6

    The TGF-β (transforming growth factor-β) system signals via protein kinase receptors and Smad mediators to regulate a plethora of biological processes, including morphogenesis, embryonic development, adult stem cell differentiation, immune regulation, wound healing and inflammation. In addition, alterations of specific components of the TGF-β signalling pathway may contribute to a broad range of pathologies such as cancer, cardiovascular pathology, fibrosis and congenital diseases. The knowledge about the mechanisms involved in TGF-β signal transduction has allowed a better understanding of the disease pathogenicity as well as the identification of several molecular targets with great potential in therapeutic interventions.

    Topics: Animals; Cardiovascular Diseases; Humans; Inflammation; Lung Diseases; Molecular Targeted Therapy; Musculoskeletal Diseases; Mutation; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
TGF-β receptor signaling pathways in angiogenesis; emerging targets for anti-angiogenesis therapy.
    Current pharmaceutical biotechnology, 2011, Volume: 12, Issue:12

    Angiogenesis, the formation of new blood vessels is essential for diverse physiological processes such as development, but also for pathological conditions like tumor growth. Most studied in this context are tyrosine kinase signaling pathways such as those involving vascular endothelial growth factor (VEGF). There is however accumulating evidence that more pathways are as essential for angiogenesis. Knockout studies of factors in transforming growth factor β (TGF-β) signaling have for example showed that also this pathway is indispensable for angiogenesis. This review highlights our understanding of TGF-β signaling in vascular development and angiogenesis. In particular, we focus on recent insights into the role of the TGF-β type I receptor ALK1 and co-receptor endoglin in tumor angiogenesis, which provide opportunities for the development of new anti-angiogenesis therapies for treatment of cancer patients.

    Topics: Animals; Endothelium, Vascular; Humans; Neoplasms; Neovascularization, Pathologic; Neovascularization, Physiologic; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2011
Antibody targeting of TGF-β in cancer patients.
    Current pharmaceutical biotechnology, 2011, Volume: 12, Issue:12

    The role of TGF-β in tumor development and progression is complex. Genetic mutations that disrupt the antiproliferative signaling effects of TGF-β play a key role in the process of malignant transformation for many types of tumors. Paradoxically, this loss of sensitivity to TGF-β's inhibitory actions often leads to TGF-β overexpression by the tumor cells or by normal cells that are recruited to the tumor microenvironment. Elevated concentrations of TGF-β in the tumor microenvironment have been shown to facilitate tumor growth and metastasis. Numerous published studies have provided evidence that inhibition of TGF-β using antibodies, soluble receptors and small molecule inhibitors of TGF-β signal transduction can have beneficial effects in murine models of cancer. Given the pleiotropic nature of TGF-β and its homeostatic role in numerous biological processes, serious concerns have been expressed regarding the safety of administering TGF-β antagonists to human patients. Interestingly, the results of numerous animal toxicology studies of TGF-β antibodies in normal rodents and primates have shown that administration of neutralizing anti-TGF-β antibodies is well tolerated and any adverse effects were reversible or self-limiting. Likewise, administration of a human anti-TGF-β antibody (fresolimumab) in three separate human phase 1 clinical trials has also been shown to be well tolerated.

    Topics: Animals; Antibodies, Monoclonal; Disease Models, Animal; Humans; Neoplasms; Transforming Growth Factor beta

2011
Tgf-beta type I receptor (Alk5) kinase inhibitors in oncology.
    Current pharmaceutical biotechnology, 2011, Volume: 12, Issue:12

    The TGFβ type I receptor kinase (ALK5) is an attractive target for intervention in TGFβ signaling due to its druggability as well as its centrality and specificity in the pathway. A number of potent, selective ALK5 inhibitors have been discovered which interact with the ATP-binding site of ALK5. Crystallographic studies of these molecules bound to ALK5 have provided an understanding of potency and selectivity achieved by these inhibitors. ALK5 kinase inhibitors are potently active in models of cancer due to mechanisms of action similar to those for other TGFβ inhibitory agents. Recent insights into the function of TGFβ in human tumors as well as in preclinical models of cancer are helping to identify potential target patient populations and drug combinations for the development of ALK5 kinase inhibitors and other TGFβ- targeted therapeutics. Differences in the toxicological effects, pharmacokinetics and clinical side effects of ALK5 kinase inhibitors and other TGFβ-targeted agents provide a useful and differentiated set of TGFβ signaling inhibitory agents to investigate in clinical studies.

    Topics: Animals; Antineoplastic Agents; Humans; Neoplasms; Protein Kinase Inhibitors; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2011
The role of miRNAs in cytokine signaling.
    Frontiers in bioscience (Landmark edition), 2011, 06-01, Volume: 16, Issue:6

    This review explores the relationship between cytokines and microRNAs (miRNAs). In particular, the regulation of miRNAs by pro-inflammatory cytokines, anti-inflammatory cytokines, interferons and transforming growth factor beta are examined, highlighting how miRNAs can mediate some of the known functions of these cytokines, as well as identifying novel gene targets, mechanisms and cross-talk between pathways.

    Topics: Animals; Cytokines; Humans; Inflammation; Interferons; Interleukin-1; Interleukin-10; Interleukin-6; MicroRNAs; Models, Biological; Neoplasms; Receptors, Cytokine; Signal Transduction; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2011
Exploring anti-TGF-β therapies in cancer and fibrosis.
    Growth factors (Chur, Switzerland), 2011, Volume: 29, Issue:4

    Transforming growth factor-β (TGF-β) is a multifunctional cytokine, with important roles in maintaining tissue homeostasis. TGF-β signals via transmembrane serine/threonine kinase receptors and intracellular Smad transcriptional regulators. Perturbed TGF-β signaling has been implicated in a large variety of pathological conditions. Increased TGF-β levels have been found in patients with cancer, fibrosis, and systemic sclerosis, and were correlated with disease severity. In cancer, TGF-β mediates tumor invasion and metastasis by affecting both tumor cells and the tumor microenvironment including fibroblast activation and immune suppression. Furthermore, TGF-β is a strong stimulator of extracellular matrix deposition. On the basis of these observations, small molecule inhibitors of the TGF-β receptor kinases, neutralizing antibodies that interfere with ligand?receptor interactions, antisense oligonucleotides reducing TGF-β expression, and soluble receptor ectodomains that sequester TGF-β have been developed to intervene with excessive TGF-β signaling activity in the aforementioned disorders. Here, we review the current state of anti-TGF-β therapy in clinical trials.

    Topics: Antibodies, Neutralizing; Fibrosis; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Oligonucleotides, Antisense; Protein Serine-Threonine Kinases; Receptors, Transforming Growth Factor beta; Scleroderma, Systemic; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
The ins and outs of the epithelial to mesenchymal transition in health and disease.
    Annual review of cell and developmental biology, 2011, Volume: 27

    The epithelial to mesenchymal transition (EMT) converts epithelial cells into migratory and invasive cells and is a fundamental event in morphogenesis. Although its relevance in the progression of cancer and organ fibrosis had been debated until recently, the EMT is now established as an important step in the metastatic cascade of epithelial tumors. The similarities between pathological and developmental EMTs validate the embryo as the best model to understand the molecular and cellular mechanisms involved in this process, identifying those that are hijacked during the progression of cancer and organ degeneration. Our ever-increasing understanding of how transcription factors regulate the EMT has revealed complex regulatory loops coupled to posttranscriptional and epigenetic regulatory programs. The EMT is now integrated into the systemic activities of whole organisms, establishing links with cell survival, stemness, inflammation, and immunity. In addition, the EMT now constitutes a promising target for the treatment of cancer and organ-degenerative diseases.

    Topics: Animals; Cadherins; Cell Differentiation; Cell Movement; Cell Transformation, Neoplastic; Disease; Disease Progression; Epigenesis, Genetic; Epithelial Cells; Epithelial-Mesenchymal Transition; Extracellular Matrix; Humans; Hypoxia; Immunity; Inflammation; Mesoderm; Neoplasms; RNA, Untranslated; Transforming Growth Factor beta

2011
Smad phosphoisoform signaling specificity: the right place at the right time.
    Carcinogenesis, 2011, Volume: 32, Issue:11

    Transforming growth factor (TGF)-β antagonizes mitogenic Ras signaling during epithelial regeneration, but TGF-β and Ras act synergistically in driving tumor progression. Insights into these apparently contradictory effects have come from recent detailed analyses of the TGF-β signaling process. Here, we summarize the different modes of TGF-β/Ras signaling in normal epithelium and neoplasms and show how perturbation of TGF-β signaling by Ras may contribute to a shift from tumor-suppressive to protumorigenic TGF-β activity during tumor progression. Smad proteins, which convey signals from TGF-β receptors to the nucleus, have intermediate linker regions between conserved Mad homology (MH) 1 and MH2 domains. TGF-β Type I receptor and Ras-associated kinases differentially phosphorylate Smad2 and Smad3 to create C-terminally (C), linker (L) or dually (L/C) phosphorylated (p) isoforms. In epithelial homeostasis, TGF-β-mediated pSmad3C signaling opposes proliferative responses induced by mitogenic signals. During carcinogenesis, activation of cytoplasmic Ras-associated kinases including mitogen-activated protein kinase confers a selective advantage on benign tumors by shifting Smad3 signaling from a tumor-suppressive pSmad3C to an oncogenic pSmad3L pathway, leading to carcinoma in situ. Finally, at the edges of advanced carcinomas invading adjacent tissues, nuclear Ras-associated kinases such as cyclin-dependent kinases, together with cytoplasmic kinases, alter TGF-β signals to more invasive and proliferative pSmad2L/C and pSmad3L/C signaling. Taken together, TGF-β signaling specificity arises from spatiotemporal dynamics of Smad phosphoisoforms. Based on these findings, we have reason to hope that pharmacologic inhibition of linker phosphorylation might suppress progression to human advanced carcinomas by switching from protumorigenic to tumor-suppressive TGF-β signaling.

    Topics: Animals; Humans; Neoplasms; Phosphorylation; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
TGF-β/SMAD/GLI2 signaling axis in cancer progression and metastasis.
    Cancer research, 2011, Sep-01, Volume: 71, Issue:17

    The Hedgehog (HH) and TGF-β signaling pathways represent essential regulators of cell proliferation and differentiation during embryogenesis. Pathway deregulation is a characteristic of various cancers. Recently, evidence for a convergence of these pathways at the level of the GLI2 transcription factor in the context of tumor initiation and progression to metastasis has emerged. This short review summarizes recent knowledge about GLI2 function and mechanisms of action downstream of TGF-β in cancer.

    Topics: Bone Neoplasms; Breast Neoplasms; Disease Progression; Female; Hedgehog Proteins; Humans; Kruppel-Like Transcription Factors; Melanoma; Neoplasm Metastasis; Neoplasms; Nuclear Proteins; Signal Transduction; Skin Neoplasms; Smad Proteins; Transforming Growth Factor beta; Zinc Finger Protein Gli2

2011
The tumor suppressor kinase LKB1: lessons from mouse models.
    Journal of molecular cell biology, 2011, Volume: 3, Issue:6

    Mutations in the tumor suppressor gene LKB1 are important in hereditary Peutz-Jeghers syndrome, as well as in sporadic cancers including lung and cervical cancer. LKB1 is a kinase-activating kinase, and a number of LKB1-dependent phosphorylation cascades regulate fundamental cellular and organismal processes in at least metabolism, polarity, cytoskeleton organization, and proliferation. Conditional targeting approaches are beginning to demonstrate the relevance and specificity of these signaling pathways in development and homeostasis of multiple organs. More than one of the pathways also appear to contribute to tumor growth following Lkb1 deficiencies based on a number of mouse tumor models. Lkb1-dependent activation of AMPK and subsequent inactivation of mammalian target of rapamycin signaling are implicated in several of the models, and other less well characterized pathways are also involved. Conditional targeting studies of Lkb1 also point an important role of LKB1 in epithelial-mesenchymal interactions, significantly expanding knowledge on the relevance of LKB1 in human disease.

    Topics: AMP-Activated Protein Kinase Kinases; Animals; Disease Models, Animal; Embryonic Development; Epithelium; Gene Targeting; Homeostasis; Humans; Mesoderm; Mice; Neoplasms; Organ Specificity; Peutz-Jeghers Syndrome; Protein Serine-Threonine Kinases; PTEN Phosphohydrolase; Signal Transduction; Stromal Cells; Transforming Growth Factor beta; Tumor Suppressor Proteins

2011
TGF-β and its coreceptors in cancerogenesis: an overview.
    Biomarkers in medicine, 2011, Volume: 5, Issue:6

    Besides signaling serine/threonine kinases, such as TGF-β receptors I and II, the TGF-β pathway involves several auxiliary receptors or coreceptors. Recent studies show that these coreceptors, particulary endoglin and β-glycan, have greater significance than previously thought. They regulate the availability of ligands to the key receptors, as well as their interaction and response, which could be variable and context-dependent. Understanding their true mechanism of action is important for delineating the complexity of the entire TGF-β signaling pathway. This is especially important in the context of cancerogenesis, because of therapeutic possibilities to manipulate the TGF-β system.

    Topics: Animals; Antigens, CD; Endoglin; Humans; Neoplasms; Proteoglycans; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2011
Signaling pathways governing tumor angiogenesis.
    Oncology, 2011, Volume: 81 Suppl 1

    Angiogenesis is regulated by the highly coordinated function of various proteins with pro- and antiangiogenic functions. Proangiogenic factors include vascular endothelial growth factor (VEGF), fibroblast growth factor, platelet-derived growth factor, insulin-like growth factor, transforming growth factor, angiopoietins, and several chemokines; antiangiogenic factors include thrombospondin-1, angiostatin, and endostatin. Matrix metalloproteinases display a dual role in vascular development. Notch signaling affects remodeling of the primary vascular network of uniformly sized vessels into functionally and morphologically distinct arteries, veins, and capillaries. Tumors, described as 'wounds that never heal', lose the appropriate balance among these factors. Although VEGF-targeted therapies are showing promise, new angiogenesis targets are needed to make additional gains. Here, we highlight recent advances in our understanding of the regulation of tumor angiogenesis and discuss the potential of molecular targeting as a new therapeutic approach.

    Topics: Angiogenesis Inhibitors; Angiopoietins; Calcium-Binding Proteins; Chemokines; Fibroblast Growth Factors; Humans; Intercellular Signaling Peptides and Proteins; JNK Mitogen-Activated Protein Kinases; Matrix Metalloproteinases; Membrane Proteins; Molecular Targeted Therapy; Neoplasms; Neovascularization, Pathologic; Platelet-Derived Growth Factor; Receptors, TIE; Serrate-Jagged Proteins; Signal Transduction; Stromal Cells; Transforming Growth Factor beta; Vascular Endothelial Growth Factors

2011
[The role of TGF-beta in the EMT of tumor cells].
    Sheng li ke xue jin zhan [Progress in physiology], 2011, Volume: 42, Issue:6

    Topics: Animals; Epithelial-Mesenchymal Transition; Humans; Neoplasm Metastasis; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
Strategies of treating cancer by cytokine regulation of chromosome end remodelling.
    Clinical and experimental pharmacology & physiology, 2010, Volume: 37, Issue:1

    1. Telomeres (ends of chromosomes) undergo constant remodelling during cell development, proliferation and differentiation, as well as in neoplastic cell immortalization and transformation. How the cellular microenvironment influences telomere remodelling (lengthening or shortening) remains largely unknown. 2. Recently, studies from our laboratories and others have indicated that growth factors and cytokines have significant roles in regulating telomere remodelling and thus influence cell functions and properties. Cancer cells must maintain their already short telomeres either by the enzyme telomerase or, more rarely, through alternative lengthening of telomeres, which functions independently of cellular regulation. 3. However, application of transforming growth factor-beta family cytokines induces transcriptional inhibition of telomerase in cancer cells, leading to telomere shortening, cell senescence (ageing) and apoptosis (death) by mechanisms dependent on telomerase inhibition. Furthermore, selective gene silencing of vascular endothelial growth factor and/or the telomerase catalytic subunit (i.e. telomerase reverse transcriptase) potently inhibits the growth of cervical cancer and laryngeal squamous cancer in mice. 4. The present paper summarizes our current understanding of the negative regulation by cytokines of telomere maintenance and thus cancer cell proliferation in cultured cells and mouse models.

    Topics: Animals; Bone Morphogenetic Protein 7; Cell Division; Cytokines; Drug Discovery; Gene Expression Regulation, Neoplastic; Humans; Models, Biological; Neoplasms; Telomerase; Telomere; Transforming Growth Factor beta

2010
Transforming growth factor-beta: a target for cancer therapy.
    Journal of immunotoxicology, 2010, Volume: 7, Issue:1

    Transforming growth factor-beta (TGF-beta) is a pleiotropic growth factor that regulates cell growth and differentiation, apoptosis, cell motility, extracellular matrix production, angiogenesis, and cellular immune responses. TGF-beta demonstrates paradoxical action whereby it can function to suppress early tumorigenesis; however, it can also facilitate malignant transformation and stimulate tumor growth by manipulating a more hospitable environment for tumor invasion and the development of metastases. Given the integral role of TGF-beta in transformation and cancer progression, various components of the TGF-beta signaling pathway offer potentially attractive therapeutic targets for cancer treatment. This review focuses on the role of TGF-beta in cancer and discusses both small and large molecule drugs currently in development that target TGF-beta, its receptor and important down stream steps along its signaling pathway.

    Topics: Antineoplastic Agents; Drug Design; Humans; Immunosuppressive Agents; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2010
Targeting the transforming growth factor-beta signaling pathway in human cancer.
    Expert opinion on investigational drugs, 2010, Volume: 19, Issue:1

    The transforming growth factor-ss (TGF-beta) signaling pathway plays a pivotal role in diverse cellular processes. TGF-beta switches its role from a tumor suppressor in normal or dysplastic cells to a tumor promoter in advanced cancers. It is widely believed that the Smad-dependent pathway is involved in TGF-beta tumor-suppressive functions, whereas activation of Smad-independent pathways, coupled with the loss of tumor-suppressor functions of TGF-beta, is important for its pro-oncogenic functions. TGF-beta signaling has been considered a useful therapeutic target. The discovery of oncogenic actions of TGF-beta has generated a great deal of enthusiasm for developing TGF-beta signaling inhibitors for the treatment of cancer. The challenge is to identify the group of patients where targeted tumors are not only refractory to TGF-beta-induced tumor suppressor functions but also responsive to the tumor-promoting effects of TGF-beta. TGF-beta pathway inhibitors, including small and large molecules, have now entered clinical trials. Preclinical studies with these inhibitors have shown promise in a variety of different tumor models. Here, we focus on the mechanisms of signaling and specific targets of the TGF-beta pathway that are critical effectors of tumor progression and invasion. This report also examines the therapeutic intervention of TGF-ss signaling in human cancers.

    Topics: Antineoplastic Agents; Clinical Trials as Topic; Drug Discovery; Humans; Neoplasms; Oligonucleotides, Antisense; Signal Transduction; Small Molecule Libraries; Transforming Growth Factor beta

2010
Transforming growth factor beta (TGF-beta) and inflammation in cancer.
    Cytokine & growth factor reviews, 2010, Volume: 21, Issue:1

    The transforming growth factor beta (TGF-beta) has been studied with regard to the regulation of cell behavior for over three decades. A large body of research has been devoted to the regulation of epithelial cell and derivative carcinoma cell populations in vitro and in vivo. TGF-beta has been shown to inhibit epithelial cell cycle progression and promote apoptosis that together significantly contribute to the tumor suppressive role for TGF-beta during carcinoma initiation and progression. TGF-beta is also able to promote an epithelial to mesenchymal transition that has been associated with increased tumor cell motility, invasion and metastasis. However, it has now been shown that loss of carcinoma cell responsiveness to TGF-beta stimulation can also promote metastasis. Interestingly, enhanced metastasis in the absence of a carcinoma cell response to TGF-beta stimulation has been shown to involve increased chemokine production resulting in recruitment of pro-metastatic myeloid derived suppressor cell (MDSC) populations to the tumor microenvironment at the leading invasive edge. When present, MDSCs enhance angiogenesis, promote immune tolerance and provide matrix degrading enzymes that promote tumor progression and metastasis. Further, the recruitment of MDSC populations in this context likely enhances the classic role for TGF-beta in immune suppression since the MDSCs are an abundant source of TGF-beta production. Importantly, it is now clear that carcinoma-immune cell cross-talk initiated by TGF-beta signaling within the carcinoma cell is a significant determinant worth consideration when designing therapeutic strategies to manage tumor progression and metastasis.

    Topics: Animals; Epithelial Cells; Humans; Inflammation; Macrophages; Models, Biological; Monocytes; Myeloid Cells; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2010
Angiogenesis and tumour progression: migration-stimulating factor as a novel target for clinical intervention.
    Eye (London, England), 2010, Volume: 24, Issue:3

    Migration-stimulating factor (MSF), a soluble genetically truncated isoform of fibronectin, is a potent oncofoetal regulatory molecule. Its 2.1-kb message is generated from the fibronectin gene by a variant of standard alternative splicing involving premature intra-intronic cleavage. MSF is constitutively expressed by both epithelial and stromal cells during foetal development and in patients with cancer, but is generally not expressed in healthy adults. MSF affects the behaviour of a broad range of potential target cells (fibroblasts, vascular, and epithelial) in terms of stimulation of their migration/invasion, matrix remodelling and induction of angiogenesis. It also functions as an autocrine survival factor for the angiogenic endothelium. MSF expression by foetal and cancer patient cells adherent to an appropriate matrix may be persistently switched off by a transient exposure to TGF-beta1; conversely, MSF expression by adult dermal fibroblasts adherent to other matrices may be persistently switched on by a transient exposure to TGF-beta or various pharmacological agents known to alter gene expression by epigenetic mechanisms. The manifestation of MSF effects on target cells is similarly dependent on the inter-dependent signalling of soluble factors and matrix molecules. The significant association between elevated MSF expression and poor survival in patients with breast and oral cancer suggests that MSF may function as a driver of tumour progression. Accordingly, we suggest that the downregulation of MSF expression (eg, by siRNA or pharmacological agents) and/or inhibition of its bioactivities (by function-neutralising antibodies or MSF inhibitors) may provide a clinically efficacious means of improving treatment outcome in cancer patients.

    Topics: Biomarkers; Cell Movement; Cytokines; Disease Progression; Fibroblasts; Fibronectins; Humans; Neoplasms; Neovascularization, Pathologic; Protein Isoforms; Transforming Growth Factor beta

2010
Cellular context-dependent "colors" of transforming growth factor-beta signaling.
    Cancer science, 2010, Volume: 101, Issue:2

    Transforming growth factor (TGF)-beta signaling has interesting characteristics in the context of cancer. Although perturbations of TGF-beta signaling are strongly implicated in cancer progression, TGF-beta signaling has both tumor-suppressive and tumor-promoting effects. For example, TGF-beta inhibits cancer cell proliferation in some cellular contexts, but promotes it in others. Although several approaches to treating cancer have been considered using TGF-beta-based therapeutic strategies, the contradictory behaviors of TGF-beta have made these approaches complex. To put them to practical use, either the tumor-suppressive or tumor-promoting arm needs to be specifically manipulated. However, there is virtually no method to specifically regulate a certain cell response induced by TGF-beta. In this review, we first consider the basic machinery of TGF-beta signaling, and describe several cell responses induced by TGF-beta stimulation in specific contexts. Mechanisms by which TGF-beta can induce several responses in a cellular context-dependent fashion are discussed with established paradigms and models. We also address perspectives on the specific control of only a subset of numerous cell responses induced by TGF-beta stimulation. Such methods will aid specific regulation of either the tumor-suppressive or tumor-promoting arm of the TGF-beta pathway and in realization of TGF-beta-based treatment of malignant tumors.

    Topics: Animals; Humans; MicroRNAs; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2010
Emerging roles of deubiquitinases in cancer-associated pathways.
    IUBMB life, 2010, Volume: 62, Issue:2

    Deubiquitinases (DUBs) are emerging as important regulators of many pathways germane to cancer. They may regulate the stability of key oncogenes, exemplified by USP28 stabilisation of c-Myc. Alternatively they can negatively regulate ubiquitin-dependent signalling cascades such as the NF-kappaB activation pathway. We review the current literature that associates DUBs with cancer and discuss their suitability as drug targets of the future.

    Topics: Animals; Carboxypeptidases; Carcinoma, Adenoid Cystic; Cell Proliferation; Deubiquitinating Enzyme CYLD; Endopeptidases; Fanconi Anemia; Fatty Acid Synthases; Forkhead Transcription Factors; Genes, myc; Humans; Male; Neoplasms; NF-kappa B; Oncogenes; Peptide Hydrolases; Prostatic Neoplasms; PTEN Phosphohydrolase; Receptor Protein-Tyrosine Kinases; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Tumor Suppressor Proteins; Ubiquitin; Ubiquitin Thiolesterase; Ubiquitin-Protein Ligases; Ubiquitin-Specific Peptidase 7; Ubiquitin-Specific Proteases; Wnt Proteins

2010
Integrin-TGF-beta crosstalk in fibrosis, cancer and wound healing.
    EMBO reports, 2010, Volume: 11, Issue:2

    Accumulating evidence indicates that there is extensive crosstalk between integrins and TGF-beta signalling. TGF-beta affects integrin-mediated cell adhesion and migration by regulating the expression of integrins, their ligands and integrin-associated proteins. Conversely, several integrins directly control TGF-beta activation. In addition, a number of integrins can interfere with both Smad-dependent and Smad-independent TGF-beta signalling in different ways, including the regulation of the expression of TGF-beta signalling pathway components, the physical association of integrins with TGF-beta receptors and the modulation of downstream effectors. Reciprocal TGF-beta-integrin signalling is implicated in normal physiology, as well as in a variety of pathological processes including systemic sclerosis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease and cancer; thus, integrins could provide attractive therapeutic targets to interfere with TGF-beta signalling in these processes.

    Topics: Animals; Disease Progression; Fibrosis; Humans; Integrins; Models, Biological; Neoplasms; Receptor Cross-Talk; Transforming Growth Factor beta; Wound Healing

2010
Growth factors as active participants in carcinogenesis: a perspective.
    Veterinary pathology, 2010, Volume: 47, Issue:1

    Growth factors are low molecular peptides active in the stimulation of cell proliferation and in the regulation of embryonic development and cellular differentiation. Significant progress has been made in developing effective strategies to treat human malignancies with new chemical compounds based on a rationale directed against various components of signaling pathways. Many of these drugs target a growth factor receptor--for instance, in the form of monoclonal antibodies or inhibitors of tyrosine kinases, such as monoclonal antibodies against epidermal growth factor receptors used in treating certain types of breast cancer. Imatinib mesylate [Gleevec]) is an excellent example of mediators of signal transduction, such as tyrosine kinases. Growth factors proper are used to ameliorate various and sometimes fatal side effects of cytotoxic and/or myelosuppressive chemotherapy. Basic characteristics of several growth families are discussed with therapeutic modalities based on growth factor activity or, more often, inhibition of such activity.

    Topics: Animals; Antineoplastic Agents; Epidermal Growth Factor; Fibroblast Growth Factors; Humans; Intercellular Signaling Peptides and Proteins; Neoplasms; Platelet-Derived Growth Factor; Progranulins; Receptors, Growth Factor; Signal Transduction; Somatomedins; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2010
Smad4-mediated TGF-beta signaling in tumorigenesis.
    International journal of biological sciences, 2010, Jan-01, Volume: 6, Issue:1

    Transforming growth factor-beta (TGF-beta) family members exert their function via specific type I and type II serine/threonine kinase receptors and intracellular Smad transcription factors, including the common mediator Smad4. The dual effects of TGF-beta signaling on tumor initiation and progression are cell-specific and yet to be determined under distinct contexts. A number of genetically manipulated mouse models with alterations in the TGF-beta pathway genes, particularly the pivotal Smad4, revealed that these genes play crucial functions in maintaining tissue homeostasis and suppressing tumorigenesis. Loss of Smad4 plays a causal role in initiating squamous cell carcinomas of skin and upper digestive tract as well as adenocarcinomas of gastrointestinal tract. However, for some cancers like pancreatic and cholangiocellular carcinomas, Smad4 deficiency does not initiate the tumorigenesis but acts as a promoter to accelerate or synergize the development and progression of cancers that are started by other oncogenic pathways. Intriguingly, emerging evidences from mouse models have highlighted the important roles of non-cell autonomous effects of Smad4-mediated TGF-beta signaling in the inhibition of oncogenesis. All these data have greatly deepened our understanding of molecular mechanisms of cell-autonomous and non-cell autonomous effect of Smad4-mediated TGF-beta signaling in suppressing carcinogenesis, which may facilitate the development of successful therapies targeting TGF-beta signaling for the treatment of human cancers.

    Topics: Animals; Humans; Neoplasms; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta

2010
Intravital imaging illuminates transforming growth factor beta signaling switches during metastasis.
    Cancer research, 2010, May-01, Volume: 70, Issue:9

    Transforming growth factor beta (TGFbeta) has seemingly contradictory roles in tumor progression: it can promote metastatic invasion but also act as a tumor suppressor. Recently, two studies have used intravital imaging to unravel the role of TGFbeta at different stages of the metastatic process. TGFbeta promotes single cell motility, which enables invasion into blood vessels. However the activation of TGFbeta signaling is a transient event and is not maintained at distant sites. The downregulation of TGFbeta signaling at secondary sites then permits growth of secondary tumors. In the absence of TGFbeta, cells are restricted to collective movement and lymphatic spread. Here, we discuss these findings and their potential implications.

    Topics: Animals; Cell Movement; Humans; Molecular Imaging; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2010
TGFbeta and cancer metastasis: an inflammation link.
    Cancer metastasis reviews, 2010, Volume: 29, Issue:2

    Dysregulated transforming growth factor beta (TGFbeta) signaling is observed in a variety of human cancers. TGFbeta is produced in large quantities by many tumor types and is known to be pro-oncogenic. Therapeutic strategies directed against TGFbeta signaling using neutralizing antibodies and small molecular inhibitors have been developed. However, TGFbeta is also found to function as a tumor suppressor. This switch from a tumor suppressor in premalignant stages of tumorigenesis to a tumor promoter in later stages of the disease poses great challenges in TGFbeta-targeted cancer therapy. It remains unclear what mechanisms underlie the dual role of TGFbeta and what factors mediate the switch. In the past, most work on dissecting underlying mechanisms was focused on differential regulation of signaling pathways by tumor cell autonomous TGFbeta signaling. Recent progress in elucidating TGFbeta effects on host immune/inflammatory reactions in the tumor microenvironment and distant organs brings exciting new perspectives to the field.

    Topics: Genes, Tumor Suppressor; Humans; Inflammation; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2010
TGFbeta, a potent regulator of tumor microenvironment and host immune response, implication for therapy.
    Current molecular medicine, 2010, Volume: 10, Issue:4

    Alterations in TGFbeta signaling are common in human cancers. TGFbeta has significant impact on tumor initiation and progression. Therapeutic strategies including neutralizing antibodies and small molecular inhibitors have been developed to target TGFbeta signaling. However, TGFbeta can work as both a tumor suppressor and a tumor promoter. A significant challenge to the development of successful TGFbeta antagonism treatment is understanding how and when TGFbeta switches its function from a tumor suppressor to a tumor promoter. Recent studies demonstrate that TGFbeta regulates the infiltration of inflammatory cells and cancer associated fibroblasts into the tumor microenvironment, resulting in changes in signaling cascade in tumor cells. Additionally, TGFbeta exerts systemic immune suppression and significantly inhibits host tumor immune surveillance. Neutralizing TGFbeta in preclinical mouse models enhances CD8+ T-cell and natural killer cell-mediated anti-tumor immune response. This new understanding of TGFbeta signaling in regulation of tumor microenvironment and immune response may provide useful information, particularly for patient selection and inflammation/immune biomarkers for TGFbeta antagonism therapy in clinical trials.

    Topics: Animals; Carcinogens; Humans; Immune Tolerance; Inflammation; Mice; Models, Biological; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2010
HtrA proteins as targets in therapy of cancer and other diseases.
    Expert opinion on therapeutic targets, 2010, Volume: 14, Issue:7

    The HtrA family proteins are serine proteases that are involved in important physiological processes, including maintenance of mitochondrial homeostasis, apoptosis and cell signaling. They are involved in the development and progression of several pathological processes such as cancer, neurodegenerative disorders and arthritic diseases.. We present characteristics of the human HtrA1, HtrA2 and HtrA3 proteins, with the stress on their function in apoptosis and in the diseases. We describe regulation of the HtrAs' proteolytic activity, focusing on allosteric interactions of ligands/substrates with the PDZ domains, and make suggestions on how the HtrA proteolytic activity could be modified. Literature cited covers years 1996 - 2010.. An overview of the HtrAs' function/regulation and involvement in diseases (cancer, neurodegenerative disorders, arthritis), and ideas how modulation of their proteolytic activity could be used in therapies.. HtrA2 is the best target for cancer drug development. An increase in the HtrAs' proteolytic activity could be beneficial in cancer treatment, by stimulation of apoptosis, anoikis or necrosis of cancer cells, or by modulation of the TGF-beta signaling cascade; modulation of HtrA activity could be helpful in therapy of neurodegenerative diseases and arthritis.

    Topics: Allosteric Regulation; Animals; Anoikis; Antineoplastic Agents; Apoptosis; Arthritis; Drug Design; Enzyme Activation; High-Temperature Requirement A Serine Peptidase 1; High-Temperature Requirement A Serine Peptidase 2; Humans; Ligands; Mitochondrial Proteins; Necrosis; Neoplasms; Neurodegenerative Diseases; PDZ Domains; Serine Endopeptidases; Signal Transduction; Transforming Growth Factor beta

2010
TGFbeta signalling: a complex web in cancer progression.
    Nature reviews. Cancer, 2010, Volume: 10, Issue:6

    The distortion of growth factor signalling is the most important prerequisite in tumour progression. Transforming growth factor-beta (TGFbeta) signalling regulates tumour progression by a tumour cell-autonomous mechanism or through tumour-stroma interaction, and has either a tumour-suppressing or tumour-promoting function depending on cellular context. Such inherent complexity of TGFbeta signalling results in arduous, but promising, assignments for developing therapeutic strategies against malignant tumours. As numerous cellular context-dependent factors tightly maintain the balance of TGFbeta signalling and contribute to the regulation of TGFbeta-induced cell responses, in this Review we discuss how they maintain the balance of TGFbeta signalling and how their collapse leads to tumour progression.

    Topics: Animals; Disease Progression; Humans; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta

2010
Transcriptional crosstalk between TGF-β and stem cell pathways in tumor cell invasion: role of EMT promoting Smad complexes.
    Cell cycle (Georgetown, Tex.), 2010, Jun-15, Volume: 9, Issue:12

    Tumor cells undergoing the epithelial-mesenchymal transition (EMT) acquire the capacity to migrate, invade the stroma and metastasize. EMT cells also acquire stem cell characteristics suggesting crosstalk between EMT and stem cell pathways and contribution of the EMT process to the generation of cancer stem cells. Indeed, transforming growth factor-beta (TGF-β), a major inducer of EMT, cooperates with stem cell pathways like Wnt, Ras, Hedgehog and Notch to induce EMT. A molecular basis for this cooperative signaling is indicated by recent data showing that many EMT associated transcription factors like Snail1, Zeb1/2, Twist, β-catenin, Lef/TCF, Foxc2 and AP-1 interact with Smads and form EMT promoting Smad complexes (EPSC) engaged in both repressing epithelial genes and activating mesenchymal genes. Thus, formation and activation of EPSC seems to represent a point of convergence between EMT and stem cell pathways. Here, we review our current understanding of the mechanisms involved in the transcriptional crosstalk between TGF-β and stem cell pathways and discuss how a fundament for the activation of these mechanisms may lead to the induction of EMT in tumors.

    Topics: Animals; Epithelial-Mesenchymal Transition; Humans; Neoplasm Invasiveness; Neoplasms; Neoplastic Stem Cells; Smad Proteins; Transcription, Genetic; Transforming Growth Factor beta

2010
TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression.
    Trends in immunology, 2010, Volume: 31, Issue:6

    Transforming growth factor beta (TGF-beta) plays an important role in tumor initiation and progression, functioning as both a suppressor and a promoter. The mechanisms underlying this dual role of TGF-beta remain unclear. TGF-beta exerts systemic immune suppression and inhibits host immunosurveillance. Neutralizing TGF-beta enhances CD8+ T-cell- and NK-cell-mediated anti-tumor immune responses. It also increases neutrophil-attracting chemokines resulting in recruitment and activation of neutrophils with an antitumor phenotype. In addition to its systemic effects, TGF-beta regulates infiltration of inflammatory/immune cells and cancer-associated fibroblasts in the tumor microenvironment causing direct changes in tumor cells. Understanding TGF-beta regulation at the interface of tumor and host immunity should provide insights into developing effective TGF-beta antagonists and biomarkers for patient selection and efficacy of TGF-beta antagonist treatment.

    Topics: Animals; Disease Progression; Humans; Killer Cells, Natural; Neoplasms; Signal Transduction; T-Lymphocytes; Transforming Growth Factor beta

2010
Signaling from membrane receptors to tumor suppressor WW domain-containing oxidoreductase.
    Experimental biology and medicine (Maywood, N.J.), 2010, Volume: 235, Issue:7

    The family of WW domain-containing proteins contains over 2000 members. The small WW domain module is responsible, in part, for protein/protein binding interactions and signaling. Many of these proteins are located at the membrane/cytoskeleton area, where they act as adaptors to receive signals from the cell surface. In this review, we provide molecular insights regarding recent novel findings on signaling from the cell surface toward WW domain-containing oxidoreductase, known as WWOX, FOR or WOX1. More specifically, transforming growth factor beta 1 utilizes cell surface hyaluronidase Hyal-2 (hyaluronoglucosaminidase 2) as a cognate receptor for signaling with WWOX and Smad4 to control gene transcription, growth and death. Complement C1q alone, bypassing the activation of classical pathway, signals a novel event of apoptosis by inducing microvillus formation and WWOX activation. Deficiency in these signaling events appears to favorably support cancer growth.

    Topics: Animals; Cell Adhesion Molecules; Complement C1q; GPI-Linked Proteins; Humans; Hyaluronoglucosaminidase; Neoplasms; Oxidoreductases; Receptors, Cytoplasmic and Nuclear; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta; Tumor Suppressor Proteins; WW Domain-Containing Oxidoreductase

2010
Divergent molecular mechanisms underlying the pleiotropic functions of macrophage inhibitory cytokine-1 in cancer.
    Journal of cellular physiology, 2010, Volume: 224, Issue:3

    Multifunctional macrophage inhibitory cytokine-1, MIC-1, is a member of the transforming growth factor-beta (TGF-beta) superfamily that plays key roles in the prenatal development and regulation of the cellular responses to stress signals and inflammation and tissue repair after acute injuries in adult life. The stringent control of the MIC-1 expression, secretion, and functions involves complex regulatory mechanisms and the interplay of other growth factor signaling networks that control the cell behavior. The deregulation of MIC-1 expression and signaling pathways has been associated with diverse human diseases and cancer progression. The MIC-1 expression levels substantially increase in cancer cells, serum, and/or cerebrospinal fluid during the progression of diverse human aggressive cancers, such as intracranial brain tumors, melanoma, and lung, gastrointestinal, pancreatic, colorectal, prostate, and breast epithelial cancers. Of clinical interest, an enhanced MIC-1 expression has been positively correlated with poor prognosis and patient survival. Secreted MIC-1 cytokine, like the TGF-beta prototypic member of the superfamily, may provide pleiotropic roles in the early and late stages of carcinogenesis. In particular, MIC-1 may contribute to the proliferation, migration, invasion, metastases, and treatment resistance of cancer cells as well as tumor-induced anorexia and weight loss in the late stages of cancer. Thus, secreted MIC-1 cytokine constitutes a new potential biomarker and therapeutic target of great clinical interest for the development of novel diagnostic and prognostic methods and/or cancer treatment against numerous metastatic, recurrent, and lethal cancers.

    Topics: Animals; Biomarkers, Tumor; Disease Progression; Female; Gene Expression Regulation, Neoplastic; Growth Differentiation Factor 15; Humans; Neoplasms; Pregnancy; Prognosis; Signal Transduction; Transforming Growth Factor beta

2010
The polarization of immune cells in the tumour environment by TGFbeta.
    Nature reviews. Immunology, 2010, Volume: 10, Issue:8

    Transforming growth factor-beta (TGFbeta) is an immunosuppressive cytokine produced by tumour cells and immune cells that can polarize many components of the immune system. This Review covers the effects of TGFbeta on natural killer (NK) cells, dendritic cells, macrophages, neutrophils, CD8(+) and CD4(+) effector and regulatory T cells, and NKT cells in animal tumour models and in patients with cancer. Collectively, many recent studies favour the hypothesis that blocking TGFbeta-induced signalling in the tumour microenvironment enhances antitumour immunity and may be beneficial for cancer therapy. An overview of the current drugs and reagents available for inhibiting TGFbeta-induced signalling and their phase in clinical development is also provided.

    Topics: Animals; Antineoplastic Agents; Cell Polarity; Cytokines; Dendritic Cells; Hematopoiesis, Extramedullary; Humans; Immunosuppression Therapy; Killer Cells, Natural; Macrophages; Neoplasms; Neutrophils; Signal Transduction; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2010
The Yin and Yang of bone morphogenetic proteins in cancer.
    Cytokine & growth factor reviews, 2010, Volume: 21, Issue:4

    Bone morphogenetic proteins (BMPs) were first studied as growth factors or morphogens of the transforming growth factor-beta superfamily. These growth molecules, originally associated with bone and cartilage development, are now known to play an important role in morphogenesis and homeostasis in many other tissues. More recently, significant contributions from BMPs, their receptors, and interacting molecules have been linked to carcinogenesis and tumor progression. On the other hand, BMPs can sometimes function as a tumor suppressor. Our report highlights these new roles in the pathogenesis of cancer that may suggest novel targets for therapeutic intervention.

    Topics: Bone Morphogenetic Protein Receptors; Bone Morphogenetic Proteins; Disease Progression; Humans; Mutation; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2010
The crosstalk of RAS with the TGF-β family during carcinoma progression and its implications for targeted cancer therapy.
    Current cancer drug targets, 2010, Volume: 10, Issue:8

    Both RAS and transforming growth factor (TGF)-β signaling cascades are central in tumorigenesis and show synergisms depending on tumor stage and tissue context. In this review we focus on the interaction of RAS subeffector proteins with signaling components of the TGF-β family including those of TGF-βs, activins and bone morphogenic proteins. Compelling evidence indicates that RAS signaling is essentially involved in the switch from tumor-suppressive to tumor-promoting functions of the TGF-β family leading to enhanced cancer growth and metastatic dissemination of primary tumors. Thus, the interface of these signaling cascades is considered as a promising target for the development of novel cancer therapeutics. The current pharmacological anti-cancer concepts combating the molecular cooperation between RAS and TGF-β family signaling during carcinoma progression are critically discussed.

    Topics: Antineoplastic Agents; Clinical Trials as Topic; Humans; Neoplasms; ras Proteins; Signal Transduction; Transforming Growth Factor beta

2010
Integrin signalling adaptors: not only figurants in the cancer story.
    Nature reviews. Cancer, 2010, Volume: 10, Issue:12

    Current evidence highlights the ability of adaptor (or scaffold) proteins to create signalling platforms that drive cellular transformation upon integrin-dependent adhesion and growth factor receptor activation. The understanding of the biological effects that are regulated by these adaptors in tumours might be crucial for the identification of new targets and the development of innovative therapeutic strategies for human cancer. In this Review we discuss the relevance of adaptor proteins in signalling that originates from integrin-mediated cell-extracellular matrix (ECM) adhesion and growth factor stimulation in the context of cell transformation and tumour progression. We specifically underline the contribution of p130 Crk-associated substrate (p130CAS; also known as BCAR1), neural precursor cell expressed, developmentally down-regulated 9 (NEDD9; also known as HEF1), CRK and the integrin-linked kinase (ILK)-pinch-parvin (IPP) complex to cancer, along with the more recently identified p140 Cas-associated protein (p140CAP; also known as SRCIN1).

    Topics: Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Animals; Apoptosis; Cell Movement; Humans; Integrins; Neoplasm Invasiveness; Neoplasms; Phosphoproteins; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-crk; Receptor, ErbB-2; Transforming Growth Factor beta

2010
[Tumor permeability of nanocarriers observed by dynamic contrast-enhanced magnetic resonance imaging].
    Yakugaku zasshi : Journal of the Pharmaceutical Society of Japan, 2010, Volume: 130, Issue:12

    The structure of tumor vasculature is crucial for the nanocarrier-mediated chemotherapy. Recently, transforming growth factor-β (TGF-β) inhibitor was reported to increase the tumor accumulation of nanocarriers by changing the structure of tumor vasculature. To identify the parameters of tumor vasculature function following TGF-β inhibitor (A-83-01) treatment, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was performed using Gd-DTPA and its liposomal formulation (Gd-L) as contrast agents. Observation of tumor MR image before, during, and after injection of contrast agent could calculate the parameters of vascular function, such as volume transfer constant between blood plasma and extracellular space (K(trans)) and fractional plasma volume (v(p)). A-83-01 treatment significantly increased these parameters within 24 h that was positively related to pericyte coverage and tumor cell proliferation. Furthermore, apparent diffusion coefficient (ADC) determined by diffusion-weighed imaging was decreased by A-83-01 treatment, suggesting the decrease of tumor interstitial fluid pressure. Vascular function of the tumor improved by A-83-01 treatment well assessed on post-Gd-L-enhanced MR images, which predicted delivery of liposomal drug to the tumor. These findings suggest that DCE-MRI and, in particular, K(trans) and v(p) quantitation, provide important additional information about tumor vasculature by A-83-01 treatment.

    Topics: Animals; Capillary Permeability; Contrast Media; Doxorubicin; Gadolinium DTPA; Humans; Liposomes; Magnetic Resonance Imaging; Mice; Neoplasms; Pyrazoles; Stimulation, Chemical; Thiocarbamates; Thiosemicarbazones; Transforming Growth Factor beta

2010
Pharmacological inhibition of TGFβ as a strategy to augment the antitumor immune response.
    Current opinion in investigational drugs (London, England : 2000), 2010, Volume: 11, Issue:12

    There is considerable evidence suggesting that a variety of malignancies utilize the TGFβ cytokine to evade immune surveillance mechanisms to facilitate tumor growth and metastatic progression. The recently developed large- and small-molecule TGFβ inhibitors have demonstrated antitumor efficacy in several preclinical tumor models. Further investigation has revealed these agents to be critically dependent upon the host's immune system, suggesting that the inhibition of TGFβ may overcome the immunosuppressive tumor microenvironment and, ultimately, augment the antitumor immune response. These findings strongly support combining this strategy with other immunotherapeutic approaches for the treatment of metastatic cancer. This review discusses the immunoregulatory and antitumor properties of these pharmacological inhibitors of TGFβ signaling as either independent agents or in combination with various immunotherapeutic strategies, their potential side effects, as well as additional avenues of research that may be necessary for their eventual clinical application.

    Topics: Antineoplastic Agents; Antineoplastic Combined Chemotherapy Protocols; Humans; Immunotherapy; Molecular Weight; Neoplasms; Transforming Growth Factor beta

2010
Ski and SnoN, potent negative regulators of TGF-beta signaling.
    Cell research, 2009, Volume: 19, Issue:1

    Ski and the closely related SnoN were discovered as oncogenes by their ability to transform chicken embryo fibroblasts upon overexpression. While elevated expressions of Ski and SnoN have also been reported in many human cancer cells and tissues, consistent with their pro-oncogenic activity, emerging evidence also suggests a potential anti-oncogenic activity for both. In addition, Ski and SnoN have been implicated in regulation of cell differentiation, especially in the muscle and neuronal lineages. Multiple cellular partners of Ski and SnoN have been identified in an effort to understand the molecular mechanisms underlying the complex roles of Ski and SnoN. In this review, we summarize recent findings on the biological functions of Ski and SnoN, their mechanisms of action and how their levels of expression are regulated.

    Topics: Animals; Chickens; DNA-Binding Proteins; Embryonic Development; Humans; Intracellular Signaling Peptides and Proteins; Neoplasms; Proto-Oncogene Proteins; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2009
The potential anti-tumorigenic and anti-metastatic side of the proprotein convertases inhibitors.
    Recent patents on anti-cancer drug discovery, 2009, Volume: 4, Issue:1

    The setup of tumorigenesis processes is generally associated with various events leading to abnormal expression of oncogenes and/or tumor suppressor genes. Recently, the expression and/or activity of a range of molecules involved in these processes were reported to require proteolytic processing of their precursor proteins by the serine pro-protein convertases (PCs) in order to mediate their biological functions. These include adhesion molecules, proteases, growth factors, cytokines and their receptors. Since their discovery, the identification of new PCs substrates and specific PCs inhibitors became an attractive strategy in cancer therapy. In this review, we will report the implication of these enzymes and the processing of their substrates in tumor progression and metastasis. Newly reported studies on the potential use of the PCs as new therapeutic targets will be also discussed.

    Topics: Animals; Antineoplastic Agents; Humans; Neoplasm Metastasis; Neoplasms; Platelet-Derived Growth Factor; Proprotein Convertases; Protease Inhibitors; Proto-Oncogene Proteins c-sis; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2009
Bioactive food components, inflammatory targets, and cancer prevention.
    Cancer prevention research (Philadelphia, Pa.), 2009, Volume: 2, Issue:3

    Various dietary components may modify chronic inflammatory processes at the stage of cytokine production, amplification of nuclear factor-kappaB-mediated inflammatory gene expression, and the release of anti-inflammatory cytokine, transforming growth factor-beta. This review provides a synopsis of the strengths and weaknesses of the evidence that specific bioactive food components influence inflammation-related targets linked to cancer. A target repeatedly surfacing as a site of action for several dietary components is transforming growth factor beta. Whereas the use of dietary intervention strategies offers intriguing possibilities for maintaining normal cell function by modifying a process that is essential for cancer development and progression, more information is needed to characterize the minimum quantity of the bioactive food components required to bring about a change in inflammation-mediated cancer, the ideal time for intervention, and the importance of genetics in determining the response. Unquestionably, the societal benefits of using foods and their components to prevent chronic inflammation and associated complications, including cancer, are enormous.

    Topics: Anti-Inflammatory Agents; Diet; Food; Histone Deacetylases; Humans; Hydroxyprostaglandin Dehydrogenases; Inflammation; Models, Biological; Neoplasms; NF-kappa B; Nutritive Value; Phosphorylation; Signal Transduction; Transforming Growth Factor beta

2009
The TGF-beta paradox in human cancer: an update.
    Future oncology (London, England), 2009, Volume: 5, Issue:2

    TGF-beta plays an essential role in maintaining tissue homeostasis through its ability to induce cell cycle arrest, differentiation and apoptosis, and to preserve genomic stability. Thus, TGF-beta is a potent anticancer agent that prohibits the uncontrolled proliferation of epithelial, endothelial and hematopoietic cells. Interestingly, tumorigenesis typically elicits aberrations in the TGF-beta signaling pathway that engenders resistance to the cytostatic activities of TGF-beta, thereby enhancing the development and progression of human malignancies. Moreover, these genetic and epigenetic events conspire to convert TGF-beta from a suppressor of tumor formation to a promoter of their growth, invasion and metastasis. The dichotomous nature of TGF-beta during tumorigenesis is known as the 'TGF-beta paradox', which remains the most critical and mysterious question concerning the physiopathological role of this multifunctional cytokine. Here we review recent findings that directly impact our understanding of the TGF-beta paradox and discuss their importance to targeting the oncogenic activities of TGF-beta in developing and progressing neoplasms.

    Topics: Cell Transformation, Neoplastic; Humans; Neoplasms; Transforming Growth Factor beta

2009
Transforming growth factor-beta in breast cancer: too much, too late.
    Breast cancer research : BCR, 2009, Volume: 11, Issue:1

    The contribution of transforming growth factor (TGF)beta to breast cancer has been studied from a myriad perspectives since seminal studies more than two decades ago. Although the action of TGFbeta as a canonical tumor suppressor in breast is without a doubt, there is compelling evidence that TGFbeta is frequently subverted in a malignant plexus that drives breast cancer. New knowledge that TGFbeta regulates the DNA damage response, which underlies cancer therapy, reveals another facet of TGFbeta biology that impedes cancer control. Too much TGFbeta, too late in cancer progression is the fundamental motivation for pharmaceutical inhibition.

    Topics: Animals; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2009
Targeting TGFbeta-mediated processes in cancer.
    Current opinion in drug discovery & development, 2009, Volume: 12, Issue:2

    TGFbeta is a pleiotropic cytokine that has regulatory roles during embryonic development and adult tissue homeostasis, and is especially important for epithelial, neural and immune system differentiation and function. An increasing number of literature reports support a role for autocrine and paracrine TGFbeta-mediated processes in cancer. Progress in delineating the effects of TGFbeta in different tumors and stages of cancer has stimulated the development of TGFbeta-targeted therapies. This review describes the causes of TGFbeta malfunction in cancer, the rationale for therapeutic targeting of TGFbeta in human neoplasia, and the molecular strategies to modulate the signal.

    Topics: Animals; Antineoplastic Agents; Cell Proliferation; Drug Design; Humans; Ligands; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Structure-Activity Relationship; Transforming Growth Factor beta

2009
Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor?
    Immunity, 2009, Volume: 30, Issue:5

    Adaptive Foxp3(+)CD4(+) regulatory T (iTreg) cells develop outside the thymus under subimmunogenic antigen presentation, during chronic inflammation, and during normal homeostasis of the gut. iTreg cells are essential in mucosal immune tolerance and in the control of severe chronic allergic inflammation, and most likely are one of the main barriers to the eradication of tumors. The Foxp3(+) iTreg cell repertoire is drawn from naive conventional CD4(+) T cells, whereas natural Treg (nTreg) cells are selected by high-avidity interactions in the thymus. The full extent of differences and similarities between iTreg and nTreg cells is yet to be defined. We speculate that iTreg cell development is driven by the need to maintain a noninflammatory environment in the gut, to suppress immune responses to environmental and food allergens, and to decrease chronic inflammation, whereas nTreg cells prevent autoimmunity and raise the activation threshold for all immune responses.

    Topics: Animals; Cell Differentiation; Dendritic Cells; Forkhead Transcription Factors; Humans; Immune Tolerance; Infections; Inflammation; Interleukin-15; Interleukin-2; Neoplasms; STAT Transcription Factors; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Thymus Gland; Transforming Growth Factor beta; Tretinoin

2009
Transforming growth factor-beta signaling and tumor angiogenesis.
    Frontiers in bioscience (Landmark edition), 2009, 06-01, Volume: 14, Issue:13

    Transforming growth factor-beta (TGF-beta) family members are secreted multifunctional cytokines that play pivotal roles in development and disease. The prototypic member of this family, TGF-beta, plays a dual role in carcinogenesis, acting as a tumor suppressor in early stages and as tumor promoter in late stages of tumor progression. Numerous studies support the notion that pathological angiogenesis is one of the hallmarks of cancer. Tumor angiogenesis is regulated by a network of growth factors, including members of the TGF-beta family. TGF-beta acts in a context-dependent manner and can either stimulate or inhibit tumor angiogenesis. In this review, we discuss our current understanding on how TGF-beta family members affect endothelial and smooth muscle cell function and how perturbed TGF-beta signaling may contribute to tumor angiogenesis and tumor progression.

    Topics: Animals; Bone Morphogenetic Proteins; Endothelial Cells; Humans; Intracellular Signaling Peptides and Proteins; Models, Biological; Myocytes, Smooth Muscle; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2009
Transforming growth factor-beta signaling in epithelial-mesenchymal transition and progression of cancer.
    Proceedings of the Japan Academy. Series B, Physical and biological sciences, 2009, Volume: 85, Issue:8

    Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that induces growth arrest, tissue fibrosis, and epithelial-mesenchymal transition (EMT) through activation of Smad and non-Smad signaling pathways. EMT is the differentiation switch by which polarized epithelial cells differentiate into contractile and motile mesenchymal cells. Cell motility and invasive capacity are activated upon EMT. Multiple transcription factors, including deltaEF1/ZEB1, SIP1/ZEB2, and Snail/SNAI1, are induced by TGF-beta-Smad signaling and play critical roles in TGF-beta-induced EMT. In addition, both non-Smad signaling activated by TGF-beta and cross-talk with other signaling pathways play important roles in induction of EMT. Of these, Ras signaling synergizes with TGF-beta-Smad signaling, and plays an important role in the induction of EMT. TGF-beta inhibitors prevent invasion and metastasis of advanced cancer through multiple mechanisms, including inhibition of EMT. The discovery of molecules that inhibit TGF-beta-induced EMT but not TGF-beta-induced growth arrest may be an ideal strategy for treatment of invasion and metastasis of cancer.

    Topics: Animals; Disease Progression; Epithelial Cells; Humans; Mesoderm; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2009
CLU "in and out": looking for a link.
    Advances in cancer research, 2009, Volume: 105

    Cancer cells need to interact synergistically with their surrounding microenvironment to form a neoplasm and to progress further to colonize distant organs. The microenvironment can exert profound epigenetic effects on cells through cell-derived interactions between cells, or through cell-derived factors deposited into the microenvironment. Tumor progression implies immune-escaping and triggers several processes that synergistically induce a cooperation among transformed and stromal cells, that compete for space and resources such as oxygen and nutrients. Therefore, the extra cellular milieu and tissue microenvironment heterotypic interactions cooperate to promote tumor growth, angiogenesis, and cancer cell motility, through elevated secretion of pleiotropic cytokines and soluble factors. Clusterin (CLU), widely viewed as an enigmatic protein represents one of the numerous cellular factors sharing the intracellular information with the microenvironment and it has also a systemic diffusion, tightly joining the "In and the Out" of the cell with a still debated variety of antagonistic functions. The multiplicity of names for CLU is an indication of the complexity of the problem and could reflect, on one hand its multifunctionality, or alternatively could mask a commonality of function. The posited role for CLU, further supported as a cytoprotective prosurvival chaperone-like molecule, seems compelling, in contrast its tumor suppressor function, as a guide of the guardians of the genome (DNA-repair proteins Ku70/80, Bax cell death inducer), could really reflect the balanced expression of its different forms, most certainly depending on the intra- and extracellular microenvironment cross talk. The complicated balance of cytokines network and the regulation of CLU forms production in cancer and stromal cells undoubtedly represent a potential link among adaptative responses, genomic stability, and bystander effect after oxidative stresses and damage. This review focuses on the tumor-microenvironment interactions strictly involved in controlling local cancer growth, invasion, and distant metastases that play a decisive role in the regulation of CLU different forms expression and release. In addition, we focus on the pleiotropic action of the extracellular form of this protein, sCLU, that may play a crucial role in redirecting stromal changes, altering intercellular communications binding cell surface receptors and contributing to influence the secretion of chemok

    Topics: Apoptosis; Clusterin; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Interleukin-6; Neoplasms; Phosphatidylinositol 3-Kinases; Platelet-Derived Growth Factor; Proto-Oncogene Proteins c-akt; Signal Transduction; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2009
Role of transforming growth factor beta in cancer microenvironment.
    Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 2009, Volume: 11, Issue:11

    Transforming growth factor Beta (TGF-Beta) family members are polypeptidic cytokines with pleiotropic physiological properties. In relation to cancer, TGF-Beta exerts a dual tumour-suppressive and oncogenic effect, which is largely dependent on microenvironment stimuli. After activation of TGF-Beta signalling, two pathways can be activated: the canonical one through the mammalian Smad family or the non-canonical one activating, among others, the cellular mitogen-activated protein kinase (MAPK) signalling downstream, which interacts with Smad signalling. During tumorigenesis, cells of many cancer types often lose their response to the tumour-suppressive effects of TGF-Beta, which, in turn, has the opposite effect, acting as an autocrine tumour-promoting factor. In this review, we summarise the current knowledge about this intriguing cytokine, with special emphasis on its immunosuppressive actions.

    Topics: Adaptive Immunity; Animals; Cell Lineage; Cytokines; Gene Expression Regulation, Neoplastic; Humans; Immunity, Innate; Immunosuppressive Agents; Models, Biological; Neoplasms; Signal Transduction; Smad Proteins; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2009
Strategies for TGF-beta modulation: a review of recent patents.
    Expert opinion on therapeutic patents, 2009, Volume: 19, Issue:12

    TGF-beta has been identified as a key factor in the progression of various diseases, in particular cancer and fibrosis. The signaling of TGF-beta can be modulated through three distinct strategies: using antisense nucleotides that block TGF-beta mRNA (trabedersen (AP 12009)), using monoclonal antibodies to block TGF-beta isoforms (lerdelimumab, metelimumab) or using small molecule inhibitors of the TGF-beta receptor 1 (TGF-betaR1 or ALK-5).. This review focuses on small molecules and summarizes the most recent TGF-betaR1 inhibitors reported in the patent literature.. We searched and analyzed the patent literature claiming chemical matter for TGF-betaR1 inhibition from the 1(st) of January 2005 to the 1(st) of January 2009.. The inhibition of TGF-beta has recently been clinically validated with antisense nucleotide trabedersen. Small molecules inhibitors of TGF-betaR1 that are now in Phase I clinical trials and in preclinical stage are, therefore, of high interest and could provide a more versatile route to TGF-beta modulation through oral dosing while maintaining the same therapeutic benefits.

    Topics: Animals; Clinical Trials as Topic; Drug Evaluation, Preclinical; Fibrosis; Humans; Neoplasms; Patents as Topic; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2009
A comparative analysis and systematic review of the wound-healing milieu: implications for body contouring after massive weight loss.
    Plastic and reconstructive surgery, 2009, Volume: 124, Issue:5

    Wound-healing complications following body contouring for massive weight loss patients are significant, with rates exceeding 40 percent. To better understand aberrant healing in this population, the authors have performed a comparative analysis of the wound milieu literature for patient populations with similar complication rates.. PubMed and Ovid databases were reviewed from January of 1985 to January of 2009 for key terms, including wound healing, obesity, cancer, burn, transplant, and body contouring. Serum and wound levels of multiple factors, including matrix metalloproteinases (MMPs) and cytokines, were assessed.. Complication rates in body contouring surgery range from 31 to 66 percent. Sixty-five studies were reviewed, and wound-healing complication rates were identified for cancer (45.8 percent), burn (30.4 percent), posttransplant (36 percent), and obese (43 percent) populations. In these groups, matrix metalloproteinases and tissue inhibitors of metalloproteinase (TIMPs) help regulate wound repair. Matrix metalloproteinase levels were elevated in cancer (4-fold increase in MMP-2), burn (20- to 30-fold increase in MMP-9), transplant (1.4-fold increase in MMP-2), and obese/chronic (79-fold increase) populations. TIMPs were increased in cancer (1.9-fold increase in TIMP-2) and burn (1.4-fold increase in TIMP-1) patients but decreased in chronic wound (55-fold decrease in TIMP-1) populations. Alterations to these regulatory proteins lead to prolonged matrix degradation, up-regulation of inflammatory mediators, and decreased growth factors, delaying the wound-healing process.. Complications after body contouring surgery are likely multifactorial; however, molecular imbalances to the massive weight loss wound milieu may contribute to poor surgical outcomes. Examining wound regulatory proteins including transforming growth factor-beta, vascular endothelial growth factor, and matrix metalloproteinases could aid in understanding the healing difficulties observed clinically.

    Topics: Bariatric Surgery; Biomarkers; Burns; Humans; Matrix Metalloproteinase 2; Matrix Metalloproteinase 3; Matrix Metalloproteinase 9; Neoplasms; Obesity; Organ Transplantation; Plastic Surgery Procedures; Tissue Inhibitor of Metalloproteinase-1; Tissue Inhibitor of Metalloproteinase-2; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A; Weight Loss; Wound Healing

2009
The soluble transforming growth factor-beta receptor: advantages and applications.
    The international journal of biochemistry & cell biology, 2009, Volume: 41, Issue:3

    Transforming growth factor-beta (TGF-beta) is a cytokine that plays a pivotal role in growth, differentiation, development, immune response and wound healing. TGF-beta is upregulated following wound infliction and inflammation, and plays an important role in the production of extracellular matrix proteins that contribute to tissue repair. However, in some diseases, TGF-beta dysregulation can lead to tumor formation, organ fibrosis and the disruption of organ function. A number of molecules have been designed to counteract the effects of TGF-beta, including anti-TGF-beta monoclonal antibodies and various small molecules. Here we discuss the design, use and advantages of the highly specific TGF-beta binding molecule, the soluble human TGF-beta receptor (sTbetaR.Fc) as a TGF-beta sequestering agent.

    Topics: Animals; Antibodies, Monoclonal; Cell Differentiation; Cell Proliferation; Extracellular Matrix; Fibrosis; Humans; Immunity, Innate; Immunotherapy; Neoplasms; Receptors, IgG; Recombinant Proteins; Signal Transduction; Transforming Growth Factor beta; Wound Healing

2009
Cytokine regulation networks in the cancer microenvironment.
    Frontiers in bioscience : a journal and virtual library, 2008, May-01, Volume: 13

    During carcinoma formation, cancer cells release various cytokines and growth factors into their surroundings and recruit and reprogram many other types of cells in order to establish a tumor microenvironment. Consequently, the tumor tissues almost always contain a large number of endothelial cells, fibroblasts, and infiltrating inflammatory cells that in turn produce a variety of cytokines. The cytokines produced by these cells have been posited as key factors in modulating immune response either against or in favor of tumorigenesis in the microenvironment. The interactions that take place between immune and cancer cells are complex, involving multiple cascades of cytokines, chemokines, and/or growth factors. In this review, we address the essential pro- and anti-tumorigenic roles of cytokines in the tumor microenvironment. As the interaction of cytokines, growth factors, and cancer cells forms a comprehensive network at the tumor site that is then responsible for the overall progression or rejection of the tumor, the current review links the microenvironment-derived cytokines and growth factors to a number of different kinds of human carcinogenesis models. Multifunctional cytokines, extracellular matrix mediators, and regulatory cytokines in the cancer environment are all shown to be key factors in the different cancer immune-editing systems. The characterization of cytokine networks in various types of cancer cells may yield important information for understanding the immune-related mechanisms of cancer development, and this knowledge may have subsequent application in cancer immunotherapy.

    Topics: Chemokine CXCL12; Cytokines; Dendritic Cells; Humans; Interferons; Neoplasms; Signal Transduction; TNF-Related Apoptosis-Inducing Ligand; Transforming Growth Factor beta

2008
SnoN in TGF-beta signaling and cancer biology.
    Current molecular medicine, 2008, Volume: 8, Issue:4

    The Transforming Growth Factor (TGF)- beta-Smad signaling pathway regulates diverse biological processes essential for normal development and homeostasis. The Smad-interacting transcriptional modulator SnoN and its related homologs have emerged as important modulators of TGF-beta signaling and responses. SnoN forms a physical complex with the TGF-beta-regulated Smad2/Smad3 and co-Smad4 proteins and either represses or stimulates TGF-beta-induced Smad-dependent transcription in a cell- and promoter-specific manner. In addition, the TGF-beta-activated Smads recruit several ubiquitin ligases to SnoN and thereby promote the ubiquitination and consequent degradation of SnoN. Additional modifications of SnoN, including sumoylation, may contribute to the regulation of SnoN function and its role in TGF-beta signaling. Collectively, these studies suggest that SnoN function is intimately linked to the TGF-beta-Smad pathway in cellular signaling. Although the mechanisms by which SnoN modulates signaling in the TGF-beta-Smad pathway are beginning to be characterized, the full range of SnoN functions and underlying mechanisms in normal development and disease processes remains to be elucidated.

    Topics: Animals; Cell Differentiation; Cell Proliferation; Humans; Neoplasms; Signal Transduction; Smad Proteins; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta

2008
Itch: a HECT-type E3 ligase regulating immunity, skin and cancer.
    Cell death and differentiation, 2008, Volume: 15, Issue:7

    The HECT-type E3 ubiquitin ligase (E3) Itch is absent in the non-agouti-lethal 18H or Itchy mice, which develop a severe immunological disease, including lung and stomach inflammation and hyperplasia of lymphoid and hematopoietic cells. The involvement of Itch in multiple signaling pathways and pathological conditions is presently an area of extensive scientific interest. This review aims to bring together a growing body of work exploring Itch-regulated biological processes, and to highlight recent discoveries on the regulatory mechanisms modulating its catalytic activity and substrate recognition capability. Our contribution is also an endeavor to correlate Itch substrate specificity with the pathological defects manifested by the mutant Itchy mice.

    Topics: Animals; Cell Death; ErbB Receptors; Immune System; Keratinocytes; Mice; Mice, Mutant Strains; Neoplasms; Phosphorylation; Protein Transport; Receptors, Chemokine; Repressor Proteins; Signal Transduction; Skin; Substrate Specificity; Transforming Growth Factor beta; TRPC Cation Channels; Ubiquitin; Ubiquitin-Protein Ligases

2008
p53 brings a new twist to the Smad signaling network.
    Science signaling, 2008, Jul-01, Volume: 1, Issue:26

    Transforming growth factor beta (TGF-beta) signaling regulates a plethora of cellular responses, including specification of developmental fate during embryogenesis, cell proliferation, differentiation, and apoptosis. Components of this pathway are often mutated in cancers and other human disorders. TGF-beta signaling involves activation of transcriptional regulators of the Smad family. The tumor suppressor p53 is an essential partner of Smads, affecting TGF-beta signaling at various points in the pathway. Inactivation of p53 may contribute to the aberrant behavior of cancer cells that escape the cytostatic action of TGF-beta despite the apparent integrity of the TGF-beta receptor or Smads. Thus, the discovery that p53 and TGF-beta cooperate in cell-fate decisions and cellular homeostatic mechanisms has important pathophysiological implications.

    Topics: Animals; Apoptosis; Cell Differentiation; Humans; Mutation; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2008
TGFbeta in Cancer.
    Cell, 2008, Jul-25, Volume: 134, Issue:2

    The transforming growth factor beta (TGFbeta) signaling pathway is a key player in metazoan biology, and its misregulation can result in tumor development. The regulatory cytokine TGFbeta exerts tumor-suppressive effects that cancer cells must elude for malignant evolution. Yet, paradoxically, TGFbeta also modulates processes such as cell invasion, immune regulation, and microenvironment modification that cancer cells may exploit to their advantage. Consequently, the output of a TGFbeta response is highly contextual throughout development, across different tissues, and also in cancer. The mechanistic basis and clinical relevance of TGFbeta's role in cancer is becoming increasingly clear, paving the way for a better understanding of the complexity and therapeutic potential of this pathway.

    Topics: Animals; Disease Progression; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2008
PTP-Pez: a novel regulator of TGFbeta signaling.
    Cell cycle (Georgetown, Tex.), 2008, Volume: 7, Issue:15

    The TGFbetas are a family of pleiotropic cytokines that mediate diverse effects including the regulation of cell cycle progression, apoptosis, tissue remodelling and epithelial-mesenchymal transition (EMT). These diverse effects allow the TGFbetas to play multiple and even opposing roles in different contexts during embryonal development, tissue homeostasis and cancer progression. We recently reported that the protein tyrosine phosphatase Pez is a novel inducer of TGFbeta signaling, regulating EMT and organogenesis in developing zebrafish embryos, and leading to TGFbeta-mediated EMT when overexpressed in vitro in epithelial MDCK cells. A number of mutations in Pez have been shown to be associated with breast and colorectal cancers, although the effect of these mutations on Pez function and their contribution to cancer progression remains unclear. Our finding that Pez regulates TGFbeta signaling is therefore of interest not only in the context of identifying a novel upstream regulator of TGFbeta signaling, but also in implicating the dysregulation of TGFbeta signaling as a possible link between Pez mutation and cancer progression. Here we discuss the implications of our research, in the context of dysregulation of TGFbeta signaling in cancer and other human pathologies.

    Topics: Animals; Disease; Growth and Development; Homeostasis; Humans; Models, Biological; Neoplasms; Protein Tyrosine Phosphatases, Non-Receptor; Signal Transduction; Transforming Growth Factor beta

2008
Decoding the quantitative nature of TGF-beta/Smad signaling.
    Trends in cell biology, 2008, Volume: 18, Issue:9

    How transforming growth factor-beta (TGF-beta) signaling elicits diverse cell responses remains elusive, despite the major molecular components of the pathway being known. We contend that understanding TGF-beta biology requires mathematical models to decipher the quantitative nature of TGF-beta/Smad signaling and to account for its complexity. Here, we review mathematical models of TGF-beta superfamily signaling that predict how robustness is achieved in bone-morphogenetic-protein signaling in the Drosophila embryo, how changes in receptor-trafficking dynamics can be exploited by cancer cells and how the basic mechanisms of TGF-beta/Smad signaling conspire to promote Smad accumulation in the nucleus. These studies demonstrate the power of mathematical modeling for understanding TGF-beta biology.

    Topics: Animals; Body Patterning; Bone Morphogenetic Proteins; Cell Nucleus; Dimerization; Drosophila; Ligands; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2008
Role of TGF-beta in the tumor stroma.
    Current cancer drug targets, 2008, Volume: 8, Issue:6

    Recent findings have demonstrated that the tumor stroma actively contributes to tumorigenesis. The communication of malignant cells and tumor stromal components is orchestrated in part by a network of growth factors. One of these growth factors is transforming growth factor-beta (TGF-beta), a secreted multifunctional protein that acts in a highly cellular contextual manner. TGF-beta can either stimulate or inhibit the tumor-promoting effects of the different components of the tumor stroma. In this review, we discuss our current understanding on how TGF-beta influences different stromal compartments.

    Topics: Humans; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2008
Cytokine-based transformation of immune surveillance into tumor-promoting inflammation.
    Oncogene, 2008, Oct-06, Volume: 27, Issue:45

    During the last decade, it has become clear that the mammalian immune system is able to recognize and partially suppress nascent tumors. Human T cells specific to oncogenes and onco-fetal antigens are present in human cancer patients and their tumors. At the same time, molecular links between tumor-associated inflammation and tumor progression have been uncovered, providing an explanation for the long recognized epidemiological link between inflammation and cancer. The synopsis of these findings suggests a new interpretation of tumor immunity. It appears that antigen recognition or antigen-specific T-cell expansion at large is not as profoundly impaired in tumor patients as the correct polarization, the survival and the effector function of tumor-infiltrating T cells. This review will focus on pro-inflammatory cytokines likely to contribute to the deregulation of tumor-specific immunity and its consequences.

    Topics: Animals; Cell Proliferation; Cytokines; Humans; Immunologic Surveillance; Inflammation; Interleukin-12; Interleukin-23; Interleukin-6; Models, Biological; Neoplasms; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2008
Emerging role of miR-106b-25/miR-17-92 clusters in the control of transforming growth factor beta signaling.
    Cancer research, 2008, Oct-15, Volume: 68, Issue:20

    Inactivation of the transforming growth factor beta (TGFbeta) tumor suppressor pathway is a main step in the development of a variety of human tumors. The miR-106b-25 and miR-17-92 clusters are emerging as key modulators of TGFbeta signaling in gastrointestinal and other tumors, interfering with cell cycle arrest and apoptosis when overexpressed in cancer cells. Genetic ablation of these microRNAs (miRNAs) reveals their physiologic role in the control of liver and central nervous system apoptosis, supporting the notion that miRNA-based homeostatic mechanisms can be usurped by cancer cells to resist TGFbeta tumor suppression.

    Topics: Animals; Apoptosis; Apoptosis Regulatory Proteins; Bcl-2-Like Protein 11; Cell Cycle Proteins; Cyclin-Dependent Kinase Inhibitor p21; DNA-Binding Proteins; E2F1 Transcription Factor; Genes, myc; Humans; Membrane Proteins; MicroRNAs; Minichromosome Maintenance Complex Component 7; Multigene Family; Neoplasms; Nuclear Proteins; Proto-Oncogene Proteins; RNA Interference; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2008
Signal transduction pathway through activin receptors as a therapeutic target of musculoskeletal diseases and cancer.
    Endocrine journal, 2008, Volume: 55, Issue:1

    Activin, myostatin and other members of the TGF-beta superfamily signal through a combination of type II and type I receptors, both of which are transmembrane serine/threonine kinases. Activin type II receptors, ActRIIA and ActRIIB, are primary ligand binding receptors for activins, nodal, myostatin and GDF11. ActRIIs also bind a subset of bone morphogenetic proteins (BMPs). Type I receptors that form complexes with ActRIIs are dependent on ligands. In the case of activins and nodal, activin receptor-like kinases 4 and 7 (ALK4 and ALK7) are the authentic type I receptors. Myostatin and GDF11 utilize ALK5, although ALK4 could also be activated by these growth factors. ALK4, 5 and 7 are structurally and functionally similar and activate receptor-regulated Smads for TGF-beta, Smad2 and 3. BMPs signal through a combination of three type II receptors, BMPRII, ActRIIA, and ActRIIB and four type I receptors, ALK1, 2, 3, and 6. BMPs activate BMP-specific Smads, Smad1, 5 and 8. Smad proteins undergo multimerization with co-mediator Smad, Smad4, and translocated into the nucleus to regulate the transcription of target genes in cooperation with nuclear cofactors. The signal transduction pathway through activin type II receptors, ActRIIA and ActRIIB, with type I receptors is involved in various human diseases. In this review, we discuss the role of signaling through activin receptors as therapeutic targets of intractable neuromuscular diseases, endocrine disorders and cancers.

    Topics: Activin Receptors; Activins; Animals; Antineoplastic Agents; Bone Morphogenetic Proteins; Drug Delivery Systems; Epidermal Growth Factor; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Models, Biological; Musculoskeletal Diseases; Myostatin; Neoplasm Proteins; Neoplasms; Protein Binding; Protein Kinase Inhibitors; Signal Transduction; Transforming Growth Factor beta

2008
CCN2, the connective tissue growth factor.
    Journal of molecular medicine (Berlin, Germany), 2008, Volume: 86, Issue:1

    Topics: Connective Tissue Growth Factor; Fibrosis; Humans; Neoplasms; Transforming Growth Factor beta

2008
The role of CD4+CD25+ T regulatory cells in autoimmune diseases.
    Clinical reviews in allergy & immunology, 2008, Volume: 34, Issue:3

    Among the several mechanisms that play role in maintaining peripheral self-tolerance is the existence of a unique CD4+CD25+ population of naturally occurring regulatory T (Treg) cells that actively prevent both the activation and the effector function of autoreactive T cells that have escaped different mechanisms of tolerance. Many studies have shown the benefit of targeting this cell population by restoring self-tolerance. Therapies that could possibly increase the suppressive ability of T regulatory cells were proven to improve that course of autoimmune diseases.

    Topics: Animals; Autoimmune Diseases; Forkhead Transcription Factors; Humans; Interleukin-10; Neoplasms; Self Tolerance; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2008
The TGFBeta pathway as a therapeutic target in cancer.
    Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 2008, Volume: 10, Issue:1

    The TGFBeta pathway has recently emerged as a putative therapeutic target against cancer. However, TGFBeta has a complex and dual role in cancer. In normal epithelial cells and early tumours, TGFBeta acts as a tumour suppressor. In contrast, during tumour progression TGFBeta becomes an oncogenic factor inducing proliferation, angiogenesis, invasion and metastasis, as well as suppressing the anti-tumoral immune response. The role of TGFBeta in oncogenesis requires the precise understanding of the TGFBeta pathway in order to design optimal therapeutic approaches and select the patient population that may benefit from an anti-TGFBeta therapy. Here we review the rationale for evaluating TGFBeta signalling inhibitors as cancer therapeutics, and the progress made in the preclinical and clinical testing of anti- TGFBeta compounds.

    Topics: Animals; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2008
[Myeloid-derived suppressor cells - the new mechanism of immunosuppression in cancer].
    Postepy higieny i medycyny doswiadczalnej (Online), 2008, Jan-21, Volume: 62

    In this paper we summarized the current knowledge about the new mechanism of immunosuppression in cancer i.e. the lately discovered myeloid-derived suppresor cells (MDSC). MDSC are produced in bone marrow under the influence of tumor cell derived substances. MDSC show negative effect on T lymphocytes function through arginine depletion and nitric oxide production. In mice MDSC are characterized by Gr-1 and CD11b expression, however in human there are no definitive markers of this subpopulation. Some laboratory experiments in turning back the negative influence of MDSC on immunological system are provided, mainly through the administration of monoclonal antibodies against MDSC or drugs which block their function. The elucidation of MDSC characteristics can allow us to build new immunotherapeutic protocols in the future.

    Topics: Animals; Arginase; Bone Marrow Cells; CD11b Antigen; Humans; Immune Tolerance; Mice; Mice, Inbred BALB C; Myeloid Cells; Neoplasms; Nitric Oxide; T-Lymphocytes; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2008
The 'short' history of regulatory B cells.
    Trends in immunology, 2008, Volume: 29, Issue:1

    The maintenance of tolerance is the sine qua non of a sophisticated regulatory apparatus to prevent or dampen overzealous immune responses. In addition to the ability of B cells to prime and activate the immune system, B cells with regulatory function (Bregs) have been identified in experimental models of autoimmunity, infections, and cancer, supporting the notion that, similar to regulatory T cells (Tregs), Breg-mediated suppression is an important means for the maintenance of peripheral tolerance. This regulatory function appears to be directly mediated by the production of IL-10 and/or TGFbeta and by the ability of B cells to interact with pathogenic T cells to inhibit harmful immune responses. The identification of their existence is of great relevance to the understanding of autoimmune diseases and to the development of new therapeutic strategies.

    Topics: Animals; Autoimmunity; B-Lymphocyte Subsets; B-Lymphocytes; Humans; Immune Tolerance; Interleukin-10; Models, Immunological; Neoplasms; Parasitic Diseases; Transforming Growth Factor beta

2008
Transforming growth factor-beta signaling and ubiquitinators in cancer.
    Endocrine-related cancer, 2008, Volume: 15, Issue:1

    Transforming growth factor-beta (TGF-beta) represents a large family of growth and differentiation factors that mobilize complex signaling networks to regulate cellular differentiation, proliferation, motility, adhesion, and apoptosis. TGF-beta signaling is tightly regulated by multiple complex mechanisms, and its deregulation plays a key role in the progression of many forms of cancer. Upon ligand binding, TGF-beta signals are transduced by Smad proteins, which in turn are tightly dependent on modulation by adaptor proteins such as embryonic liver fodrin, Smad anchor for receptor activation, filamin, and crkl. A further layer of regulation is imposed by ubiquitin-mediated targeting and proteasomal degradation of specific components of the TGF-beta signaling pathway. This review focuses on the ubiquitinators that regulate TGF-beta signaling and the association of these ubiquitin ligases with various forms of cancer. Delineating the role of ubiquitinators in the TGF-beta signaling pathway could yield powerful novel therapeutic targets for designing new cancer treatments.

    Topics: Animals; Humans; Neoplasms; Proteasome Endopeptidase Complex; Signal Transduction; Transforming Growth Factor beta; Ubiquitin

2008
Role of transforming growth factor-beta superfamily signaling pathways in human disease.
    Biochimica et biophysica acta, 2008, Volume: 1782, Issue:4

    Transforming growth factor beta (TGF-beta) superfamily signaling pathways are ubiquitous and essential regulators of cellular processes including proliferation, differentiation, migration, and survival, as well as physiological processes, including embryonic development, angiogenesis, and wound healing. Alterations in these pathways, including either germ-line or somatic mutations or alterations in the expression of members of these signaling pathways often result in human disease. Appropriate regulation of these pathways is required at all levels, particularly at the ligand level, with either a deficiency or an excess of specific TGF-beta superfamily ligands resulting in human disease. TGF-beta superfamily ligands and members of these TGF-beta superfamily signaling pathways also have emerging roles as diagnostic, prognostic or predictive markers for human disease. Ongoing studies will enable targeting of TGF-beta superfamily signaling pathways for the chemoprevention and treatment of human disease.

    Topics: Biomarkers; Cardiovascular Diseases; Chromosome Aberrations; Disease; Female; Genetic Diseases, Inborn; Humans; Ligands; Musculoskeletal Diseases; Mutation; Neoplasms; Pregnancy; Pregnancy Complications; Prognosis; Signal Transduction; Transforming Growth Factor beta

2008
The multiple roles of CUX1: insights from mouse models and cell-based assays.
    Gene, 2008, Apr-15, Volume: 412, Issue:1-2

    Cux (Cut homeobox) genes are present in all metazoans. Early reports described many phenotypes caused by cut mutations in Drosophila melanogaster. In vertebrates, CUX1 was originally characterized as the CCAAT-displacement protein (CDP). Another line of investigation revealed the presence of CUX1 within a multi-protein complex called the histone nuclear factor D (HiNF-D). Recent studies led to the identification of several CUX1 isoforms with distinct DNA binding and transcriptional properties. While the CCAAT-displacement activity was implicated in the transcriptional repression of several genes, some CUX1 isoforms were found to participate in the transcriptional activation of some genes. The expression and activity of CUX1 was shown to be regulated through the cell cycle and to be a target of TGF-beta signaling. Mechanisms of regulation include alternative transcription initiation, proteolytic processing, phosphorylation and acetylation. Cell-based assays have established a role for CUX1 in the control of cell cycle progression, cell motility and invasion. In the mouse, gene inactivation as well as over-expression in transgenic mice has revealed phenotypes in multiple organs and cell types. While some phenotypes could be explained by the presumed functions of CUX1 in the affected cells, other phenotypes invoked non-cell-autonomous effects that suggest regulatory functions with an impact on cell-cell interactions. The implication of CUX1 in cancer was suggested first from its over-expression in primary tumors and cancer cell lines and was later confirmed in mouse models.

    Topics: Animals; Cell Differentiation; Cell Line; DNA-Binding Proteins; Genes, Homeobox; Homeodomain Proteins; Humans; Mice; Mice, Knockout; Mice, Transgenic; Models, Animal; Mutation; Neoplasms; Nuclear Proteins; Protein Processing, Post-Translational; Repressor Proteins; RNA, Messenger; Transforming Growth Factor beta

2008
Endoglin (CD105): a marker of tumor vasculature and potential target for therapy.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2008, Apr-01, Volume: 14, Issue:7

    Endoglin (CD105) is an accessory protein of the transforming growth factor-beta receptor system expressed on vascular endothelial cells. Mutation of the endoglin gene is associated with hereditary hemorrhagic telangiectasias, or Osler-Weber-Rendu syndrome, and has been studied extensively in the context of this disease. The expression of endoglin is elevated on the endothelial cells of healing wounds, developing embryos, inflammatory tissues, and solid tumors. Endoglin is a marker of activated endothelium, and its vascular expression is limited to proliferating cells. Recent studies identified endoglin expression in several solid tumor types, with the level of expression correlating with various clinicopathologic factors including decreased survival and presence of metastases. Attempts to target endoglin and the cells that express this protein in tumor-bearing mice have yielded promising results.

    Topics: Animals; Antigens, CD; Biomarkers, Tumor; Endoglin; Endothelial Cells; Humans; Neoplasms; Neovascularization, Pathologic; Receptors, Cell Surface; Transforming Growth Factor beta

2008
To (TGF)beta or not to (TGF)beta: fine-tuning of Smad signaling via post-translational modifications.
    Cellular signalling, 2008, Volume: 20, Issue:9

    Smad proteins are key signal transducers for the TGF-beta superfamily and are frequently inactivated in human cancers, yet the molecular basis of how their levels and activities are regulated remains unclear. Recent progress, discussed herein, illustrates the critical roles of Smad post-translational modifications in the cellular outcome to TGF-beta signaling.

    Topics: Amino Acid Motifs; Animals; Humans; Neoplasms; Protein Processing, Post-Translational; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2008
TGF-beta and tumors--an ill-fated alliance.
    Current opinion in immunology, 2008, Volume: 20, Issue:2

    Mechanisms of host defense can form an unwitting alliance with tumor cells to promote tumor progression, invasion, and dissemination to distant sites. By secreting TGF-beta, an immunoregulatory molecule designated for both promoting inflammation and dampening immune responses, the tumor tricks the host into supporting its expansion and survival. TGF-beta not only recruits leukocytes to secrete chemokines, growth factors, cytokines, and proteases in support of a tumor-friendly niche but also in a context-specific manner, incapacitates the emergent immune response. As a profound immunosuppressant, TGF-beta, both directly and through the generation of regulatory T cells, blunts immune surveillance, favoring tumor escape. Collectively, the ability of the tumor to hijack these host defense pathways can tip the balance in favor of the tumor.

    Topics: Animals; Carcinoma, Squamous Cell; Head and Neck Neoplasms; Humans; Mice; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Escape

2008
Actions of TGF-beta as tumor suppressor and pro-metastatic factor in human cancer.
    Biochimica et biophysica acta, 2007, Volume: 1775, Issue:1

    Transforming growth factor-beta (TGF-beta) is a secreted polypeptide that signals via receptor serine/threonine kinases and intracellular Smad effectors. TGF-beta inhibits proliferation and induces apoptosis in various cell types, and accumulation of loss-of-function mutations in the TGF-beta receptor or Smad genes classify the pathway as a tumor suppressor in humans. In addition, various oncogenic pathways directly inactivate the TGF-beta receptor-Smad pathway, thus favoring tumor growth. On the other hand, all human tumors overproduce TGF-beta whose autocrine and paracrine actions promote tumor cell invasiveness and metastasis. Accordingly, TGF-beta induces epithelial-mesenchymal transition, a differentiation switch that is required for transitory invasiveness of carcinoma cells. Tumor-derived TGF-beta acting on stromal fibroblasts remodels the tumor matrix and induces expression of mitogenic signals towards the carcinoma cells, and upon acting on endothelial cells and pericytes, TGF-beta regulates angiogenesis. Finally, TGF-beta suppresses proliferation and differentiation of lymphocytes including cytolytic T cells, natural killer cells and macrophages, thus preventing immune surveillance of the developing tumor. Current clinical approaches aim at establishing novel cancer drugs whose mechanisms target the TGF-beta pathway. In conclusion, TGF-beta signaling is intimately implicated in tumor development and contributes to all cardinal features of tumor cell biology.

    Topics: Animals; Antibodies, Neoplasm; Apoptosis; Carcinogens; Cell Cycle; Cell Transformation, Neoplastic; Disease Models, Animal; Epigenesis, Genetic; Epithelial Cells; Humans; Mesoderm; Mice; Mutation; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Oncogenes; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Suppressor Proteins

2007
The commonality of plasticity underlying multipotent tumor cells and embryonic stem cells.
    Journal of cellular biochemistry, 2007, Jul-01, Volume: 101, Issue:4

    Aggressive cancer cells and pluripotent stem cells converge in their capacity for self-renewal, proliferation and plasticity. Recent studies have capitalized on these similarities by demonstrating that tumors arise from specific cancer stem cell populations that, in a manner reminiscent of normal stem cells, are able to both self-renew and give rise to a heterogeneous tumor population. This stem cell like function of aggressive cancer cells is likely attributable to the ectopic expression of embryonic factors such as Nodal and Cancer Testis Specific Antigens (CTAs), which maintain a functional plasticity by promoting pluripotency and immortality. During development, the expression of these embryonic factors is tightly regulated by a dynamic array of mediators, including the spatial and temporal expression of inhibitors such as Lefty, and the epigenetic modulation of the genome. In aggressive cancer cells, particularly melanoma, this balance of regulatory mediators is disrupted, leading to the aberrant expression of pluripotency-associated genes. By exposing aggressive cancer cells to embryonic microenvironments, this balance of regulatory mediators is restored, thereby reprogramming tumor cells to a more benign phenotype. These stem cell-derived mediators, as well as the genes they regulate, provide therapeutic targets designed to specifically differentiate and eradicate aggressive cancers.

    Topics: Cell Lineage; Embryonic Stem Cells; Humans; Left-Right Determination Factors; Models, Biological; Neoplasms; Neoplastic Stem Cells; Nodal Protein; Pluripotent Stem Cells; Smad Proteins; Transforming Growth Factor beta

2007
The IL-6/sIL-6R complex as a novel target for therapeutic approaches.
    Expert opinion on therapeutic targets, 2007, Volume: 11, Issue:5

    IL-6 plays a pivotal role in immune responses and certain oncologic conditions. The intense investigation of its biological activity and function led to the discovery of two different IL-6-driven signalling pathways. Binding to the membrane-bound IL-6 receptor (mIL-6R, CD126) causes the recruitment of two gp130 co-receptor molecules (CD130) and the activation of intracellular signalling cascades via gp130. Although this classical pathway is mainly limited to hepatocytes, neutrophils, monocytes/macrophages and certain other leukocyte populations, which express IL-6R on their surface, an alternative mechanism has also been described. Proteolytic cleavage of the mIL-6R protein or translation from alternatively spliced mRNA leads to the generation of a soluble form of the IL-6R (sIL-6R), which is likewise able to bind to IL-6. The resulting IL-6/sIL-6R complex is also capable of binding to gp130 and inducing intracellular signalling. Through this so-called 'trans-signalling' mechanism, IL-6 is able to stimulate cells that lack an endogenous mIL-6R. High levels of IL-6 and sIL-6R have been reported in several chronic inflammatory and autoimmune diseases as well as in cancer. Preclinical animal disease models have provided strong evidence that specific blockade of IL-6-regulated signalling pathways represents a promising approach for the therapy of these diseases. An optimised variant of the recently described fusion protein sgp30Fc is now heading towards its clinical evaluation.

    Topics: Alternative Splicing; Animals; Anti-Inflammatory Agents; Antineoplastic Agents; Arthritis; Arthritis, Rheumatoid; Asthma; Autoimmune Diseases; Cell Line; Clinical Trials as Topic; Colitis; Colonic Neoplasms; Cytokine Receptor gp130; Disease Models, Animal; Drug Delivery Systems; Drug Evaluation, Preclinical; Humans; Inflammation; Interleukin-6; Leukocytes; Male; Mice; Neoplasms; Receptors, Interleukin-6; Recombinant Fusion Proteins; Signal Transduction; Solubility; Transforming Growth Factor beta

2007
A delicate balance: TGF-beta and the tumor microenvironment.
    Journal of cellular biochemistry, 2007, Jul-01, Volume: 101, Issue:4

    The activated form of TGF-beta is a known regulator of epithelial cell autonomous tumor initiation, progression, and metastasis. Recent studies have also indicated that TGF-beta mediates interactions between cancer cells and their local tumor microenvironment. Specifically, the loss of TGF-beta signaling in stromal components including fibroblasts and T-cells can result in an "activated" microenvironment that supports and even initiates transformation of adjacent epithelial cells. TGF-beta signaling in cancer can be regulated through mechanisms involving ligand activation and expression of essential components within the pathway including the receptors and downstream effectors. TGF-beta signaling in the tumor microenvironment significantly impacts carcinoma initiation, progression, and metastasis via epithelial cell autonomous and interdependent stromal-epithelial interactions in vivo.

    Topics: Animals; Cell Transformation, Neoplastic; Epithelial Cells; Humans; Mesoderm; Models, Biological; Neoplasms; Transforming Growth Factor beta

2007
Inhibition of the TGF-beta signaling pathway in tumor cells.
    Recent results in cancer research. Fortschritte der Krebsforschung. Progres dans les recherches sur le cancer, 2007, Volume: 172

    Topics: Antineoplastic Agents; Cell Transformation, Neoplastic; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2007
TGFBIp/betaig-h3 protein: a versatile matrix molecule induced by TGF-beta.
    The international journal of biochemistry & cell biology, 2007, Volume: 39, Issue:12

    TGFBIp/betaig-h3 protein is an extracellular matrix molecule initially cloned from human adenocarcinoma cells treated with TGF-beta. Its precise function remains obscure but a number of studies have demonstrated it to be an intriguingly versatile molecule role in a wide range of physiological and pathological conditions. To date, the most extensively studied and reported action of TGFBIp/betaig-h3 protein is in corneal dystrophy and several excellent reviews are available on this. Work from various laboratories on this molecule has compiled a tremendous amount of information over the past decade and a half. Here we review the current understanding on TGFBIp/betaig-h3 protein and its functions in morphogenesis, extracellular matrix interactions, adhesion/migration, corneal dystrophy, tumorigenesis, angiogenesis, nephropathies, osteogenesis, wound healing and inflammation.

    Topics: Animals; Corneal Dystrophies, Hereditary; Extracellular Matrix; Extracellular Matrix Proteins; Humans; Mutation; Neoplasms; Transforming Growth Factor beta; Wound Healing

2007
Tgf-Beta signaling in development.
    Science's STKE : signal transduction knowledge environment, 2007, Aug-14, Volume: 2007, Issue:399

    The transforming growth factor-beta (TGF-beta) superfamily comprises nearly 30 growth and differentiation factors that include TGF-betas, activins, inhibins, and bone morphogenetic proteins (BMPs). Multiple members of the TGF-beta superfamily serve key roles in stem cell fate commitment. The various members of the family can exhibit disparate roles in regulating the biology of embryonic stem (ES) cells and tumor suppression. For example, TGF-beta inhibits proliferation of multipotent hematopoietic progenitors, promotes lineage commitment of neural precursors, and suppresses epithelial tumors. BMPs block neural differentiation of mouse and human ES cells, contribute to self-renewal of mouse ES cells, and also suppress tumorigenesis. ES cells and tumors may be exposed to multiple TGF-beta members, and it is likely that the combination of growth factors and cross-talk among the intracellular signaling pathways is what precisely defines stem cell fate commitment. This Connections Map Pathway in the Database of Cell Signaling integrates signaling not only from TGF-beta and BMP but also from the ligands nodal and activin, and describes the role of the signaling pathways activated by these ligands in mammalian development. Much of the evidence for the connections shown comes from studies on mouse and human ES cells or mouse knockouts. This pathway is important for understanding not only stem cell biology, but also the molecular effectors of TGF-beta and BMP signaling that may contribute to cancer suppression or progression and thus are potential targets for therapeutic intervention.

    Topics: Animals; Cell Lineage; Humans; Mice; Mice, Knockout; Neoplasms; Signal Transduction; Stem Cells; Transforming Growth Factor beta

2007
Myeloid-derived suppressor cells.
    Advances in experimental medicine and biology, 2007, Volume: 601

    The development of tumor-specific T cell tolerance is largely responsible for tumor escape. Accumulation of myeloid-derived suppressor cells (MDSCs) in animal tumor models as well as in cancer patients is involved in tumor-associated T cell tolerance. In recent years, it has become increasingly evident that MDSCs bring about antigen-specific T cell tolerance by various mechanisms, which is the focus of this chapter.

    Topics: Animals; Antigens; Arginine; Cell Differentiation; Epitopes; Humans; Immune Tolerance; Models, Biological; Myeloid Cells; Neoplasms; Peroxynitrous Acid; Reactive Oxygen Species; T-Lymphocytes; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2007
Pathobiology of transforming growth factor beta in cancer, fibrosis and immunologic disease, and therapeutic considerations.
    Laboratory investigation; a journal of technical methods and pathology, 2007, Volume: 87, Issue:11

    Transforming growth factor beta (TGF-beta) is a highly pleiotropic cytokine that plays an important role in wound healing, angiogenesis, immunoregulation and cancer. The cells of the immune system produce the TGF-beta1 isoform, which exerts powerful anti-inflammatory functions, and is a master regulator of the immune response. However, this is context dependent, because TGF-beta can contribute to the differentiation of both regulatory (suppressive) T cells (Tr cells) and inflammatory Th17 cells. While TGF-beta might be underproduced in some autoimmune diseases, it is overproduced in many pathological conditions. This includes pulmonary fibrosis, glomerulosclerosis, renal interstitial fibrosis, cirrhosis, Crohn's disease, cardiomyopathy, scleroderma and chronic graft-vs-host disease. In neoplastic disease, TGF-beta suppresses the progression of early lesions, but later this effect is lost and cancer cells produce TGF-beta, which then promotes metastasis. This cytokine also contributes to the formation of the tumor stroma, angiogenesis and immunosuppression. In view of this, several approaches are being studied to inhibit TGF-beta activity, including neutralizing antibodies, soluble receptors, receptor kinase antagonist drugs, antisense reagents and a number of less specific drugs such as angiotensin II antagonists and tranilast. It might be assumed that TGF-beta blockade would result in severe inflammatory disease, but this has not been the case, presumably because the neutralization is only partial. In contrast, the systemic administration of TGF-beta for therapeutic purposes is limited by toxicity and safety concerns, but local administration appears feasible, especially to promote wound healing. Immunotherapy or vaccination stimulating TGF-beta production and/or Tr differentiation might be applied to the treatment of autoimmune diseases. The benefits of new therapies targeting TGF-beta are under intense investigation.

    Topics: Animals; Autoimmune Diseases; Cancer Vaccines; Fibrosis; Genetic Therapy; Humans; Immune System Diseases; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Stromal Cells; Transforming Growth Factor beta; Transforming Growth Factor beta1; Wound Healing

2007
Role of TIEG1 in biological processes and disease states.
    Journal of cellular biochemistry, 2007, Oct-15, Volume: 102, Issue:3

    A novel TGFbeta Inducible Early Gene-1 (TIEG1) was discovered in human osteoblast (OB) cells by our laboratory. Over the past decade, a handful of laboratories have revealed a multitude of organismic, cellular, and molecular functions of this gene. TIEG1 is now classified as a member of the 3 zinc finger family of Krüppel-like transcription factors (KLF10). Other closely related factors [TIEG2 (KLF11) and TIEG3/TIEG2b] have been reported and are briefly compared. As described in this review, TIEG1 is shown to play a role in regulating estrogen and TGFbeta actions, the latter through the Smad signaling pathway. In both cases, TIEG1 acts as an inducer or repressor of gene transcription to enhance the TGFbeta/Smad pathway, as well at other signaling pathways, to regulate cell proliferation, differentiation, and apoptosis. This review outlines TIEG1's molecular functions and roles in skeletal disease (osteopenia/osteoporosis), heart disease (hypertrophic cardiomyopathy), and cancer (breast and prostate).

    Topics: Animals; Apoptosis; Bone and Bones; Disease Models, Animal; Early Growth Response Transcription Factors; Estrogens; Gene Expression Regulation; Humans; Kruppel-Like Transcription Factors; Mice; Models, Biological; Myocardium; Neoplasms; Osteoblasts; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2007
Transforming growth factor-beta signaling in cancer invasion and metastasis.
    International journal of cancer, 2007, Nov-15, Volume: 121, Issue:10

    Transforming growth factor-beta (TGF-beta) family members are polypeptides with dual tumor suppressive and oncogenic effects. They signal through serine/threonine kinase receptor complexes, which phosphorylate cytoplasmic mediators, the Smads. Upon phosphorylation, Smads translocate to the nucleus and associate with transcriptional coactivators or corepressors, and regulate the transcriptional activation of various TGF-beta responsive genes. In addition, TGF-beta activates cellular mitogen-activated protein kinase signaling pathways, which crosstalk with Smad signaling and regulate growth, survival and motility of cells. During tumorigenesis, malignantly transformed cells often lose the response to the tumor suppressive effects of TGF-beta, which, in turn, starts to act as an autocrine tumor promoting factor by enhancing cancer invasion and metastasis. In this review, we summarize current view on the role of TGF-beta signaling in tumorigenesis, with emphasis on cancer invasion and metastasis. On the basis of these recent observations, we discuss new therapeutic strategies targeting TGF-beta signaling at distinct levels as a basis for inhibiting tumor growth, angiogenesis, invasion and metastasis.

    Topics: Animals; Cell Transformation, Neoplastic; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2007
Signaling pathways in cancer and embryonic stem cells.
    Stem cell reviews, 2007, Volume: 3, Issue:1

    Cancer cells have the ability to divide indefinitely and spread to different parts of the body during metastasis. Embryonic stem cells can self-renew and, through differentiation to somatic cells, provide the building blocks of the human body. Embryonic stem cells offer tremendous opportunities for regenerative medicine and serve as an excellent model system to study early human development. Many of the molecular mechanism underlying tumorigenesis in cancer and self-renewal in stem cells have been elucidated in the past decade. Here we present a systematic analysis of seven major signaling pathways implicated in both cancer and stem cells. We present on overview of the JAK/STAT, Notch, MAPK/ERK, PI3K/AKT, NF-kB, Wnt and TGF-beta pathways and analyze their activation status in the context of cancer and stem cells. We focus on their role in stem cell self-renewal and development and identify key molecules, whose aberrant expression has been associated with malignant phenotypes. We conclude by presenting a map of the signaling networks involved in cancer and embryonic stem cells.

    Topics: Animals; Embryonic Stem Cells; Humans; Janus Kinases; MAP Kinase Signaling System; Models, Biological; Neoplasms; NF-kappa B; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Receptors, Notch; Signal Transduction; STAT Transcription Factors; Transforming Growth Factor beta; Wnt Proteins

2007
The Wnt signal transduction pathway in stem cells and cancer cells: influence on cellular invasion.
    Stem cell reviews, 2007, Volume: 3, Issue:1

    The regulative network conducting adult stem cells in endogenous tissue repair is of prime interest for understanding organ regeneration as well as preventing degenerative and malignant diseases. One major signal transduction pathway which is involved in the control of these (patho)physiological processes is the Wnt pathway. Recent results obtained in our laboratories showed for the first time that canonical Wnt signaling is critically involved in the control of the migration/invasion behaviour of human mesenchymal stem cells (hMSC). In the first part of this review, we describe that the regenerative state is closely linked to the activation of the Wnt pathway. Central hallmarks of activated stem cells are recapitulated in a similar way also in cancer metastasis, where the acquisition of an invasive cancer stem cell phenotype is associated with the induction of Wnt-mediated epithelial to mesenchymal transition (EMT). In the second part, the influence of proinflammatory cytokines such as transforming growth factor (TGF-)beta1, interleukin (Il-)1beta, and tumor necrosis factor (TNF-)alpha is discussed with regard to the invasive characteristics of hMSC. In this context, special attention has been paid on the role of matrix metalloproteinases (MMPs), such as MMP-2, MMP-9 and membrane type 1 (MT1)-MMP, as well as on the tissue inhibitors of metalloproteinases TIMP-1 and TIMP-2. Putative cross-talks between different signal transduction pathways that may amplify the invasive capacity of this stem cell population are also discussed. Finally, the consequences towards future drug-mediated therapeutical modifications of Wnt signaling in stem cells and tumor cells are highlighted.

    Topics: Animals; Bone Marrow Cells; Cell Proliferation; Humans; Matrix Metalloproteinases; Models, Biological; Neoplasm Invasiveness; Neoplasms; Peptide Hydrolases; Regeneration; Signal Transduction; Stem Cells; Tissue Inhibitor of Metalloproteinases; Transforming Growth Factor beta; Wnt Proteins

2007
Transforming growth factor-beta and the immune response: implications for anticancer therapy.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2007, Sep-15, Volume: 13, Issue:18 Pt 1

    Immune homeostasis is a delicate balance between the immune defense against foreign pathogens and suppression of the immune system to maintain self-tolerance and prevent autoimmune disease. Maintenance of this balance involves several crucial networks of cytokines and various cell types. Among these regulators, transforming growth factor-beta (TGF-beta) is a potent cytokine with diverse effects on hematopoietic cells. Its pivotal function within the immune system is to maintain tolerance via the regulation of lymphocyte proliferation, differentiation, and survival. In addition, TGF-beta controls the initiation and resolution of inflammatory responses through the regulation of chemotaxis and activation of leukocytes in the periphery, including lymphocytes, natural killer cells, dendritic cells, macrophages, mast cells, and granulocytes. Through its pleiotropic effects on these immune cells, TGF-beta prevents the development of autoimmune diseases without compromising immune responses to pathogens. However, overactivation of this pathway can lead to several immunopathologies under physiologic conditions including cancer progression, making it an attractive target for antitumor therapies. This review discusses the biological functions of TGF-beta and its effects on the immune system and addresses how immunosuppression by this cytokine can promote tumorigenesis, providing the rationale for evaluating the immune-enhancing and antitumor effects of inhibiting TGF-beta in cancer patients.

    Topics: Animals; Dendritic Cells; Humans; Killer Cells, Natural; Neoplasms; T-Lymphocytes; Transforming Growth Factor beta

2007
Targeting aberrant TGF-beta signaling in pre-clinical models of cancer.
    Anti-cancer agents in medicinal chemistry, 2007, Volume: 7, Issue:5

    The TGF-beta signaling pathway is central to the control of diverse biological processes including cellular proliferation, cell survival, apoptosis, extracellular matrix deposition/remodeling, migration, invasion and immune regulation/inflammation. Given the pleiotropic effects of this cytokine, it comes as no surprise that numerous pathological conditions are associated with alterations in the TGF-beta pathway, including chronic fibrosis, airway remodeling (asthma), cardiovascular disease and cancer. Thus, there are increasing efforts to develop reagents and therapeutic strategies to impair TGF-beta signaling. Here we review several classes of inhibitors, including knock-down strategies aimed at signaling components of the TGF-beta pathway, TGF-beta neutralizing antibodies, TGF-beta receptor extracellular domains that function as ligand traps and small molecule kinase inhibitors. Strategies with potential for application as anti-cancer therapeutics that have been evaluated in pre-clinical animal models will be discussed. TGF-beta action is complex, shifting from a tumor suppressor to a promoter of tumor cell invasion and metastasis in several types of cancer. This raises important issues regarding not only the status of the TGF-beta pathway in the individual patient but also the precise stage during disease progression that such inhibitors should be employed. Potential consequences of targeting the TGF-beta pathway will also be considered.

    Topics: Animals; Antineoplastic Agents; Drug Evaluation, Preclinical; Humans; Neoplasms; Transforming Growth Factor beta

2007
Targeting transforming growth factor-beta signaling.
    Current opinion in oncology, 2007, Volume: 19, Issue:6

    Most cancers are characterized by excessive transforming growth factor-beta production by tumors, which can promote tumor growth and mediate epithelial-to-mesenchymal transition. Transforming growth factor-beta also has the ability to overproduce extracellular matrix components in response to injury and other stimuli. There are many strategies undergoing current evaluation for inhibiting the deleterious biological effects of transforming growth factor-beta by disrupting its signaling at various levels. The current review focuses on the recent advances made in this area, and the potential of these strategies in the clinical treatment of cancer and fibrosis.. Four main strategies used most recently for disrupting transforming growth factor-beta signaling are brought into focus in this review: inhibition or sequestration of the transforming growth factor-beta protein ligands, inhibition of transforming growth factor-beta receptor kinase activity, inhibition of SMAD signaling downstream of transforming growth factor-beta kinase activity and restoration of antitumor immunity upon transforming growth factor-beta inhibition. Various techniques currently used to employ these four strategies are discussed.. Several lines of evidence suggest that altered transforming growth factor-beta signaling contributes to tumor progression and metastasis as well as development of fibrosis. Accumulating data from preclinical and clinical studies indicate that antagonizing aberrant transforming growth factor-beta signaling is a promising novel therapeutic approach in cancer and fibrotic disorders.

    Topics: Antineoplastic Agents; Clinical Trials as Topic; Disease Progression; Extracellular Matrix; Fibrosis; Gene Expression Regulation, Neoplastic; Humans; Ligands; Models, Biological; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2007
Mechanisms of action of TGF-beta in cancer: evidence for Smad3 as a repressor of the hTERT gene.
    Annals of the New York Academy of Sciences, 2007, Volume: 1114

    Transforming growth factor-beta (TGF-beta) induces cell differentiation and suppresses cell proliferation, but the mechanisms underlying the actions of TGF-beta remain to be fully elucidated. Recent studies suggest that TGF-beta suppresses neoplastic cell development by employing Smad3 protein to repress the gene of human telomerase reverse transcriptase (hTERT). In human breast cancer cells, TGF-beta induces rapid phosphorylation and subsequent entry of Smad3 into the nucleus. In the nucleus, Smad3 binds to the hTERT gene promoter directly and inhibits hTERT gene transcription activity. By interacting with c-myc, Smad3 also represses the c-myc gene. Thus, TGF-beta prevents continuous cell proliferation by switching off telomerase activity through Smad3 repression of the hTERT gene and the action of c-myc. Modulating the interface between Smad3 and the hTERT gene, and the potential feedback loop from telomeres to Smad3 via Smurf2, may represent a novel approach to regulate cell lifespan of proliferation.

    Topics: Animals; Gene Expression Regulation, Enzymologic; Humans; Neoplasms; Smad3 Protein; Telomerase; Transforming Growth Factor beta; Tumor Suppressor Proteins

2007
Transforming growth factor-beta and the immune response to malignant disease.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2007, Nov-01, Volume: 13, Issue:21

    Transforming growth factor-beta (TGF-beta) is a key player in malignant disease through its actions on host tissues and cells. Malignant cells often secrete large amounts of TGF-beta that act on nontransformed cells present in the tumor mass as well as distal cells in the host to suppress antitumor immune responses creating an environment of immune tolerance, augmenting angiogenesis, invasion and metastasis, and increasing tumor extracellular matrix deposition. Cells of the innate immune system contribute to the high concentrations of TGF-beta found in tumor masses. In addition, dendritic cell subpopulations secreting TGF-beta contribute to the generation of regulatory T cells that actively inhibit the activity of other T cells. Elevated levels of plasma TGF-beta are associated with advanced stage disease and may separate patients into prognostically high-risk populations. Anti-TGF-beta therapy could reverse the immunosuppressive effects of this cytokine on the host as well as decrease extracellular matrix formation, decrease angiogenesis, decrease osteolytic activity, and increase the sensitivity of the malignant cells to cytotoxic therapies and immunotherapies. Phase I clinical trials of an inhibitor of TGF-beta receptor type I kinase activity and a TGF-beta neutralizing antibody are under way.

    Topics: Animals; Humans; Immune System; Immune Tolerance; Immunosuppressive Agents; Models, Biological; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Prognosis; T-Lymphocytes; Transforming Growth Factor beta

2007
SEL1L a multifaceted protein playing a role in tumor progression.
    Journal of cellular physiology, 2006, Volume: 208, Issue:1

    Since the cloning in 1997 of SEL1L, the human ortholog of the sel-1 gene of C. elegans, most studies have focused on its role in cancer progression and have provided significant evidences to link its increased expression to a decrease in tumor aggressiveness. SEL1L resides on a "Genome Desert area" on chromosome 14q24.3-31 and is highly conserved in evolution. The function of the SEL1L encoded protein is still very elusive although, several evidences from lower organisms indicate that it plays a major role in protein degradation using the ubiquitin-proteosome system. SEL1L has a very complex structure made up of modules: genomically it consists of 21 exons featuring several alternative transcripts encoding for putative protein isoforms. This structural complexity ensures protein flexibility and specificity, indeed the protein was found in different sub-cellular compartments and may turn on a particular transcript in response to specific stimuli. The overall architecture of SEL1L guarantees an exquisite regulation in the expression of the gene.

    Topics: Amino Acid Sequence; Animals; Cell Proliferation; Cell Transformation, Neoplastic; Chromosome Deletion; Chromosomes, Human, Pair 14; Disease Progression; DNA Mutational Analysis; DNA, Neoplasm; Exons; Fetus; Gene Expression Regulation, Neoplastic; Humans; Molecular Sequence Data; Neoplasm Metastasis; Neoplasms; Polymorphism, Genetic; Protein Isoforms; Proteins; Receptors, Notch; Signal Transduction; Transforming Growth Factor beta

2006
Inhibition of TGFbeta signaling in cancer therapy.
    Current opinion in genetics & development, 2006, Volume: 16, Issue:1

    Recent evidence continues to support a central role for TGFbeta in tumor maintenance and progression. Although this may involve TGFbeta-mediated paracrine effects that modulate the tumor microenvironment and the host immune system, some studies causally implicate autocrine TGFbeta in cancer cell motility and survival. Other recent evidence indicates synergy between oncogene and TGFbeta signaling in epithelial cell transformation. This suggests opportunities for dissecting molecular mechanisms of cross-talk as well as providing insights into possible combinatorial molecular anticancer therapies that will include TGFbeta inhibitors.

    Topics: Animals; Biomarkers, Tumor; Humans; Mice; Models, Biological; Neoplasms; Oncogenes; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2006
Mechanisms of pericyte recruitment in tumour angiogenesis: a new role for metalloproteinases.
    European journal of cancer (Oxford, England : 1990), 2006, Volume: 42, Issue:3

    Pericytes occur in tumour blood vessels and are critical for the development of a functional vascular network. Targeting tumour pericytes is a promising anti-angiogenic therapy but requires identifying the mechanisms of their recruitment in tumour and addressing whether these mechanisms can be selectively harnessed. Among the pathways involved in pericyte recruitment during embryonic development, the contribution of platelet-derived growth factor B and sphingosine 1-phosphate is confirmed in tumour angiogenesis. The effect of angiopoietin 1 depends on the tumour model. Transforming growth factor-beta1 enhances tumour vascularization and microvessel maturation. Recent reports suggest a participation of matrix metalloproteinases (MMP) in tumour pericyte recruitment that is consistent with the effect of certain MMPs in the development of microvasculature in embryonic development and in in vitro models of vascular remodelling. Here, we discuss the possibility for MMPs to contribute to pericyte recruitment at six levels: (1) direct promotion of pericyte invasion by extracellular matrix degradation; (2) stimulation of pericyte proliferation and protection against apoptosis by modification of the ECM; (3) activation of pericytes through the release of growth factor bound to the ECM; (4) transactivation of angiogenic cell surface receptor; (5) propagation of angiogenic signalling as cofactor; and (6) recruitment of bone marrow-derived stem cells.

    Topics: Angiopoietin-1; Cell Movement; Cell Proliferation; Humans; Lysophospholipids; Matrix Metalloproteinases; Neoplasms; Neovascularization, Pathologic; Pericytes; Proto-Oncogene Proteins c-sis; Sphingosine; Transforming Growth Factor beta; Transforming Growth Factor beta1

2006
Tissue fibrosis and carcinogenesis: divergent or successive pathways dictate multiple molecular therapeutic targets for oligo decoy therapies.
    Journal of cellular biochemistry, 2006, Apr-15, Volume: 97, Issue:6

    The extracellular matrix (ECM) is composed of several families of macromolecular components: fibrous proteins such as collagens, type I collagen (COL1), type III collagen (COL3), fibronectin, elastin, and glycoconjugates such as proteoglycans and matrix glycoproteins. Their receptors on the cell membrane, most of which in the case of the ECM belong to the integrins, which are heterodimeric proteins composed of alpha and beta chains. COL1 is the major fibrous collagen of bone, tendon, and skin; while COL3 is the more pliable collagen of organs like liver. Focus will not only be given to the regulation of synthesis of several fibrogenic parameters but also modulation of their degradation during growth factor-induced tissue fibrosis and cancer development. Evidence will be provided that certain tissues, which undergo fibrosis, also become cancerous. Why does there exist a divergency between tissues, which undergo frank fibrosis as an endpoint, and those tissues that undergo fibrosis and subsequently are susceptible to carcinogenicity; resulting from the etiological factor(s) causing the initial injury? For example, why does a polyvinyl alcohol (PVA) sponge implant become encapsulated and filled with fibrous tissue then fibrosis tissue growth stops? Why does the subcutaneous injection of a fibrogenic growth factor cause a benign growth and incisional wounding results in fibrosis and ultimately scarring? There are many examples of tissues, which undergo fibrosis as a prerequisite to carcinogenesis. Is there a cause-effect relationship? If you block tissue fibrosis in these precancerous tissues, would you block cancer formation? What are the molecular targets for blocking fibrosis and ultimately carcinogenesis? How can oligo decoys may be used to attenuate carcinogenesis and which oligo decoys specifically attenuate fibrogenesis as a prelude to carcinogenesis? What are other molecular targets for oligo decoy therapy in carcinogenesis?

    Topics: Animals; Cell Transformation, Neoplastic; Connective Tissue; Extracellular Matrix; Fibrosis; Gene Expression Regulation; Humans; Inflammation; Matrix Metalloproteinases; Models, Biological; Neoplasms; Oligonucleotides; Signal Transduction; Smad Proteins; Tissue Inhibitor of Metalloproteinases; Transcription Factors; Transforming Growth Factor beta

2006
Effective cancer therapy through immunomodulation.
    Annual review of medicine, 2006, Volume: 57

    MAbs directed toward tumor cells, tumor neovasculature, and host negative immunoregulatory elements (checkpoints) have emerged as useful immunotherapeutic agents against cancer. However, effective active modulation of the immune response with anticancer vaccines will require identifying appropriate tumor-rejection antigens; optimizing the interactions of peptides, antigen-presenting cells, and T cells; and blockading negative immunological checkpoints that impede an effective immune response. Checkpoints being targeted include CTLA-4 and PD1 that are negative signaling receptors expressed on activated T cells, CD4+CD25+ Foxp3-expressing Tregs (suppressor T cells), IL-2-mediated activation-induced cell death (AICD), and the cytokine TGFbeta.

    Topics: Antibodies, Monoclonal; Antigens, CD; Antigens, Differentiation; Antigens, Surface; Apoptosis Regulatory Proteins; Cancer Vaccines; CTLA-4 Antigen; Cytokines; Humans; Immunologic Factors; Neoplasms; Programmed Cell Death 1 Receptor; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2006
TGF-beta and cancer: is Smad3 a repressor of hTERT gene?
    Cell research, 2006, Volume: 16, Issue:2

    Transforming growth factor beta (TGF-beta) carries out tumor suppressor activity in epithelial and lymphoid cells, whereas telomerase is required for most cancers. Although the molecular mechanisms by which TGF-beta acts as a tumor suppressor are yet to be fully established, a link between TGFb and its tumor suppressor activity by telomerase has been suggested. Recently, we have noted a novel mode of action for TGF-beta through which human telomerase reverse transcriptase (hTERT) gene is repressed in immortal and neoplastic cells, confirming that one of the mechanisms underlying TGF-beta suppression of tumor growth may be through inhibiting hTERT gene transcription. Moreover, the inhibition of hTERT gene by TGF-beta suggests a cis action of the TGF-beta signaling molecule Smad3 on hTERT promoter directly. This article examines our current understanding and investigation of TGF-beta regulation of telomerase activity, and presents a model in which Smad3 participates in regulating hTERT gene transcription by acting as a repressor directly. Engineering the interface between Smad3 and hTERT gene may lead to a new strategy to inhibit telomerase activity in cancer.

    Topics: Gene Expression Regulation, Enzymologic; Humans; Neoplasms; Promoter Regions, Genetic; Repressor Proteins; Signal Transduction; Smad3 Protein; Telomerase; Transforming Growth Factor beta

2006
The role of the stroma in carcinogenesis.
    Bosnian journal of basic medical sciences, 2006, Volume: 6, Issue:1

    This systematic review considers the most recent attitudes and news regarding the influence of the stroma on tumor initiation and progression. It is now widely accepted that tumor stroma plays an active role in carcinogenesis. Many different signaling molecules, ligands and signaling pathways recently have been discovered. This review considers the complexity of interactions between malignant cells and its stroma (cross-talk). The recent advances and better understanding of the tumor-stroma interactions will have important impact on the new and combined therapeutic approaches and modalities.

    Topics: Cell Communication; Cellular Senescence; Female; Fibroblasts; Humans; Male; Models, Biological; Neoplasms; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2006
Transforming growth factor-beta regulation of immune responses.
    Annual review of immunology, 2006, Volume: 24

    Transforming growth factor-beta (TGF-beta) is a potent regulatory cytokine with diverse effects on hemopoietic cells. The pivotal function of TGF-beta in the immune system is to maintain tolerance via the regulation of lymphocyte proliferation, differentiation, and survival. In addition, TGF-beta controls the initiation and resolution of inflammatory responses through the regulation of chemotaxis, activation, and survival of lymphocytes, natural killer cells, dendritic cells, macrophages, mast cells, and granulocytes. The regulatory activity of TGF-beta is modulated by the cell differentiation state and by the presence of inflammatory cytokines and costimulatory molecules. Collectively, TGF-beta inhibits the development of immunopathology to self or nonharmful antigens without compromising immune responses to pathogens. This review highlights the findings that have advanced our understanding of TGF-beta in the immune system and in disease.

    Topics: Animals; Atherosclerosis; Autoimmune Diseases; B-Lymphocytes; Cell Differentiation; Cell Proliferation; Cell Survival; Dendritic Cells; Granulocytes; Humans; Immunoglobulin A; Infections; Killer Cells, Natural; Macrophages; Mast Cells; Models, Immunological; Neoplasms; Signal Transduction; T-Lymphocyte Subsets; Transforming Growth Factor beta

2006
TGF-beta in cancer and other diseases.
    Future oncology (London, England), 2006, Volume: 2, Issue:2

    Topics: Animals; Humans; Neoplasms; Transforming Growth Factor beta

2006
Cachexia: pathophysiology and clinical relevance.
    The American journal of clinical nutrition, 2006, Volume: 83, Issue:4

    Cachexia causes weight loss and increased mortality. It affects more than 5 million persons in the United States. Other causes of weight loss include anorexia, sarcopenia, and dehydration. The pathophysiology of cachexia is reviewed in this article. The major cause appears to be cytokine excess. Other potential mediators include testosterone and insulin-like growth factor I deficiency, excess myostatin, and excess glucocorticoids. Numerous diseases can result in cachexia, each by a slightly different mechanism. Both nutritional support and orexigenic agents play a role in the management of cachexia.

    Topics: Aging; Anorexia; Arthritis, Rheumatoid; Cachexia; Chronic Disease; Cytokines; Glucocorticoids; HIV Wasting Syndrome; Humans; Insulin-Like Growth Factor I; Kidney Failure, Chronic; Myostatin; Neoplasms; Pulmonary Disease, Chronic Obstructive; Testosterone; Transforming Growth Factor beta; Weight Loss

2006
Targeting the stromal microenvironment in chronic inflammation.
    Current opinion in pharmacology, 2006, Volume: 6, Issue:4

    A characteristic feature of chronic inflammatory reactions is their persistence and predilection for certain sites. The molecular basis for such tissue tropism (as, for example, seen with metastatic spread) has until recently remained obscure, but recent studies have strongly implicated tissue-resident, stromal cells, such as macrophages, endothelial cells and fibroblasts. These cell types make attractive therapeutic targets as they help define the three-dimensional structure of tissues and are key orchestrators of the inflammatory infiltrate. Most current anti-inflammatory therapies target immune cells in an attempt to inhibit the production of pro-inflammatory mediators; however, an equally important target is the active induction of anti-inflammatory mediators involved in the resolution of inflammation. Recent work suggests that stromal cells are an important source of these mediators. Targeting of multiple signals may be required to inhibit tissue damage associated with inflammatory disease. Cells of the monocyte lineage are present as tissue-resident cells and interact closely with other stromal populations. These cells form an ideal target for modulation of the inflammatory environment as, in some cases, they appear to induce tissue repair. Therapeutic manipulation of the stromal microenvironment has been particularly effective in treating cancer and is likely to provide a novel method to achieve improved control of chronic inflammatory disease.

    Topics: Angiogenic Proteins; Animals; Anti-Inflammatory Agents; Antibodies, Monoclonal; Chronic Disease; Cytokines; Endothelial Cells; Fibroblasts; Humans; Inflammation; Monocytes; Neoplasms; Stromal Cells; Transforming Growth Factor beta

2006
Escaping from the TGFbeta anti-proliferative control.
    Carcinogenesis, 2006, Volume: 27, Issue:11

    Transforming growth factor-beta (TGFbeta) has a crucial role in tissue homeostasis and disruption of the TGFbeta pathway has been implicated in many human diseases including cancer. As a potent inhibitor of epithelial cell proliferation, TGFbeta is a tumor suppressor. Tumor cells evade the antitumoral effect of TGFbeta, either by acquiring somatic mutations that blunt TGFbeta signaling or by selectively preventing the cytostatic responses to TGFbeta. During tumor progression, TGFbeta not only loses the anti-proliferative response but can also become an oncogenic factor. Recent work has provided insights into the specific molecular mechanisms involved in the loss of the TGFbeta anti-proliferative response. This review is an overview of the mechanisms that lead to the impairment of the tumor-suppressive function of TGFbeta in cancer. The understanding of how the TGFbeta signal is disrupted in cancer might facilitate the design and development of rational and successful therapeutic strategies.

    Topics: Animals; Antineoplastic Agents; Cell Differentiation; Cell Proliferation; Disease Progression; Epithelial Cells; Humans; Models, Biological; Mutation; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2006
Large- and small-molecule inhibitors of transforming growth factor-beta signaling.
    Current opinion in investigational drugs (London, England : 2000), 2006, Volume: 7, Issue:6

    A variety of drugs have been developed to inhibit transforming growth factor (TGF)beta signaling. These drugs have been designed to block TGFbeta synthesis, ligand/receptor binding or receptor kinase signaling. Preclinical studies using TGFbeta inhibitors have demonstrated efficacy in reducing metastasis and have shown improvements in cytotoxic drug delivery. Results of phase I/II clinical trials of TGFbeta inhibitors in patients with glioblastoma suggest improved survival rates compared with conventional chemotherapy. The predominant cellular target, whether cancer or stromal cell, immune cell or angiogenesis, may differ between tumor types. Different individuals may show variable responses to drug therapy dependent on both germline genetic variation and the somatic mutation profile of the tumor. A deeper understanding of these issues will assist in targeting the right patients for such therapy, and in limiting unwanted side effects.

    Topics: Animals; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Clinical Trials as Topic; Humans; Neoplasms; Oligonucleotides, Antisense; Patient Selection; Pteridines; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2006
Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer.
    Nature reviews. Cancer, 2006, Volume: 6, Issue:7

    Transforming growth factor-beta (TGFbeta) signalling regulates cancer through mechanisms that function either within the tumour cell itself or through host-tumour cell interactions. Studies of tumour-cell-autonomous TGFbeta effects show clearly that TGFbeta signalling has a mechanistic role in tumour suppression and tumour promotion. In addition, factors in the tumour microenvironment, such as fibroblasts, immune cells and the extracellular matrix, influence the ability of TGFbeta to promote or suppress carcinoma progression and metastasis. The complex nature of TGFbeta signalling and crosstalk in the tumour microenvironment presents a unique challenge, and an opportunity to develop therapeutic intervention strategies for targeting cancer.

    Topics: Animals; Cell Communication; Cell Transformation, Neoplastic; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2006
Inhibition of transforming growth factor-beta signaling in human cancer: targeting a tumor suppressor network as a therapeutic strategy.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2006, Jul-15, Volume: 12, Issue:14 Pt 1

    Topics: Antineoplastic Agents; Apoptosis; Genes, Tumor Suppressor; Humans; Ligands; Models, Biological; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2006
[TGF-beta signaling].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 2006, Volume: 51, Issue:10 Suppl

    Topics: Animals; Carrier Proteins; Genes, Tumor Suppressor; Humans; Intracellular Signaling Peptides and Proteins; Neoplasms; Proteasome Endopeptidase Complex; Protein Binding; Proto-Oncogene Proteins; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Ubiquitin; Ubiquitin-Protein Ligases

2006
Ubiquitin-dependent regulation of TGFbeta signaling in cancer.
    Neoplasia (New York, N.Y.), 2006, Volume: 8, Issue:8

    The transforming growth factorbeta (TGFbeta) superfamily regulates a broad spectrum of biological responses throughout embryonic development and adult life, including cell proliferation and differentiation, epithelial-to-mesenchymal transition, apoptosis, and angiogenesis. TGFbeta members initiate signaling by bringing together a complex of serine/threonine kinase receptors that transmit signals through intracellular Smad proteins. Genetic alterations in numerous components of the TGFbeta signaling pathway have been associated with several human cancers. In addition, tight regulation of TGFbeta signaling is pivotal to the maintenance of homeostasis and the prevention of carcinogenesis. The ubiquitin/proteosome system is one mechanism by which cells regulate the expression and activity of effectors of the TGFbeta signaling cascade. Mounting evidence also suggests that disruption of the ubiquitin-dependent degradation of components of the TGFbeta pathway leads to the development and progression of cancer. Therefore, understanding how these two pathways intertwine will contribute to the advancement of our knowledge of cancer development.

    Topics: Apoptosis; Cell Transformation, Neoplastic; Humans; Neoplasms; Neovascularization, Pathologic; Smad Proteins; Transforming Growth Factor beta; Ubiquitin

2006
Transforming growth factor-beta in cancer and metastasis.
    Cancer metastasis reviews, 2006, Volume: 25, Issue:3

    Transforming growth factor-beta (TGF-beta) is a multifunctional regulatory polypeptide that is the prototypical member of a large family of cytokines that controls many aspects of cellular function, including cellular proliferation, differentiation, migration, apoptosis, adhesion, angiogenesis, immune surveillance, and survival. The actions of TGF-beta are dependent on several factors including cell type, growth conditions, and the presence of other polypeptide growth factors. One of the biological effects of TGF-beta is the inhibition of proliferation of most normal epithelial cells using an autocrine mechanism of action, and this suggests a tumor suppressor role for TGF-beta. Loss of autocrine TGF-beta activity and/or responsiveness to exogenous TGF-beta appears to provide some epithelial cells with a growth advantage leading to malignant progression. This suggests a pro-oncogenic role for TGF-beta in addition to its tumor suppressor role. During the early phase of epithelial tumorigenesis, TGF-beta inhibits primary tumor development and growth by inducing cell cycle arrest and apoptosis. In late stages of tumor progression when tumor cells become resistant to growth inhibition by TGF-beta due to inactivation of the TGF-beta signaling pathway or aberrant regulation of the cell cycle, the role of TGF-beta becomes one of tumor promotion. Resistance to TGF-beta-mediated inhibition of proliferation is frequently observed in multiple human cancers, as are various alterations in the complex TGF-beta signaling and cell cycle pathways. TGF-beta can exert effects on tumor and stromal cells as well as alter the responsiveness of tumor cells to TGF-beta to stimulate invasion, angiogenesis, and metastasis, and to inhibit immune surveillance. Because of the dual role of TGF-beta as a tumor suppressor and pro-oncogenic factor, members of the TGF-beta signaling pathway are being considered as predictive biomarkers for progressive tumorigenesis, as well as molecular targets for prevention and treatment of cancer and metastasis.

    Topics: Antineoplastic Agents; Disease Progression; Humans; Neoplasm Metastasis; Neoplasms; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Tumor Suppressor Proteins

2006
TGF-beta: a mobile purveyor of immune privilege.
    Immunological reviews, 2006, Volume: 213

    Functionally barricaded immune responses or sites of immune privilege are no longer considered dependent on specific anatomical considerations, but rather, they can develop in any location where immunoregulatory cells congregate and express or release products capable of deviating the host response to foreign antigens. Among the pivotal molecules involved in orchestrating these ectopic sites of immune suppression is transforming growth factor-beta (TGF-beta), a secreted and cell-associated polypeptide with a multiplicity of actions in innate and adaptive immunity. While beneficial in initiating and controlling immune responses and maintaining immune homeostasis, immunosuppressive pathways mediated by TGF-beta may obscure immune surveillance mechanisms, resulting in failure to recognize or respond adequately to self, foreign, or tumor-associated antigens. CD4+CD25+Foxp3+ regulatory T cells represent a dominant purveyor of TGF-beta-mediated suppression and are found in infiltrating tumors and other sites of immune privilege, where they influence CD8+ T cells; CD4+ T-helper (Th)1, Th2, and Th17 cells; natural killer cells; and cells of myeloid lineage to choreograph and/or muck up host defense. Defining the cellular sources, mechanisms of action, and networking that distinguish the dynamic establishment of localized immune privilege is vital for developing strategic approaches to diminish or to embellish these tolerogenic events for therapeutic benefit.

    Topics: Animals; Humans; Immune System; Immune Tolerance; Neoplasms; Transforming Growth Factor beta

2006
Mutations of TGFbeta signaling molecules in human disease.
    Annals of medicine, 2006, Volume: 38, Issue:6

    The transforming growth factor beta (TGFbeta) signaling pathway regulates several biological processes including cellular proliferation, differentiation, apoptosis, migration, and extracellular matrix deposition. Ligand and receptor family members signal through two main Smad signaling branches, TGFbeta/activin to Smad2/3 (Sma and MAD-related proteins) and bone morphogenetic protein (BMP) to Smad1/5. At the molecular level, TGFbeta acts by modifying cytoskeletal organization and ultimately regulating expression of specific target genes. Germline disruption of TGFbeta signaling leads to several types of hereditary congenital malformation or dysfunction of the skeletal, muscular and/or cardiovascular systems, and to cancer predisposition syndromes. In this review, the molecular etiology of TGFbeta-associated disorders is examined, together with a discussion of clinical overlap between syndromes and possible biological explanations underlying the variable penetrance and expressivity of clinical characteristics. Increasing our understanding of the molecular etiology underlying genotype-phenotype correlations will ultimately provide a molecular-based approach that should result in better prognostic tools, smart therapeutics and individualized disease management, not only for these rare syndromes, but for more generalized disorders of the cardiovascular and musculoskeletal systems and cancer. The clinical consequence of TGFbeta signaling mutations appears to depend on environmental factors and on the basal levels of ongoing signaling transduction networks specific to each individual. In this respect, genetic background might be a central factor in determining disease outcome and treatment strategy for TGFbeta-associated diseases.

    Topics: Genetic Diseases, Inborn; Genetic Predisposition to Disease; Humans; Mutation; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2006
TGF beta inhibition for cancer therapy.
    Current cancer drug targets, 2006, Volume: 6, Issue:7

    The importance of perturbation in transforming growth factor beta (TGFbeta) signaling for the onset and progression of cancer is well established. Many tumors over express TGFbeta, and high circulating levels of TGFbeta1 in cancer patients are frequently associated with poor prognosis. TGFbeta has context-dependent biphasic action during tumorigenesis. Because of this, it is essential to take due care about the selection of patients most likely to benefit from anti-TGFbeta therapy. Anti-TGFbeta therapy aims to target both the tumor cell and the tumor microenvironment and may well have systemic effects of relevance to tumorigenesis. Extra-tumoral targets include stromal fibroblasts, endothelial and pericyte cells during angiogenesis, and the local and systemic immune systems, all of which can contribute to the pro-oncogenic effects of TGFbeta. Many different approaches have been considered, such as interference with ligand synthesis using oligonucleotides, sequestration of extracellular ligand using naturally-occurring TGFbeta binding proteins, recombinant proteins or antibodies, targeting activation of latent TGFbeta at the cell surface, or signal transduction within the cell. Consideration of which patients might benefit most from anti-TGFbeta therapy should include not only tumor responses to TGFbeta (which depend on activation of other oncogenic pathways in the cancer cell), but also germline genetic variation between individuals. Ultimately, a deep understanding of the interacting networks of signal pathways that regulate TGFbeta outcome in tumor and host cells should allow judicial choice of drugs. This review discusses the progress made in the pre-clinical and clinical testing of TGFbeta inhibitors, and discusses considerations of target populations and potential drug regimens.

    Topics: Animals; Antibodies, Monoclonal; Drug Design; Humans; Neoplasms; Oligonucleotides, Antisense; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2006
Inhibitors of the TGF-beta superfamily and their clinical applications.
    Mini reviews in medicinal chemistry, 2006, Volume: 6, Issue:11

    The transforming growth factor-beta (TGF-beta) superfamily includes TGF-betas, activin, myostatin and bone morphogenetic proteins. Misregulation of the activity of TGF-beta family members is involved in pathogenesis of cancer, muscular dystrophy, obesity and bone and tooth remodeling. Natural inhibitors for the TGF-beta superfamily regulate fine-tuning of activity of TGF-beta family in vivo. In addition to natural inhibitors for the TGF-beta family, soluble forms of receptors for the TGF-beta family, blocking monoclonal antibodies and small chemical TGF-beta inhibitors have been developed. In this review, we summarize recent advances in our understanding of inhibitors for the TGF-beta superfamily and their medical applications.

    Topics: Animals; Bone Morphogenetic Proteins; Humans; Muscular Diseases; Myostatin; Neoplasms; Protein Binding; Transforming Growth Factor beta

2006
Role of transforming growth factor-beta in cancer progression.
    Future oncology (London, England), 2006, Volume: 2, Issue:6

    Invasion and metastasis are the most lethal characteristics of cancer and the leading causes of cancer-related death. Transforming growth factor (TGF)-beta is a multifunctional cytokine that normally functions to prevent the uncontrolled proliferation of epithelial, endothelial and hematopoietic cells. Quite dichotomously, however, aberrant genetic or epigenetic events often negate the cytostatic function of TGF-beta in these cells, leading to tumor formation. Once freed from the growth-inhibitory effects of TGF-beta, cancer cells acquire the ability to proliferate, invade and metastasize when stimulated by TGF-beta. A thorough understanding of the molecular mechanisms underlying these paradoxical functions of TGF-beta remains elusive. Here, the authors review the tumor-suppressing and -promoting activities of TGF-beta and discuss the potential use and targeting of the TGF-beta-signaling system to prevent the progression and acquisition of metastatic phenotypes by human malignancies.

    Topics: Antineoplastic Agents; Disease Progression; Humans; Mutation; Neoplasms; Neoplastic Processes; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta

2006
Antisense therapy in clinical oncology: preclinical and clinical experiences.
    Methods in molecular medicine, 2005, Volume: 106

    Topics: Animals; Clinical Trials as Topic; Cyclic AMP-Dependent Protein Kinases; DNA Modification Methylases; Genes, ras; Humans; Leukemia; Neoplasms; Oligonucleotides, Antisense; Protein Kinase C; Protein Kinase C-alpha; Proto-Oncogene Proteins c-bcl-2; Proto-Oncogene Proteins c-myb; Proto-Oncogene Proteins c-raf; Transforming Growth Factor beta; Transforming Growth Factor beta2

2005
Duel nature of TGF-beta signaling: tumor suppressor vs. tumor promoter.
    Current opinion in oncology, 2005, Volume: 17, Issue:1

    Transforming growth factor beta type I (TGF-beta) is a ubiquitous cytokine that is well known for its ability to inhibit epithelial cell proliferation. Somatic mutations abrogating the TGF-beta signal transduction pathway are found in many gastrointestinal cancers, confirming its importance as a tumor suppressor. In contrast, many nongastrointestinal epithelial malignancies lack these somatic alterations, yet these cancers still acquire resistance to the growth-inhibitory effects of TGF-beta. In many instances, this resistance is part of a signaling switch whereby TGF-beta loses its growth inhibitory effects and is then used by the epithelial cell in a growth-promoting fashion. The mechanisms that underlie this change in the phenotypic growth response to TGF-beta are now being elucidated. This review focuses on recent advances in understanding the dual nature of the TGF-beta pathway as it relates to human carcinogenesis.. Elucidating the molecular basis that enables epithelial cells to change from a growth-suppressive to growth-stimulatory phenotype on TGF-beta exposure is an area of active research. Besides enhancing cancer cell growth, TGF-beta is also thought to promote a malignant cell's ability to metastasize by mediating changes in the cytoskeletal architecture, known as an epithelial-to-mesenchymal transition. This process enables a cancer cell to invade and spread to distal sites. Strong evidence has now emerged suggesting that the ability of a cell to use TGF-beta as a growth-promoting/invasive cytokine is a result of a number of different cellular and nuclear factors, including the absence or disruption of cyclin-dependent kinase inhibitors. This imbalance in cell cycle regulators may be the key element that dictates a cell's response to TGF-beta as growth-inhibitory versus growth-stimulatory, thus explaining the dual nature of TGF-beta signaling.. Current studies are beginning to shed light on the mechanisms that allow some nongastrointestinal epithelial cancers to evade the growth inhibitory effects of TGF-beta while simultaneously using this cytokine for growth advantage. By dissecting this phenotypic switch during tumor development, important genes, proteins, and pathways that are involved with TGF-beta signaling continue to be discovered. Knowledge of how premalignant cells and tumor cells respond to the growth promoting effects of TGF-beta and the genes that regulate this process will aid in the development of novel therapeutics and treatment strategies.

    Topics: Cell Transformation, Neoplastic; Cytokines; Epithelial Cells; Genes, Tumor Suppressor; Humans; Neoplasms; Phenotype; Promoter Regions, Genetic; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2005
Integrative radiation carcinogenesis: interactions between cell and tissue responses to DNA damage.
    Seminars in cancer biology, 2005, Volume: 15, Issue:2

    Tissue function requires coordinated multicellular behavior as a consequence of diverse signals integrated through the tissue microenvironment; importantly, these cell-cell and cell-microenvironment interactions also actively suppress cancer. Ionizing radiation (IR) elicits a well-defined cellular response to DNA damage that mediates the fate of the individual cell, concomitantly with a less well-characterized overarching tissue stress response that coordinates the response of multiple cell types via microenvironment signaling. We have now shown that these programs to reestablish homeostasis intersect via mutual regulation by transforming growth factor beta1 (TGF beta 1), which acts as an extracellular sensor and signal of stress. In this review, the concept that this type of functional integration of cell and tissue stress response programs is essential to cancer suppression will be discussed. Our experiments using IR, and several recent studies that experimentally manipulate stromal TGF beta, show that disruption of microenvironment signaling actively promotes malignant progression. Understanding the dynamic interactions between tissue and cell stress responses will be necessary for an accurate assessment of cancer risk and may also provide targets for prevention.

    Topics: Animals; Cell Communication; Cell Transformation, Neoplastic; DNA Damage; Humans; Neoplasms; Radiation, Ionizing; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2005
Transforming growth factor -beta receptor signaling in cancer.
    Frontiers in bioscience : a journal and virtual library, 2005, May-01, Volume: 10

    Transforming growth factor-beta (TGF-beta) is an ubiquitous cytokine that affects various biological processes, such as regulation of cell proliferation, immune responses, growth, differentiation, angiogenesis, and apoptosis of various cell types. The TGF-beta ligand initiates signaling by binding to and joining type I and II receptors of serine/threonine kinases. This review discusses the TGF-beta ligands and receptors, its positive and negative regulators in signaling, as well as its important roles with respect to tumor suppression and progression.

    Topics: Animals; Disease Progression; Humans; Neoplasms; Proteasome Endopeptidase Complex; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2005
Role of transforming growth factor Beta in human cancer.
    Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 2005, Mar-20, Volume: 23, Issue:9

    Transforming growth factor beta (TGF-beta) is a ubiquitous and essential regulator of cellular and physiologic processes including proliferation, differentiation, migration, cell survival, angiogenesis, and immunosurveillance. Alterations in the TGF-beta signaling pathway, including mutation or deletion of members of the signaling pathway and resistance to TGF-beta-mediated inhibition of proliferation are frequently observed in human cancers. Although these alterations define a tumor suppressor role for the TGF-beta pathway in human cancer, TGF-beta also mediates tumor-promoting effects, either through differential effects on tumor and stromal cells or through a fundamental alteration in the TGF-beta responsiveness of the tumor cells themselves. TGF-beta and members of the TGF-beta signaling pathway are being evaluated as prognostic or predictive markers for cancer patients. Ongoing advances in understanding the TGF-beta signaling pathway will enable targeting of this pathway for the chemoprevention and treatment of human cancers.

    Topics: Apoptosis; DNA-Binding Proteins; Humans; Neoplasm Metastasis; Neoplasms; Signal Transduction; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2005
Targeting TGFbeta signaling for cancer therapy.
    Cancer biology & therapy, 2005, Volume: 4, Issue:3

    Transforming growth factor (TGF) betas are multifunctional polypeptides that regulate several cellular functions, including cell growth and differentiation, extra cellular matrix production, motility and immunosuppression. The growth-inhibiting properties of TGFbeta have gained much attention into its role as a tumor suppressor. There is, however, now increasing evidence that TGFbeta switches roles, from tumor suppressor to tumor promoter, as the tumor progresses. Given the integral role of TGFbeta in the tumor progression, it follows that TGFbeta signaling offers an attractive target for cancer therapy. Several strategies including the use of antisense oligonucleotides for TGFbeta, TGFbeta antibodies, dominant negative TGFbeta receptor II, and small drug-molecules to inhibit TGFbeta receptor I kinase have shown great promise in the preclinical studies. These new findings, coupled with progressing clinical trials indicate that inhibition of TGFbeta signaling may, indeed, be a viable option to cancer therapy. This review summarizes the TGFbeta signaling, the dual role of TGFbeta--as a tumor suppressor and tumor promoter, and various strategies targeted against TGFbeta signaling for cancer therapy. The next few years promise to better our understanding of approaching cancer therapy with an eye to the inhibition of TGFbeta signaling.

    Topics: Animals; Humans; Mice; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2005
[TGF-beta family (TGF-beta, activin, BMP)].
    Nihon rinsho. Japanese journal of clinical medicine, 2005, Volume: 63 Suppl 4

    Topics: Activins; Apoptosis; Bone Morphogenetic Proteins; Cell Differentiation; DNA-Binding Proteins; Fibrosis; Neoplasms; Neurons; Osteoblasts; Proteasome Endopeptidase Complex; Protein Serine-Threonine Kinases; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Trans-Activators; Transcription, Genetic; Transforming Growth Factor beta; Ubiquitin

2005
[TGF-beta signaling and carcinogenesis].
    Nihon rinsho. Japanese journal of clinical medicine, 2005, Volume: 63 Suppl 4

    Topics: Animals; Cell Division; DNA-Binding Proteins; Humans; MDS1 and EVI1 Complex Locus Protein; Multigene Family; Mutation; Neoplasm Proteins; Neoplasms; Protein Binding; Proto-Oncogenes; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Trans-Activators; Transcription Factors; Transforming Growth Factor beta

2005
The RUNX genes: gain or loss of function in cancer.
    Nature reviews. Cancer, 2005, Volume: 5, Issue:5

    The RUNX genes have come to prominence recently because of their roles as essential regulators of cell fate in development and their paradoxical effects in cancer, in which they can function either as tumour-suppressor genes or dominant oncogenes according to context. How can this family of transcription factors have such an ambiguous role in cancer? How and where do these genes impinge on the pathways that regulate growth control and differentiation? And what is the evidence for a wider role for the RUNX genes in non-haematopoietic cancers?

    Topics: Animals; Core Binding Factor alpha Subunits; DNA-Binding Proteins; Genes, Tumor Suppressor; Humans; Leukemia; Mice; Mice, Knockout; Models, Biological; Mutation; Neoplasm Proteins; Neoplasms; Oncogenes; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Translocation, Genetic

2005
Transforming growth factor-beta signaling during epithelial-mesenchymal transformation: implications for embryogenesis and tumor metastasis.
    Cells, tissues, organs, 2005, Volume: 179, Issue:1-2

    The molecular mechanisms of epithelial-mesenchymal transformation (EMT) have long been studied to gain a greater understanding of this distinct change in cellular morphology. Early studies of the developing embryo have designated the involvement of Wnt signaling in EMT, through an activated complex of the lymphoid-enhancing factor-1 (LEF-1) transcription factor and the cell adhesion molecule beta-catenin. However, more recent studies have implicated a significant role of the transforming growth factor-beta (TGF-beta) in causing EMT in both development and pathology. The ability of TGF-beta isoforms to signal through a variety of molecules such as Smads, phosphatidylinositol 3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) creates an incredible complexity as to their role in this transition. Here we assess the biochemical signaling pathways of TGF-beta and their potential cross-interaction with traditional Wnt signaling molecules to bring about EMT during embryogenesis and tumor metastasis.

    Topics: Animals; beta Catenin; Cytoskeletal Proteins; DNA-Binding Proteins; Embryonic Development; Epithelium; Gene Expression Regulation, Developmental; Gene Expression Regulation, Neoplastic; Humans; Ligands; Lymphoid Enhancer-Binding Factor 1; MAP Kinase Signaling System; Mesoderm; Models, Biological; Neoplasm Metastasis; Neoplasms; Neural Crest; Neurons; Phosphorylation; Signal Transduction; Smad Proteins; Trans-Activators; Transcription Factors; Transforming Growth Factor beta

2005
TGF beta signalling and its role in tumour pathogenesis.
    Acta biochimica Polonica, 2005, Volume: 52, Issue:2

    Transforming growth factor beta (TGF-beta) is a multifunctional cytokine involved in the regulation of cell proliferation, differentiation and survival/or apoptosis of many cells. Knock-out experiments in mice for the three isoforms of TGF-beta have demonstrated their importance in regulating inflammation and tissue repair. TGF-beta is implicated in the pathogenesis of human diseases, including tissue fibrosis and carcinogenesis. TGF-beta receptors act through multiple intracellular pathways. Upon binding of TGF-beta with its receptor, receptor-regulated Smad2/3 proteins become phosphorylated and associate with Smad4. Such complex translocates to the nucleus, binds to DNA and regulates transcription of specific genes. Negative regulation of TGF-beta/Smad signalling may occur through the inhibitory Smad6/7. Furthermore, TGF-beta-activated kinase-1 (TAK1) is a component of TGF-beta signalling and activates stress-activated kinases: p38 through MKK6 or MKK3 and c-Jun N-terminal kinases (JNKs) via MKK4. In the brain TGF-beta, normally expressed at the very low level, increases dramatically after injury. Increased mRNA levels of the three TGF-beta isoforms correlate with the degree of malignancy of human gliomas. TGF-betas are secreted as latent precursors requiring activation into the mature form. TGF-beta may contribute to tumour pathogenesis by direct support of tumour growth and influence on local microenvironment, resulting in immunosuppression, induction of angiogenesis, and modification of the extracellular matrix. TGF-beta1,2 may stimulate production of vascular endothelial growth factor (VEGF) as well as plasminogen activator inhibitor (PAI-I), that are involved in vascular remodelling occurring during angiogenesis. Blocking of TGF-beta action inhibits tumour viability, migration, metastases in mammary cancer, melanoma and prostate cancer model. Reduction of TGF-beta production and activity may be a promising target of therapeutic strategies to control tumour growth.

    Topics: Animals; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2005
TGF-beta inhibitors for the treatment of cancer.
    Expert opinion on investigational drugs, 2005, Volume: 14, Issue:6

    Advances in understanding the role of transforming growth factor (TGF)-beta in tumorigenesis have led to the development of TGF-beta inhibitors for cancer treatment. Three platforms of TGF-beta inhibitors have evolved: antisense oligonucleotides, monoclonal antibodies and small molecules. In this review, the current stage of development of each known TGF-beta inhibitor will be discussed. As part of the risk/benefit assessment of TGF-beta inhibitors, the known effects of TGF-beta deficiency in mice, non-clinical toxicology studies with TGF-beta inhibitors in rats, and the clinical studies with monoclonal antibodies against TGF-beta will be summarised.

    Topics: Animals; Antineoplastic Agents; Clinical Trials as Topic; Drug Industry; Humans; Neoplasms; Transforming Growth Factor beta

2005
PTHrP gene expression in cancer: do all paths lead to Ets?
    Critical reviews in eukaryotic gene expression, 2005, Volume: 15, Issue:2

    Parathyroid hormone-related protein (PTHrP) came to the attention of the scientific community in the mid-1980s because of its association with the paraneoplastic syndrome of humoral hypercalcemia of malignancy. Recently, a crucial role for the peptide has been identified in the metastatic growth of cancer cells in bone. Efforts to understand the peptide's role in these pathological processes have evolved into the study of PTHrP gene expression. Currently, regulation of the third PTHrP promoter is beginning to be understood in the context of activation of certain signaling pathways involved in the growth and progression of specific neoplasms. In addition, factors that modulate the entire PTHrP-transcriptional unit, as well as the stability of the mRNA, are being elucidated at the level of cis-acting sequences.

    Topics: Base Sequence; Bone Neoplasms; Gene Expression; Gene Expression Regulation; Gene Expression Regulation, Neoplastic; Humans; Methylation; Mitogen-Activated Protein Kinase Kinases; Neoplasms; Parathyroid Hormone-Related Protein; Promoter Regions, Genetic; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-ets; Transcription Factors; Transforming Growth Factor beta; Vitamin D

2005
Role of the small leucine-rich proteoglycan (SLRP) family in pathological lesions and cancer cell growth.
    Journal of Nippon Medical School = Nippon Ika Daigaku zasshi, 2005, Volume: 72, Issue:3

    The roles of lumican, a member of the small-leucine-rich-proteoglycan (SLRP) family, in pathological fibrosis, cancer tissues and tumor cell growth were reviewed. Lumican is predominantly localized in the areas of pathological fibrosis including the thickened intima of human coronary arteries, ischemic and reperfused hearts, and acute pancreatitis and chronic pancreatitis (CP)-like lesions adjacent to pancreatic cancer nests. In these lesions, lumican mRNA and protein were transiently and ectopically overexpressed in most of the vascular smooth muscle cells (VSMCs) that migrated into the thickened intima, myocardial cells adjacent to an ischemic lesion, acinar cells, islet cells and fibroblasts of pathological pancreatic tissues. The low expression level of lumican in breast cancer is associated with rapid progression and poor survival. Lumican mRNA in breast cancer is overexpressed in fibroblasts adjacent to cancer cells but not in cancer cells. Furthermore, the high expression level of lumican is associated with a high pathological tumor grade, a low estrogen receptor level in the cancer tissues, and young age of patients. The suppression of lumican expression in culture cells induces their cell growth. Lumican-transfected tumor cells are characterized by a strong suppression of their anchorage-independent growth and capacity of invasion. Lumican significantly suppressed subcutaneous tumor formation in syngenic mice, with a concomitant decrease in cyclin D1 expression level, and induced and/or enhanced the apoptosis of these cells. The autocrine mechanism in cancer cells and the paracrine mechanism in cancer cells and fibroblasts via transforming growth factor (TGF)-beta and Smad signals may play important roles in the regulation of tumor growth by SLRPs.

    Topics: Animals; Apoptosis; Cell Communication; Cell Division; Chondroitin Sulfate Proteoglycans; DNA-Binding Proteins; Fibrosis; Gene Expression; Humans; Keratan Sulfate; Lumican; Mice; Muscle, Smooth, Vascular; Neoplasm Invasiveness; Neoplasm Staging; Neoplasms; RNA, Messenger; Smad Proteins; Trans-Activators; Transforming Growth Factor beta

2005
CD25+ CD4+ regulatory T-cells in cancer.
    Immunologic research, 2005, Volume: 32, Issue:1-3

    Regulatory T-cells (Treg) protect the host from autoimmune disease by suppressing self-reactive immune cells. As such, Treg may also block antitumor immune responses. Recent observations by us and others showed that the prevalence of Treg is increased in cancer patients, particularly in the tumor environment. Our studies in a mouse pancreas cancer model suggest that the tumor actively promotes the accrual of Treg through several mechanisms involving activation of naturally occurring Treg as well as conversion of non-Treg into Treg. Our studies focus on further defining these mechanisms with the ultimate goal of designing strategies that block Treg-mediated suppression in cancer patients.

    Topics: Animals; Biomarkers, Tumor; Breast Neoplasms; Cancer Vaccines; Carcinoma, Ductal, Breast; CD4 Antigens; Dendritic Cells; Female; Forkhead Transcription Factors; Humans; Immune Tolerance; Lymphocyte Depletion; Mice; Mice, Knockout; Neoplasms; Pancreatic Neoplasms; Phenotype; Receptors, Interleukin-2; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2005
Cripto-1: a multifunctional modulator during embryogenesis and oncogenesis.
    Oncogene, 2005, Aug-29, Volume: 24, Issue:37

    It is increasingly evident that genes known to perform critical roles during early embryogenesis, particularly during stem cell renewal, pluripotentiality and survival, are also expressed during the development of cancer. In this regard, oncogenesis may be considered as the recapitulation of embryogenesis in an inappropriate temporal and spatial manner. The epidermal growth factor-Cripto-1/FRL1/cryptic family of proteins consists of extracellular and cell-associated proteins that have been identified in several vertebrate species. During early embryogenesis, epidermal growth factor-Cripto-1/FRL1/cryptic proteins perform an obligatory role as coreceptors for the transforming growth factor-beta subfamily of proteins, which includes Nodal. Cripto-1 has also been shown to function as a ligand through a Nodal/Alk4-independent signaling pathway that involves binding to glypican-1 and the subsequent activation through src of phosphoinositol-3 kinase/Akt and ras/mitogen-activated protein kinase intracellular pathways. Expression of Cripto-1 is increased in several human cancers and its overexpression is associated with the development of mammary tumors in mice. Here, we review the role of Cripto-1 during embryogenesis, cell migration, invasion and angiogenesis and how these activities may relate to cellular transformation and tumorigenesis. We also briefly discuss evidence suggesting that Cripto-1 may be involved in stem cell maintenance.

    Topics: Activin Receptors, Type I; Cell Movement; Cell Transformation, Neoplastic; DNA-Binding Proteins; Embryonic Development; Epidermal Growth Factor; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; MAP Kinase Signaling System; Membrane Glycoproteins; Milk Proteins; Neoplasm Proteins; Neoplasms; Neovascularization, Physiologic; Nodal Protein; Protein Serine-Threonine Kinases; Proteins; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Signal Transduction; Smad2 Protein; Trans-Activators; Transforming Growth Factor beta; Wnt Proteins

2005
Crosstalk mechanisms between the mitogen-activated protein kinase pathways and Smad signaling downstream of TGF-beta: implications for carcinogenesis.
    Oncogene, 2005, Aug-29, Volume: 24, Issue:37

    Transforming growth factor-beta (TGF-beta) superfamily members signal via membrane-bound heteromeric serine-threonine kinase receptor complexes. Upon ligand-binding, receptor activation leads to phosphorylation of cytoplasmic protein substrates of the Smad family. Following phosphorylation and oligomerization, the latter move into the nucleus to act as transcription factors to regulate target gene expression. TGF-beta responses are not solely the result of the activation Smad cascade, but are highly cell-type specific and dependent upon interactions of Smad signaling with a variety of other intracellular signaling mechanisms, initiated or not by TGF-beta itself, that may either potentiate, synergize, or antagonize, the rather linear TGF-beta/Smad pathway. These include, (a), regulation of Smad activity by mitogen-activated protein kinases (MAPKs), (b), nuclear interaction of activated Smads with transcriptional cofactors, whether coactivators or corepressors, that may be themselves be regulated by diverse signaling mechanisms, and (c), negative feedback loops exerted by inhibitory Smads, transcriptional targets of the Smad cascade. This review focuses on how MAPKs modulate the outcome of Smad activation by TGF-beta, and how cross-signaling mechanisms between the Smad and MAPK pathways may take place and affect cell fate in the context of carcinogenesis.

    Topics: DNA-Binding Proteins; Enzyme Activation; Humans; MAP Kinase Signaling System; Neoplasms; Signal Transduction; Smad Proteins; Trans-Activators; Transcription Factors; Transforming Growth Factor beta

2005
[The role of TGF beta].
    Polskie Archiwum Medycyny Wewnetrznej, 2005, Volume: 113, Issue:4

    Topics: Apoptosis; Extracellular Matrix; Fibrosis; Humans; Neoplasms; Transforming Growth Factor beta

2005
RNF11 is a multifunctional modulator of growth factor receptor signalling and transcriptional regulation.
    European journal of cancer (Oxford, England : 1990), 2005, Volume: 41, Issue:16

    Our laboratory has found that the 154aa RING finger protein 11 (RNF11), has modular domains and motifs including a RING-H2 finger domain, a PY motif, an ubiquitin interacting motif (UIM), a 14-3-3 binding sequence and an AKT phosphorylation site. RNF11 represents a unique protein with no other known immediate family members yet described. Comparative genetic analysis has shown that RNF11 is highly conserved throughout evolution. This may indicate a conserved and non-redundant role for the RNF11 protein. Molecular binding assays using RNF11 have shown that RNF11 has important roles in growth factor signalling, ubiquitination and transcriptional regulation. RNF11 has been shown to interact with HECT-type E3 ubiquitin ligases Nedd4, AIP4, Smurf1 and Smurf2, as well as with Cullin1, the core protein in the multi-subunit SCF E3 ubiquitin ligase complex. Work done in our laboratory has shown that RNF11 is capable of antagonizing Smurf2-mediated inhibition of TGFbeta signalling. Furthermore, RNF11 is capable of degrading AMSH, a positive regulator of both TGFbeta and EGFR signalling pathways. Recently, we have found that RNF11 can directly enhance TGFbeta signalling through a direct association with Smad4, the common signal transducer and transcription factor in the TGFbeta, BMP, and Activin pathways. Through its association with Smad4 and other transcription factors, RNF11 may have a role in direct transcriptional regulation. Our laboratory and others have found nearly 80 protein interactions for RNF11, placing RNF11 at the cross-roads of cell signalling and transcriptional regulation. RNF11 is highly expressed in breast tumours. Deregulation of RNF11 function may prove to be harmful to patient therapeutic outcomes. RNF11 may therefore provide a novel target for cancer therapeutics. The purpose of this review is to discuss the role of RNF11 in cell signalling and transcription factor modulation with special attention given to the ubiquitin-proteasomal pathway, TGFbeta pathway and EGFR pathway.

    Topics: Carrier Proteins; DNA Damage; DNA-Binding Proteins; Humans; Neoplasms; Phosphorylation; Receptors, Growth Factor; Signal Transduction; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Ubiquitin

2005
Mdm2 in growth signaling and cancer.
    Growth factors (Chur, Switzerland), 2005, Volume: 23, Issue:3

    Genetic and biochemical evidence have demonstrated a direct link between Mdm2 and cancer development. Elevated expression of Mdm2 is observed in a significant proportion of different types of cancer. The major contribution of Mdm2 to the development of cancer is through a tight inhibition of the activities and stability of the tumor suppressor p53. However, extensive studies over the past few years have identified p53-independent functions of Mdm2, in the regulation of several important cellular processes and multiple signaling pathways. The promotion of cell cycle progression by Mdm2 is mediated via p53 inhibition, and by regulating the pRb/E2F complex. Mdm2 is an important mediator of growth and survival signaling in the PI3K/Akt pathway, an activator of certain steroid hormone receptors, and an inhibitor of the TGF-beta growth restrictive pathway. Thus, the impact on these pathways by deregulated Mdm2, as often observed in cancer, can be oncogenic in a permissible environment. This renders Mdm2 as an important target for the development of anti-cancer drugs.

    Topics: Animals; Cell Survival; Growth Substances; Humans; Mice; Neoplasms; Oncogene Proteins; Proto-Oncogene Proteins c-mdm2; Receptors, Steroid; Signal Transduction; Transforming Growth Factor beta

2005
Integrin-mediated activation of latent transforming growth factor beta.
    Cancer metastasis reviews, 2005, Volume: 24, Issue:3

    Members of the integrin family recognize a variety of spatially-restricted extracellular ligands. Classically, ligation of integrins activates cytoplasmic signals in the integrin-expressing cell and contributes to cell adhesion, migration, proliferation and survival. At least two members of this family, alphavbeta6 and alphavbeta8 perform an additional function, activation of latent complexes of transforming growth factor beta. In effect, this process allows integrins on one cell to activate signals on adjacent (in the case of alphavbeta6) or nearby cells (in the case of alphavbeta8). Integrin-mediated TGFbeta activation has been shown to play important roles in modulating tissue fibrosis, acute lung injury and pulmonary emphysema. Given the important roles that TGFbeta plays in modulating epithelial cell growth, epithelial-to-mesenchymal transformation and tumor invasion and metastasis, integrin-mediated TGFbeta activation is likely to play important roles in tumor growth and metastasis.

    Topics: Animals; Antigens, Neoplasm; Cell Adhesion; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Survival; Cytoplasm; Epithelial Cells; Gene Expression Regulation, Neoplastic; Humans; Integrins; Models, Biological; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Transforming Growth Factor beta

2005
Can risk of radiotherapy-induced normal tissue complications be predicted from genetic profiles?
    Acta oncologica (Stockholm, Sweden), 2005, Volume: 44, Issue:8

    Over the last decade, increasing efforts have been taken to establish associations between various genetic germline alterations and risk of normal tissue complications after radiotherapy. Though the studies have been relatively small and methodologically heterogeneous, preliminary indications have been provided that single nucleotide polymorphisms in the genes TGFB1 and ATM may modulate risk of particularly late toxicity. In addition, rare ATM alterations may enhance complication susceptibility. Nevertheless, we are still far from having an exhaustive understanding of the genetics that may underlie differences in clinical normal tissue radiosensitivity. Recent technical advances and emerging insights to the structure of inter-individual genetic variation open up unprecedented opportunities to dissect the molecular and genetic basis of normal tissue radiosensitivity. However, to fully exploit these new possibilities well-planed large-scale clinical studies are mandatory. Currently, international initiatives are taken to establish the bio banks and databases needed for this task.

    Topics: Ataxia Telangiectasia Mutated Proteins; Cell Cycle Proteins; Cell Survival; DNA-Binding Proteins; Genetic Markers; Humans; Models, Genetic; Neoplasms; Protein Serine-Threonine Kinases; Radiation Genetics; Radiation Injuries; Radiation Tolerance; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Suppressor Proteins

2005
The fibroblastic coconspirator in cancer progression.
    Cold Spring Harbor symposia on quantitative biology, 2005, Volume: 70

    A remarkable change has occurred in the thinking about epithelial-derived cancer in recent years: From almost entirely focusing on oncogenes and tumor suppressor genes has come the realization that the tumor microenvironment is a coconspirator in the carcinogenic process. Many types of stromal cells, including fibroblasts, adipocytes, macrophages, mast cells, and cells of the vascular system, are crucial contributors to epithelial carcinogenesis. Here, we focus on the fibroblast's role in cancer progression and the molecules involved in the communications between the fibroblasts and the cancer cells, including fibroblast secreted protein 1 (FSP-1 or S100A4), transforming growth factor beta (TGF-beta), the chemokine CXCL-12 (stromal derived factor 1 alpha, SDF-1alpha), type I collagen, and matrix metalloproteinase 13 (MMP-13).

    Topics: Animals; Calcium-Binding Proteins; Chemokine CXCL12; Chemokines, CXC; Collagen Type I; Fibroblasts; Humans; Matrix Metalloproteinases; Models, Biological; Neoplasms; S100 Calcium-Binding Protein A4; S100 Proteins; Transforming Growth Factor beta

2005
Antiangiogenic cancer therapy.
    Seminars in cancer biology, 2004, Volume: 14, Issue:2

    Like most embryonic tissues, tumors have the ability to build up their own blood vessel networks. However, the architecture of tumor vessels is fundamentally different from that found in healthy tissues. Tumor vessels are usually irregular, heterogeneous, leaky, and poorly associated with mural cells. Endothelial cells in tumor vessels are also disorganized and express imbalanced surface molecules. These unusual features may provide some molecular and structural basis for selective inhibition or even destruction of tumor vessels by angiogenesis inhibitors. In animal tumor models, several angiogenesis inhibitors seem to inhibit tumor angiogenesis specifically without obvious effects on the normal vasculature. As a result, these inhibitors produced potent antitumor effects in mice. Excited by these preclinical studies, more than 60 angiogenesis inhibitors are being evaluated for their anticancer effects in human patients. Although the ultimate outcome of antiangiogenic clinical trials remains to be seen, several early observations have reported some disappointing results. These early clinical data have raised several important questions. Can we cure human cancers with angiogenesis inhibitors? Have we found the ideal angiogenesis inhibitors for therapy? What is the difference between angiogenesis in an implanted mouse tumor and in a spontaneous human tumor? What are the molecular mechanisms of these angiogenesis inhibitors? Should angiogenesis inhibitors be used alone or in combinations with other existing anticancer drugs? In this review, we will discuss these important issues in relation to ongoing antiangiogenic clinical trials.

    Topics: Administration, Oral; Angiogenesis Inhibitors; Angiostatins; Animals; Fibroblast Growth Factor 2; Humans; Mice; Neoplasms; Neovascularization, Pathologic; Thymidine Phosphorylase; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2004
TGFbeta1, back to the future: revisiting its role as a transforming growth factor.
    Cancer biology & therapy, 2004, Volume: 3, Issue:3

    TGFbeta1 was initially identified in culture media from transformed cells as part of a factor that could produce a transformed phenotype in a nontransformed cell line. Subsequently this activity was separated into TGFbeta and TGFalpha an EGF receptor ligand. With the discovery that TGFbeta1 was a potent growth inhibitor of epithelial cells, and the identification of inactivating mutations within the TGFbeta1 signaling pathway in cancers it became clear that TGFbeta1 signaling is a tumor suppressor pathway for early stages of cancer. However many human carcinomas overexpress TGFbeta1 and this is associated with poor patient prognosis and increased frequency of metastasis. Similar results have been obtained with tumor cell lines and experimental animal models. Thus stage specific duality of function is the emerging paradigm for the role of TGFbeta1 in cancer. This review will focus on the evidence for TGFbeta1 as a tumor promoting and metastasis factor and examine the biological and molecular basis for these effects. It is proposed that the switch from tumor suppressor to oncogene reflects genetic or epigenetic alterations in signaling pathways in tumor cells that alter the readout from the TGFbeta1 pathway.

    Topics: Animals; Carcinoma; Disease Models, Animal; Disease Progression; Humans; Immune Tolerance; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Phenotype; Prognosis; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factor beta1

2004
Epithelial metaplasia: an inadequate environment for antitumour immunity?
    Trends in immunology, 2004, Volume: 25, Issue:4

    Topics: Animals; Antigen-Presenting Cells; Cell Adhesion Molecules; Chemokines; Cytokines; Epithelium; Humans; Metaplasia; Models, Biological; Neoplasms; Prostaglandins; Transforming Growth Factor beta; Transforming Growth Factor beta1

2004
The role of translation in neoplastic transformation from a pathologist's point of view.
    Oncogene, 2004, Apr-19, Volume: 23, Issue:18

    Increased cell proliferation, which is a hallmark of aggressive malignant neoplasms, requires a general increase in protein synthesis and a specific increase in the synthesis of replication-promoting proteins. Transient increase in the general protein synthesis rate, as well as preferential translation of specific mRNAs coding for growth promoting proteins (e.g. cyclin D1), takes place during normal mitogenic response. A number of extensively studied growth signal transduction pathways (Ras, PI3K, MAPK, mTOR-dependent pathways) activate the function and expression of various components of the translational machinery. In abnormal situations, constitutive activation of signal transduction pathways (e.g. oncogenic activation of Ras or Myc) leads to continuous upregulation of key elements of translational machinery. On the other hand, tumor suppressor genes (p53, pRb) downregulate ribosomal and tRNA synthesis, and their inactivation results in uncontrolled production of these translational components. During recent years, a significant effort has been dedicated to determining whether expression of translation factors is increased in human tumors using clinical biopsy specimens. The results of these studies indicate that expression of particular translation initiation factors is not always increased in human neoplasms. The pattern of expression is characteristic for a particular tumor type. For example, eIF-4E is usually increased in bronchioloalveolar carcinomas but not in squamous cell carcinomas of the lung. Interestingly, in certain highly proliferative and aggressive neoplasms (e.g. squamous cell carcinoma of the lung, melanoma), the expression of eIF-4E is barely detectable. These findings suggest that mechanisms for increasing general protein synthesis in various neoplasms differ significantly. Finally, the possibility of qualitative alterations in the translational machinery, rather than a simple increase in the activity of its components, is discussed along with the possibility of targeting those qualitative differences for tumor therapy.

    Topics: Animals; Apoptosis; Cell Division; Cell Transformation, Neoplastic; Eukaryotic Initiation Factor-2; Eukaryotic Initiation Factor-4E; Gene Expression Regulation; Humans; Neoplasms; Protein Biosynthesis; Proto-Oncogene Proteins c-myc; Retinoblastoma Protein; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2004
Postgenomic global analysis of translational control induced by oncogenic signaling.
    Oncogene, 2004, Apr-19, Volume: 23, Issue:18

    It is commonly assumed that developmental and oncogenic signaling achieve their phenotypic effects primarily by directly regulating the transcriptional profile of cells. However, there is growing evidence that the direct effect on transcription may be overshadowed by differential effects on the translational efficiency of specific existing mRNA species. Global analysis of this effect using microarrays indicates that this mechanism of controlling protein production provides a highly specific, robust, and rapid response to oncogenic and developmental stimuli. The mRNAs so affected encode proteins involved in cell-cell interaction, signal transduction, and growth control. Furthermore, a large number of transcription factors capable of secondarily rearranging the transcriptional profile of the cell are controlled at this level as well. To what degree this translational control is either necessary or sufficient for tumor formation or maintenance remains to be determined.

    Topics: 3' Untranslated Regions; Animals; Eukaryotic Initiation Factor-4E; Gene Expression Profiling; Humans; Neoplasms; Oligonucleotide Array Sequence Analysis; Phosphatidylinositol 3-Kinases; Protein Biosynthesis; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; ras Proteins; Ribosomes; RNA, Messenger; Signal Transduction; Transforming Growth Factor beta

2004
The role of Fas ligand and transforming growth factor beta in tumor progression: molecular mechanisms of immune privilege via Fas-mediated apoptosis and potential targets for cancer therapy.
    Cancer, 2004, Jun-01, Volume: 100, Issue:11

    Despite the fact that expression of Fas ligand (FasL) in cytotoxic T lymphocytes (CTLs) and in natural killer (NK) cells plays an important role in Fas-mediated tumor killing, During tumor progression FasL-expressing tumor cells are involved in counterattacking to kill tumor-infiltrating lymphocytes (TILs). Soluble FasL levels also increase with tumor progression in solid tumors, and this increase inhibits Fas-mediated tumor killing by CTLs and NK cells. The increased expression of FasL in tumor cells is associated with decreased expression of Fas; and the promoter region of the FASL gene is regulated by transcription factors, such as neuronal factor kappaB (NF-kappaB) and AP-1, in the tumor microenvironment. Although the ratio of FasL expression to Fas expression in tumor cells is not strongly related to the induction of apoptosis in TILs, increased expression of FasL is associated with decreased Fas levels in tumor cells that can escape immune surveillance and facilitate tumor progression and metastasis. Transforming growth factor beta (TGF-beta) is a potent growth inhibitor and has tumor-suppressing activity in the early phases of carcinogenesis. During subsequent tumor progression, the increased secretion of TGF-beta by both tumor cells and, in a paracrine fashion, stromal cells, is involved in the enhancement of tumor invasion and metastasis accompanied by immunosuppression. Herein, the authors review the clinical significance of FasL and TGF-beta expression patterns as features of immune privilege accompanying tumor progression in the tumor microenvironment. Potential strategies for identifying which molecules can serve as targets for effective antitumor therapy also are discussed.

    Topics: Apoptosis; Fas Ligand Protein; Humans; Lymphocytes, Tumor-Infiltrating; Membrane Glycoproteins; Neoplasms; Neutrophils; Receptors, Tumor Necrosis Factor; Signal Transduction; Transforming Growth Factor beta

2004
Role of TGF-beta in cancer and the potential for therapy and prevention.
    Expert review of anticancer therapy, 2004, Volume: 4, Issue:4

    Transforming growth factor (TGF)-beta is a naturally occurring potent inhibitor of cell growth. TGF-beta binds first to a Type II (TGFBR2), then a Type I receptor (TGFBR1). TGFBR1 activation results in the phosphorylation of intracellular messengers, the SMADs. Unrestricted cell growth due to decreased growth inhibitory activity is a paramount feature of a defect in TGF-beta function. There is growing evidence that common variants of the TGF-beta pathway ligand and receptors that alter TGF-beta signaling modify cancer risk. Approximately 14% of the general population carry TGFBR1*6A, a variant of the TGFBR1 gene that results in decreased TGF-beta-mediated growth inhibition. Recent studies show that overall cancer risk is increased by 70 and 19% among TGFBR1*6A homozygotes and heterozygotes, respectively. This suggests that TGFBR1*6A may contribute to the development of a large proportion of common forms of cancer and may become a target for cancer chemoprevention. While decreased TGF-beta signaling increases cancer risk, TGF-beta secretion and activated TGF-beta signaling enhances the aggressiveness of several types of tumors. The activated TGF-beta signaling pathway is emerging as an attractive target in cancer and the authors predict that assessment of functionally relevant variants of this pathway will lead to the identification of individuals with a higher cancer risk and account for some forms of familial cancer susceptibility. In addition, it is predicted that inhibitors of the TGF-beta signaling pathway will find their way into cancer clinical trials, leading to delays in tumor progression and improvements in overall survival.

    Topics: Activin Receptors, Type I; Genetic Predisposition to Disease; Humans; Ligands; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Risk Factors; Signal Transduction; Transforming Growth Factor beta

2004
APC dosage effects in tumorigenesis and stem cell differentiation.
    The International journal of developmental biology, 2004, Volume: 48, Issue:5-6

    It is well established that concentration gradients of signaling molecules (the so-called "morphogens") organize and pattern tissues in developing animals. In particular, studies in Drosophila and different vertebrates have shown that gradients of the Wnt, Hedgehog (Hh) and transforming growth factor-beta (TGF-beta) families of morphogens play critical roles in limb patterning. Morphogens are often expressed in organizing centres and can act over a long range to coordinate the patterning of an entire field of cells. These observations imply that exposure to different concentrations of these diffusible factors may trigger differential cellular responses. In order to study these dosage-dependent Wnt/beta-catenin signaling effects, we have generated several hypomorphic mutant alleles at the mouse Apc locus and studied their cellular and phenotypic outcomes in stem cell renewal and differentiation, and in tumorigenesis. The results clearly show that Apc mutations differentially affect the capacity of stem cells to differentiate in a dosage-dependent fashion. Likewise, different Apc mutations (and the corresponding Wnt signaling dosages) confer different degrees of susceptibility to tumorigenesis in the corresponding mouse models. These results have implications for the understanding of the molecular and cellular basis of tumor initiation by defects in the Wnt pathway. We propose a model in which adult somatic stem cell compartments are characterized by tissue-specific beta-catenin threshold levels for cell proliferation, differentiation and apoptosis. Different APC mutations will result in different levels of beta-catenin signaling, thus conferring different degrees of tumor susceptibility in different tissues. Hence, beta-catenin dosage-dependent effects may not only explain how a single pathway is involved in the development and homeostasis of different tissues, but also its pleiotrophic role in tumorigenesis.

    Topics: Adenomatous Polyposis Coli Protein; Alleles; Animals; Cell Differentiation; Colorectal Neoplasms; Disease Progression; Dose-Response Relationship, Drug; Genes, APC; Mice; Models, Biological; Models, Genetic; Mutation; Neoplasms; Phenotype; Signal Transduction; Stem Cells; Transforming Growth Factor beta

2004
Cancer progression: is inhibin alpha from Venus or Mars?
    Cytokine & growth factor reviews, 2004, Volume: 15, Issue:5

    The inhibin field has been perplexed by the information that inhibin alpha is a tumour suppressor in mice yet is elevated in women with ovarian cancer. Furthermore, we have consistently observed a down-regulation or loss of inhibin alpha in prostate cancer patient samples and cell lines. However, our latest data have prompted us to re-evaluate the role of inhibin alpha in prostate and other cancers. Using the analogy of TGF-beta as a springboard for our hypothesis, we offer a unifying model whereby the previously conflicting observations in mice, men and women can be explained. We propose that initially inhibin alpha is tumour-suppressive and is expressed in benign and early-stage primary cancers. Tumour-suppressive inhibin alpha is then silenced as the tumour progresses but is reactivated as a pro-metastatic factor in advanced, aggressive cancers.

    Topics: Animals; Carcinogens; Disease Progression; Female; Genes, Tumor Suppressor; Humans; Inhibins; Male; Mice; Models, Biological; Neoplasm Proteins; Neoplasms; Ovarian Neoplasms; Prostatic Neoplasms; Severity of Illness Index; Transforming Growth Factor beta

2004
Cytokine overexpression and constitutive NFkappaB in cancer.
    Cell cycle (Georgetown, Tex.), 2004, Volume: 3, Issue:9

    The NFkappaB family of transcription factors, central mediators of immune responses, are also involved in oncogenesis. Loss of regulation of the normally latent NFkappaB contributes importantly to the deregulated growth, resistance to apoptosis and propensity to metastasize observed in many cancers. Thus, pathways for activation of NFkappaB are promising targets for new agents that may help to prevent or treat cancer. We find that the abnormal secretion of multiple cytokines that activate NFkappaB by binding to cell-surface receptors is one of the major causes of constitutive NFkappaB activity in cancer. A novel finding is that the latent form of TGFbeta, secreted by some of these cells, can activate NFkappaB. To understand the basis of this abnormal cytokine secretion, we are using forward genetic methods to identify specific causative mutations in cancer cells.

    Topics: Animals; Cell Transformation, Neoplastic; Cytokines; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; NF-kappa B; Oncogenes; Receptors, Cell Surface; Transcriptional Activation; Transforming Growth Factor beta

2004
High interstitial fluid pressure - an obstacle in cancer therapy.
    Nature reviews. Cancer, 2004, Volume: 4, Issue:10

    Many solid tumours show an increased interstitial fluid pressure (IFP), which forms a barrier to transcapillary transport. This barrier is an obstacle in tumour treatment, as it results in inefficient uptake of therapeutic agents. There are a number of factors that contribute to increased IFP in the tumour, such as vessel abnormalities, fibrosis and contraction of the interstitial matrix. Lowering the tumour IFP with specific signal-transduction antagonists might be a useful approach to improving anticancer drug efficacy.

    Topics: Alprostadil; Animals; Antineoplastic Agents; Biological Transport; Bradykinin; Extracellular Fluid; Humans; Hyaluronoglucosaminidase; Hydrostatic Pressure; Mice; Neoplasms; Niacinamide; Platelet-Derived Growth Factor; Signal Transduction; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Vascular Endothelial Growth Factor A

2004
Development of TGF-beta signalling inhibitors for cancer therapy.
    Nature reviews. Drug discovery, 2004, Volume: 3, Issue:12

    The transforming growth factor-beta (TGF-beta) superfamily of ligands has a pivotal role in the regulation of a wide variety of physiological processes from development to pathogenesis. Since the discovery of the prototypic member, TGF-beta, almost 20 years ago, there have been tremendous advances in our understanding of the complex biology of this superfamily. Deregulation of TGF-beta has been implicated in the pathogenesis of a variety of diseases, including cancer and fibrosis. Here we present the rationale for evaluating TGF-beta signalling inhibitors as cancer therapeutics, the structures of small-molecule inhibitors that are in development and the targeted drug discovery model that is being applied to their development.

    Topics: Animals; Antineoplastic Agents; Drug Design; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2004
Cellular signaling pathways affect the function of ribonucleotide reductase mRNA binding proteins: mRNA stabilization, drug resistance, and malignancy (Review).
    International journal of oncology, 2003, Volume: 22, Issue:1

    Ribonucleotide reductase is an enzyme that is essential for DNA synthesis and repair. It is composed of 2 dimeric proteins called R1 and R2 that are both necessary for enzymatic activity that reduces ribonucleotides to deoxyribonucleotides. This is the rate-limiting reaction that provides a supply of precursors for DNA synthesis therefore it is essential for cell proliferation. The importance of understanding the complex regulation of ribonucleotide reductase is emphasized by observations that mechanisms controlling its expression and activity may be altered during malignant cell proliferation which leads to drug resistance, making it a useful target to develop chemotherapeutic compounds in the treatment of cancer. Expression studies with the R1 and R2 genes have provided evidence for a direct role for the components of ribonucleotide reductase in determining malignant potential. Ribonucleotide reductase is regulated by transcriptional activation of gene expression and post-transcriptional mechanisms that alter mRNA message stability. Post-transcriptional regulation of mRNA turnover plays an important role in modulating mRNA steady state levels and therefore directly influences gene expression. The 3'-untranslated region (UTR) of R1 and R2 messages contain sequences that are important in regulating gene expression through changes in message stability. Studies have found that mRNA message stability is mediated by growth factors, cytokines and tumor promoters. Several studies have elucidated signal transduction pathways of tumor promoters, TGF-beta and oxidation/reduction agents. This report reviews how knowledge of these signaling pathways is revealing new insights into how ribonucleotide reductase mRNA binding proteins are important in regulating cellular proliferation, drug resistance and malignancy.

    Topics: 3' Untranslated Regions; Animals; Drug Resistance, Neoplasm; Humans; Neoplasms; Protein Binding; Protein Kinase C; Ribonucleotide Reductases; RNA-Binding Proteins; RNA, Messenger; Tetradecanoylphorbol Acetate; Transforming Growth Factor beta; Transforming Growth Factor beta1

2003
TGF beta/Smad signaling system and its pathologic correlates.
    American journal of medical genetics. Part A, 2003, Jan-01, Volume: 116A, Issue:1

    Topics: Animals; Autoimmune Diseases; Craniosynostoses; DNA-Binding Proteins; Humans; Mutation; Neoplasms; Signal Transduction; Smad2 Protein; Trans-Activators; Transforming Growth Factor beta

2003
Antitumor effects of bisphosphonates.
    Cancer, 2003, Feb-01, Volume: 97, Issue:3 Suppl

    Bisphosphonates are widely used to treat skeletal complications of malignancy. These drugs accumulate in bone where they inhibit osteoclastic bone resorption and reduce the local release of factors that stimulate tumor growth. The mechanism of action of bisphosphonates is dependent on chemical structure: Nonnitrogen-containing compounds (e.g., etidronate, clodronate) are metabolized into cytotoxic analogues of ATP, whereas the more potent nitrogen-containing compounds (N-BPs; e.g., pamidronate, ibandronate, zoledronic acid) inhibit protein prenylation, thus affecting cell function and survival. Because protein prenylation is required by all cells, not just osteoclasts, the possibility arises that N-BPs could also affect the viability of tumor cells.. Several groups have investigated the in vitro effects of bisphosphonates, either alone or in combination with other antineoplastic agents, on the viability and metastatic properties of many tumor cell types. Similarly, the effect of bisphosphonate treatment on osteolysis and tumor burden has been studied in a variety of animal tumor models.. In vitro, submicromolar concentrations of N-BPs inhibited tumor cell adhesion and reduced invasion through extracellular matrix. At higher concentrations, antiproliferative and proapoptotic effects have been reported. In animal models of bone metastases, bisphosphonate treatment markedly reduced osteolytic lesions. There is also evidence of a reduction in tumor burden in bone and occasionally in other organs. Survival may be prolonged, but bisphosphonates do not appear to inhibit the growth of primary soft tissue tumors or orthotopic xenografts.. The cell culture data clearly demonstrated that N-BPs exert antitumor properties and interact synergistically with other antineoplastic agents. As bisphosphonates accumulate in bone, they can also exert cytostatic effects on tumor cells in bone metastases, either directly or indirectly via osteoclast inhibition and alterations in the bone microenvironment. Further in vivo research is now required to optimize the dosing regimen of N-BPs to exploit fully their antitumor potential.

    Topics: Apoptosis; Bone Neoplasms; Cell Division; Diphosphonates; Humans; Neoplasms; Neovascularization, Pathologic; Osteoclasts; Transforming Growth Factor beta

2003
Stress and radiation-induced activation of multiple intracellular signaling pathways.
    Radiation research, 2003, Volume: 159, Issue:3

    Exposure of cells to a variety of stresses induces compensatory activations of multiple intracellular signaling pathways. These activations can play critical roles in controlling cell survival and repopulation effects in a stress-specific and cell type-dependent manner. Some stress-induced signaling pathways are those normally activated by mitogens such as the EGFR/RAS/PI3K-MAPK pathway. Other pathways activated by stresses such as ionizing radiation include those downstream of death receptors, including pro-caspases and the transcription factor NFKB. This review will attempt to describe some of the complex network of signals induced by ionizing radiation and other cellular stresses in animal cells, with particular attention to signaling by growth factor and death receptors. This includes radiation-induced signaling via the EGFR and IGFI-R to the PI3K, MAPK, JNK, and p38 pathways as well as FAS-R and TNF-R signaling to pro-caspases and NFKB. The roles of autocrine ligands in the responses of cells and bystander cells to radiation and cellular stresses will also be discussed. Based on the data currently available, it appears that radiation can simultaneously activate multiple signaling pathways in cells. Reactive oxygen and nitrogen species may play an important role in this process by inhibiting protein tyrosine phosphatase activity. The ability of radiation to activate signaling pathways may depend on the expression of growth factor receptors, autocrine factors, RAS mutation, and PTEN expression. In other words, just because pathway X is activated by radiation in one cell type does not mean that pathway X will be activated in a different cell type. Radiation-induced signaling through growth factor receptors such as the EGFR may provide radioprotective signals through multiple downstream pathways. In some cell types, enhanced basal signaling by proto-oncogenes such as RAS may provide a radioprotective signal. In many cell types, this may be through PI3K, in others potentially by NFKB or MAPK. Receptor signaling is often dependent on autocrine factors, and synthesis of autocrine factors will have an impact on the amount of radiation-induced pathway activity. For example, cells expressing TGFalpha and HB-EGF will generate protection primarily through EGFR. Heregulin and neuregulins will generate protective signals through ERBB4/ERBB3. The impact on radiation-induced signaling of other autocrine and paracrine ligands such as TGFbeta and interleukin 6 is like

    Topics: Animals; Cell Division; Cell Nucleus; Humans; JNK Mitogen-Activated Protein Kinases; MAP Kinase Signaling System; Mitogen-Activated Protein Kinases; Models, Biological; Neoplasms; Oncogene Proteins v-erbB; p38 Mitogen-Activated Protein Kinases; Phosphatidylinositol 3-Kinases; Signal Transduction; Transforming Growth Factor beta

2003
TGF-beta signaling alterations in cancer.
    Cancer treatment and research, 2003, Volume: 115

    Topics: Animals; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2003
From TGF-beta to cancer therapy.
    Current drug targets, 2003, Volume: 4, Issue:3

    This article will introduce a novel concept in the use of TGF-beta insensitive host immune cells in cancer therapy. TGF-beta is a multi-functional cytokine. At a cellular level, it mediates cellular proliferation, growth arrest, differentiation and apoptosis. Because of the above cellular effects, TGF-beta is able to regulate a host of patho-physiological events in vivo, such as normal embryonic development, angiogenesis in tumor tissues, malignant transformation and immune surveillance. As a general rule, its direct effect on cancer cells is inhibition to cancer growth. However cancer cells are able to acquire the ability to evade this inhibitory effect of TGF-beta by becoming insensitive to TGF-beta. Furthermore, these malignant cells are able to produce large quantities of TGF-beta. The consequence of over expression of TGF-beta by cancer cells is an important factor for subsequent tumor progression. The excess amount of TGF-beta promotes tumor angiogenesis and immune suppression. The latter effect of TGF-beta is the most devastating to the host. The present discussion is focused on the role of TGF-beta insensitive immune cells in cancer growth. The host immune system offers a natural defense program against cancer. But, this natural immune surveillance is rendered ineffective by an overproduction of TGF-beta derived from the tumor cells. Rendering the host immune cells insensitive to TGF-beta in a gene therapy program offers a hope for us to successfully combat against cancer. Based on the above discussion, it is encouraging that there is a possibility for us to achieve a cure in cancer using TGF-beta insensitive immune cells in gene therapy.

    Topics: Animals; Bone Marrow Transplantation; Cell Division; Drug Resistance, Neoplasm; Humans; Immunotherapy; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2003
Inhibiting the TGF-beta signalling pathway as a means of cancer immunotherapy.
    Expert opinion on biological therapy, 2003, Volume: 3, Issue:2

    Cancers have developed numerous mechanisms for escaping the immune response, either by successfully evading a fully functional immune system or by actively suppressing the immune system so that they are no longer recognised or effectively eliminated. Current evidence supports active cancer cell-mediated immunosuppression via the secretion of the potent immunosuppressive cytokine, transforming growth factor-beta (TGF-beta), as the most general and potent mechanism for human cancer cells to escape the immune system. Efforts to bypass TGF-beta-mediated immunosuppression thereby represent an attractive therapeutic strategy for the chemoprevention and treatment of human cancers, both by directly increasing the efficacy of immunosurveillance and by increasing the efficacy of current immunotherapy strategies. Current approaches are limited by their nonspecific effects on the TGF-beta signalling pathway, as TGF-beta pathways which specifically mediate immunosuppression have not yet been defined. Future efforts should be directed towards elucidating specific TGF-beta pathways so that these can be targeted for the chemoprevention and treatment of human cancers.

    Topics: Animals; Antineoplastic Agents; Humans; Immunosuppressive Agents; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2003
Endoglin (CD105): a target for anti-angiogenetic cancer therapy.
    Current drug targets, 2003, Volume: 4, Issue:4

    Targeting of tumor vasculature is a promising strategy for cancer treatment. Among endothelial cell markers, Endoglin, a cell membrane glycoprotein, is emerging as an attractive therapeutic target on angiogenetic blood vessels, and it currently represents a powerful marker to quantify tumor angiogenesis. In normal human tissues, Endoglin is weakly expressed on erytroid precursors, stromal cells and activated monocytes, whereas it is strongly expressed on proliferating endothelial cells. In human neoplasias of different histotype, Endoglin is mainly present on endothelial cells of both peri- and intra-tumoral blood vessels, while it is weakly expressed or absent on neoplastic cells. Endoglin is an accessory component of the receptor complex of Transforming Growth Factor (TGF)-beta, a pleiotropic cytokine that modulates angiogenesis by the regulation of different cellular functions including proliferation, differentiation and migration. Interestingly, the over-expression of Endoglin antagonizes several cellular responses to TGF-beta1, while its down-regulation potentiates cellular responses to TGF-beta1. In animal models, administration of radiolabeled anti-Endoglin monoclonal antibodies (mAb) efficiently images primary tumors, and naked or conjugated anti-Endoglin mAb suppress angiogenesis and tumor growth. In this review we will summarize the complex of experimental evidences pointing to Endoglin as a vascular target to design innovative bioimmunotherapeutic strategies in human neoplasias.

    Topics: Angiogenesis Inhibitors; Animals; Antigens, CD; Biomarkers, Tumor; Endoglin; Humans; Neoplasms; Prognosis; Receptors, Cell Surface; Transforming Growth Factor beta; Vascular Cell Adhesion Molecule-1

2003
Genealogy, expression, and cellular function of transforming growth factor-beta.
    Pharmacology & therapeutics, 2003, Volume: 98, Issue:2

    The transforming growth factor-beta (TGF-beta) gene superfamily expresses a large set of structurally and functionally related polypeptides. Three TGF-beta isoforms are regulated by specific genes and have been identified in mammals (TGF-beta1, -beta2, and -beta3). All three-protein isoforms are observed abundantly during development and display overlapping and distinct spatial and temporal patterns of expressions. Each isoform plays a distinct role, the nature of which depends on the cell type, its state of differentiation, and growth conditions, and on the other growth factors present. TGF-beta regulates many of the processes common to both tissue repair and disease, including angiogenesis, chemotoxins, fibroblast proliferation and the controlled synthesis, and degradation of matrix proteins, such as collagen and fibronectin. This review will examine the genealogy and mode of actions of TGF-beta on the cell types involved in inflammation and repair, as well as in carcinoma.

    Topics: Gene Expression Regulation; Humans; Inflammation; Neoplasms; Neovascularization, Physiologic; Receptors, Transforming Growth Factor beta; Signal Transduction; Transcription, Genetic; Transforming Growth Factor beta

2003
Reversal of tumor-induced immunosuppression by TGF-beta inhibitors.
    Investigational new drugs, 2003, Volume: 21, Issue:1

    The immune system is responsible for the early detection and destruction of newly transformed malignant cells. Some transformed cells become immunologically invisible by passive avoidance of immune surveillance (i.e., when tumor cells are immunologically indistinguishable from normal cells). Other transformed cells actively secrete cytokines that effectively blind the immune system to the presence of abnormal antigens on the tumor cell surface. Transforming growth factor-beta ("TGF-beta"), which is expressed by a majority of malignant tumors, is the most potent immunosuppressor and therefore, the most likely cytokine to be responsible for the latter phenomenon. In addition to playing a key role in tumor-induced immunosuppression, TGF-beta stimulates angiogenesis. Interestingly, tumor cells eventually become refractory to TGF-beta-mediated growth arrest, either due to loss of TGF-beta receptors or due to dysregulation in TGF-beta signaling pathways. Neutralization of TGF-beta or inhibition of its production is an effective method of cancer treatment in variety of animal models. Several agents targeting TGF-beta are in the early stages of development and include anti-TGF-beta antibodies, small molecule inhibitors of TGF-beta, Smad inhibitors and antisense gene therapy. Since tumors may express more than one isoform of TGF-beta, these new drugs should target all three TGF-beta isoforms produced by human tumors. The effects of therapies targeting TGF-beta are likely to be synergistic with cytotoxic chemotherapy and immunotherapy. Reversal of TGF-beta-induced immunosuppression is a new and promising approach to cancer therapy, with potential applications in other diseases such as AIDS.

    Topics: Animals; Antibodies, Monoclonal; DNA-Binding Proteins; Humans; Immune Tolerance; Immunotherapy, Active; Neoplasm Metastasis; Neoplasms; Smad Proteins; Trans-Activators; Transforming Growth Factor beta

2003
Regulation of TGF-beta signaling and its roles in progression of tumors.
    Cancer science, 2003, Volume: 94, Issue:3

    Transforming growth factor-beta (TGF-beta) is a potent growth inhibitor of most types of cells; therefore, perturbations of TGF-beta signaling are believed to result in progression of various tumors. On the other hand, TGF-beta has been shown to act as an oncogenic cytokine through induction of extracellular matrices, angiogenesis, and immune suppression. A wide variety of effects of TGF-beta are mediated by physical interaction of signal transducer Smad proteins with various transcription factors. Among these, Runx3 plays a pivotal role in prevention of gastric cancer. TGF-beta signaling is regulated by various mechanisms in the cytoplasm and nucleus. Inhibitory Smads (I-Smads) repress TGF-beta signaling mainly by interacting with activated TGF-beta receptors. Smad ubiquitin regulatory factors (Smurfs) play important roles in facilitating the inhibitory signals induced by I-Smads. In addition, the transcriptional co-repressors c-Ski and SnoN interact with Smads, and repress transcription induced by TGF-beta. Abnormalities of these regulators of TGF-beta signaling may thus participate in the progression of various tumors.

    Topics: Animals; Disease Progression; Gastric Mucosa; Humans; Models, Biological; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2003
Targeting the TGF beta signaling network in human neoplasia.
    Cancer cell, 2003, Volume: 3, Issue:6

    Topics: Animals; Antineoplastic Agents; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2003
Role of tissue stroma in cancer cell invasion.
    The Journal of pathology, 2003, Volume: 200, Issue:4

    Maintenance of epithelial tissues needs the stroma. When the epithelium changes, the stroma inevitably follows. In cancer, changes in the stroma drive invasion and metastasis, the hallmarks of malignancy. Stromal changes at the invasion front include the appearance of myofibroblasts, cells sharing characteristics with fibroblasts and smooth muscle cells. The main precursors of myofibroblasts are fibroblasts. The transdifferentiation of fibroblasts into myofibroblasts is modulated by cancer cell-derived cytokines, such as transforming growth factor-beta (TGF-beta). TGF-beta causes cancer progression through paracrine and autocrine effects. Paracrine effects of TGF-beta implicate stimulation of angiogenesis, escape from immunosurveillance and recruitment of myofibroblasts. Autocrine effects of TGF-beta in cancer cells with a functional TGF-beta receptor complex may be caused by a convergence between TGF-beta signalling and beta-catenin or activating Ras mutations. Experimental and clinical observations indicate that myofibroblasts produce pro-invasive signals. Such signals may also be implicated in cancer pain. N-Cadherin and its soluble form act as invasion-promoters. N-Cadherin is expressed in invasive cancer cells and in host cells such as myofibroblasts, neurons, smooth muscle cells, and endothelial cells. N-Cadherin-dependent heterotypic contacts may promote matrix invasion, perineural invasion, muscular invasion, and transendothelial migration; the extracellular, the juxtamembrane and the beta-catenin binding domain of N-cadherin are implicated in positive invasion signalling pathways. A better understanding of stromal contributions to cancer progression will likely increase our awareness of the importance of the combinatorial signals that support and promote growth, dedifferentiation, invasion, and ectopic survival and eventually result in the identification of new therapeutics targeting the stroma.

    Topics: Cadherins; Epithelial Cells; Extracellular Matrix; Fibroblasts; Humans; Muscle Cells; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Pain; Receptors, Transforming Growth Factor beta; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2003
The two faces of transforming growth factor beta in carcinogenesis.
    Proceedings of the National Academy of Sciences of the United States of America, 2003, Jul-22, Volume: 100, Issue:15

    Topics: Animals; Female; Genes, Tumor Suppressor; Humans; Mammary Neoplasms, Experimental; Mice; Models, Biological; Neoplasm Metastasis; Neoplasms; Oncogenes; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2003
Id proteins in development, cell cycle and cancer.
    Trends in cell biology, 2003, Volume: 13, Issue:8

    Id proteins are important parts of signaling pathways involved in development, cell cycle and tumorigenesis. They were first shown to act as dominant negative antagonists of the basic helix-loop-helix family of transcription factors, which positively regulate differentiation in many cell lineages. The Id proteins do this by associating with the ubiquitous E proteins and preventing them from binding DNA or other transcription factors. Id proteins also associate with Ets transcription factors and the Rb family of tumor suppressor proteins, and are downstream targets of transforming growth factor beta and bone morphogenic protein signaling. Thus, the Id proteins have become important molecules for understanding basic biological processes as well as targets for potential therapeutic intervention in human disease.

    Topics: Aging; Amino Acid Motifs; Animals; Apoptosis; Bone Morphogenetic Proteins; Cell Cycle; Cell Differentiation; Gene Expression Regulation; Helix-Loop-Helix Motifs; Humans; Inhibitor of Differentiation Protein 1; Mice; Mice, Knockout; Models, Biological; Neoplasms; Neovascularization, Physiologic; Repressor Proteins; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2003
Decrypting the role of Cripto in tumorigenesis.
    The Journal of clinical investigation, 2003, Volume: 112, Issue:4

    The cell-surface associated molecule Cripto is overexpressed in a wide range of epithelial cancers, yet little is known about potential mechanisms by which Cripto expression might enhance tumorigenesis. A new study reveals that binding of Cripto to the TGF-beta ligand Activin B can block Activin B-mediated suppression of cell proliferation. Furthermore, this study also demonstrates that antibody blockade of Cripto function may prove useful in the inhibition of tumorigenesis.

    Topics: Activins; Amino Acid Motifs; Animals; Cell Division; Epidermal Growth Factor; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Models, Biological; Neoplasm Proteins; Neoplasms; Nodal Protein; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
TGFBR1*6A and cancer risk: a meta-analysis of seven case-control studies.
    Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 2003, Sep-01, Volume: 21, Issue:17

    TGFBR1*6A is a hypomorphic polymorphic allele of the type I transforming growth factor beta receptor (TGFBR1). TGFBR1*6A is a candidate tumor susceptibility allele that has been associated with an increased incidence of various types of cancer. This study was undertaken to analyze all published case-control studies on TGFBR1*6A and cancer and determine whether TGFBR1*6A is associated with cancer.. All published case-control studies assessing the germline frequency of TGFBR1*6A were included. Studies assessing TGFBR1*6A in tumors were excluded. The results of seven studies comprising 2,438 cases and 1,846 controls were pooled and analyzed.. Overall, TGFBR1*6A carriers have a 26% increased risk of cancer (odds ratio [OR], 1.26; 95% confidence interval [CI], 1.07 to 1.49). Cancer risk for TGFBR1*6A homozygotes (OR, 2.53; 95% CI, 1.39 to 4.61) is twice that of TGFBR1*6A heterozygotes (OR, 1.26; 95% CI, 1.04 to 1.51). Analysis of various types of tumors shows that TGFBR1*6A carriers are at increased risk of developing breast cancer (OR, 1.48; 95% CI, 1.11 to 1.96), hematological malignancies (OR, 1.70; 95% CI, 1.13 to 2.54), and ovarian cancer (OR, 1.53; 95% CI, 1.07 to 2.17). Carriers of TGFBR1*6A who are from the United States are at increased risk of colorectal cancer (OR, 1.38; 95% CI, 1.02 to 1.86). However, Southern European TGFBR1*6A carriers have no increased colorectal cancer risk. There is no association between TGFBR1*6A and bladder cancer.. TGFBR1*6A is emerging as a highfrequency, low-penetrance tumor susceptibility allele that predisposes to the development of breast, ovarian, and colorectal cancer, as well as hematologic malignancies.

    Topics: Adolescent; Adult; Aged; Alleles; Case-Control Studies; Female; Genetic Predisposition to Disease; Genotype; Heterozygote; Homozygote; Humans; Male; Middle Aged; Neoplasms; Odds Ratio; Polymorphism, Genetic; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

2003
From developmental disorder to heritable cancer: it's all in the BMP/TGF-beta family.
    Nature reviews. Genetics, 2003, Volume: 4, Issue:10

    Transforming growth factor-beta (TGF-beta) regulates many cellular processes through complex signal-transduction pathways that have crucial roles in normal development. Disruption of these pathways can lead to a range of diseases, including cancer. Mutations in the genes that encode members of the TGF-beta pathway are involved in vascular diseases as well as gastrointestinal neoplasia. More recently, they have been implicated in Cowden syndrome, which is normally associated with mutations in the phosphatase and tensin homologue gene PTEN. Molecular studies of TGF-beta signalling are now showing why mutations in genes that encode components of this pathway result in inherited cancer and developmental diseases.

    Topics: Animals; Bone Diseases, Developmental; Bone Morphogenetic Proteins; Humans; Mutation; Neoplasms; Transforming Growth Factor beta

2003
Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer.
    Nature reviews. Cancer, 2003, Volume: 3, Issue:11

    Topics: Apoptosis; Disease Progression; Homeostasis; Humans; Immunosuppression Therapy; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2003
On and off: proteasome and TGF-beta signaling.
    Experimental cell research, 2003, Dec-10, Volume: 291, Issue:2

    Transforming growth factor-beta (TGF-beta) signaling mainly relies on the TGF-beta receptor-Smad pathway. Meanwhile, TGF-beta binding to its receptors initiates the degradation of several key components of its signaling pathway. The degradation of these components, including both positive and negative transducers, is mediated by the ubiquitin-proteasome system. Inhibition of the proteasome activity causes accumulation of these components in the cells and modulates TGF-beta signaling in a time-dependent and gene-specific manner. The accelerated degradation of TGF-beta signaling components via the proteasome system has been found in a number of tumors, indicating that dysregulated proteasomal degradation is a novel pathway how tumor cells silence TGF-beta signaling.

    Topics: Animals; Cysteine Endopeptidases; DNA-Binding Proteins; Humans; Multienzyme Complexes; Neoplasms; Proteasome Endopeptidase Complex; Signal Transduction; Smad Proteins; Trans-Activators; Transforming Growth Factor beta; Ubiquitin

2003
The molecular basis of retinoid action in tumors.
    Trends in molecular medicine, 2003, Volume: 9, Issue:12

    Topics: Animals; Apoptosis; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Humans; Medulloblastoma; Models, Biological; Neoplasms; Retinoids; Transforming Growth Factor beta

2003
[Transcriptional regulation by the transforming growth factor-beta signaling].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 2003, Volume: 48, Issue:16

    Topics: Animals; Cell Division; DNA-Binding Proteins; Humans; Hypertension, Pulmonary; Neoplasms; Signal Transduction; Smad Proteins; Trans-Activators; Transcription, Genetic; Transforming Growth Factor beta

2003
Cellular and molecular mechanisms of carcinogenesis.
    Gastroenterology clinics of North America, 2002, Volume: 31, Issue:2

    Our understanding of the cellular and molecular mechanisms of cancer of the gastrointestinal tract has increased dramatically over the last several decades. We are identifying new players in the pathways toward cancer with increasing frequency. In addition, we have come to understand that no single pathway acts by itself; in vivo, the effects are combinatorial. As new and better cell culture and animal models of carcinogenesis arise, our knowledge will continue to grow. As we learn more, we will be able to translate the results of our research into new and better techniques for the diagnosis and treatment of gastrointestinal cancers.

    Topics: Apoptosis; Base Pair Mismatch; Cell Cycle; Cell Transformation, Neoplastic; DNA Methylation; DNA Repair; DNA-Binding Proteins; Genes, p53; Genes, ras; Histones; Humans; Neoplasms; Proto-Oncogene Proteins; Signal Transduction; Transforming Growth Factor beta

2002
Role of genetic polymorphisms in tumour angiogenesis.
    British journal of cancer, 2002, Nov-04, Volume: 87, Issue:10

    Angiogenesis plays a crucial role in the development, growth and spread of solid tumours. Pro- and anti-angiogenic factors are abnormally expressed in tumours, influencing tumour angiogenesis, growth and progression. Polymorphisms in genes encoding angiogenic factors or their receptors may alter protein expression and/or activity. This article reviews the literature to determine the possible role of angiogenesis-related polymorphisms in cancer. Further research studies in this potentially crucial area of tumour biology are proposed.

    Topics: Animals; Cytokines; Endothelial Growth Factors; Hepatocyte Growth Factor; Humans; Intercellular Signaling Peptides and Proteins; Lymphokines; Matrix Metalloproteinases; Neoplasms; Neovascularization, Pathologic; Polymorphism, Genetic; Receptors, Fibroblast Growth Factor; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2002
The tumor microenvironment: a potential arbitrator of the tumor suppressive and promoting actions of TGFbeta.
    Differentiation; research in biological diversity, 2002, Volume: 70, Issue:9-10

    Transforming growth factor beta (TGFbeta) members are secreted in biologically inactive complexes that must be activated in order to enable binding to their cell surface receptors. Interestingly, many of the proteins that can activate TGFbeta have been implicated in either suppressing or promoting tumorigenesis. Included among these are matrix proteins (thrombospondin-1), receptors (integrins alphanubeta6 and alphanubeta8) and proteases (matrix metalloproteases and plasmin). These proteins cannot only activate TGFbeta, but can also modulate cell responsiveness to TGFbeta. In this section, we review data highlighting the complexity and bidirectionality of TGFbeta matrix interactions within the tumor microenvironment, and propose that these dynamic interactions are a critical spatial and temporal determinant of the effects of TGFbeta on tumorigenesis.

    Topics: Animals; Cell Movement; Endopeptidases; Extracellular Matrix; Humans; Mice; Neoplasms; Neoplastic Processes; Neovascularization, Pathologic; Receptors, Transforming Growth Factor beta; Signal Transduction; Thrombospondins; Transforming Growth Factor beta

2002
Inhibitors of apoptosis proteins in injury and disease.
    IUBMB life, 2002, Volume: 54, Issue:5

    The inhibitor of apoptosis (IAP) gene family prevents cell death by binding to and inhibiting caspases. In addition to negatively regulating apoptosis, IAPs bind to signaling intermediates and receptors, and participate in diverse cellular functions. Here, we describe the physiological functions of IAPs and their participation in injury and disease processes.

    Topics: Animals; Apoptosis; Disease Models, Animal; Humans; Inhibitor of Apoptosis Proteins; Microtubule-Associated Proteins; Models, Biological; Multigene Family; Neoplasm Proteins; Neoplasms; Nerve Tissue Proteins; Neuronal Apoptosis-Inhibitory Protein; Neurons; Protein Biosynthesis; Protein Structure, Tertiary; Proteins; Signal Transduction; Survivin; Transcription, Genetic; Transforming Growth Factor beta; X-Linked Inhibitor of Apoptosis Protein

2002
TGFbeta-regulated transcriptional mechanisms in cancer.
    International journal of gastrointestinal cancer, 2002, Volume: 31, Issue:1-3

    Transforming growth factor-beta (TGFbeta) has been implicated in oncogenesis for many years. The multifunctional activities of TGFbeta endow it with both tumor suppressor and tumor promoting activities, depending on the stage of carcinogenesis and the responsivity of the tumor cell. In early tumor stages, TGFbeta inhibits epithelial cell growth through induction of apoptosis and cell cycle arrest. During tumor development, however, many tumor cells lose their growth-inhibitory responses to TGFbeta owing to genetic alterations or signaling perturbations such as oncogenic Ras signaling. Loss of TGFbeta-growth inhibition is commonly associated with increased tumor cell invasion and metastasis of tumor cells that undergo an epithelial-mesenchymal transition. Interestingly, the tumor-promoting effects of TGFbeta on the tumor cells are observed particularly in cells in which TGFbeta-signaling remains functional despite loss of growth control by TGFbeta. New insights into transcriptional mechanisms activated by TGFbeta are providing a better understanding of the cellular changes involved in the switch of TGFbeta from a tumor suppressor to a tumor promotor.

    Topics: Cell Differentiation; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Signal Transduction; Transcription, Genetic; Transforming Growth Factor beta

2002
[Epstein-Barr virus and oncogenesis].
    Uirusu, 2002, Volume: 52, Issue:2

    Topics: Apoptosis; B-Lymphocytes; Cell Division; Epstein-Barr Virus Infections; Epstein-Barr Virus Nuclear Antigens; Genes, Viral; Herpesvirus 4, Human; Humans; Neoplasms; Ribosomal Proteins; RNA-Binding Proteins; Signal Transduction; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Transforming Growth Factor beta1; Viral Matrix Proteins; Virus Replication

2002
The TGF-beta family: signaling pathways, developmental roles, and tumor suppressor activities.
    TheScientificWorldJournal, 2002, Apr-05, Volume: 2

    Intercellular communication is a critical process for all multicellular organisms, and communication among cells is required for proper embryonic development and adult physiology. Members of the Transforming Growth Factor-beta (TGF-beta) family of secreted proteins communicate information between cells via a complex signaling pathway, and family members are capable of inducing a wide range of cellular responses. The purpose of this review is to provide the reader with a broad introduction to our current understanding of three aspects of the TGF-beta family. These are the molecular mechanisms utilized by TGF-beta signaling pathways, the developmental roles played by TGF-beta family members in a variety of species, and the growing list of cancers in which various TGF-beta signaling pathways display tumor suppressor activity.

    Topics: Animals; Gene Expression Regulation; Humans; Mice; Morphogenesis; Mutation; Neoplasms; Nuclear Proteins; Protein Processing, Post-Translational; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2002
TGF-beta signaling: positive and negative effects on tumorigenesis.
    Current opinion in genetics & development, 2002, Volume: 12, Issue:1

    TGF-beta binding to the cell surface triggers activation of multiple signal transduction pathways that are connected in intricate ways with each other, and with other response networks involved in sensing cellular information input. Recent data indicate that changes in signal intensity and connectivity of these pathways may underlie the complex transition of the TGF-beta pathway from tumor suppressor to oncogene during tumorigenesis.

    Topics: Animals; DNA-Binding Proteins; Gene Dosage; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Humans; Neoplasms; Proto-Oncogene Proteins; Receptors, Transforming Growth Factor beta; Signal Transduction; Transcription, Genetic; Transcriptional Activation; Transforming Growth Factor beta

2002
Strike against cancer.
    Trends in molecular medicine, 2002, Volume: 8, Issue:1

    Topics: Animals; CD4-Positive T-Lymphocytes; Humans; Mice; Mice, Transgenic; Neoplasms; Signal Transduction; T-Lymphocytes, Helper-Inducer; Transforming Growth Factor beta

2002
The TGF-beta--Smad network: introducing bioinformatic tools.
    Trends in genetics : TIG, 2002, Volume: 18, Issue:2

    The TGF-beta superfamily is an important class of intercellular signalling molecule, including TGF-beta and bone morphogenetic proteins. Intracellular signalling cascades triggered by these molecules eventually activate transcription factors of the Smad family, which then regulate expression of their respective target genes. This article will discuss the TGF-beta--Smad signalling networks and how these processes are represented in databases of signal transduction and transcription control mechanisms. These databases can provide a well-structured overview of the subject and a basis for advanced bioinformatics analyses to interpret the function of genomic sequences or to analyse signalling networks.

    Topics: Animals; Binding Sites; Databases, Factual; DNA; DNA-Binding Proteins; Feedback, Physiological; Humans; Mutation; Neoplasms; Protein Conformation; Signal Transduction; Smad Proteins; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2002
[Molecular mechanisms of oncogenic transformation: what's new?].
    Bulletin du cancer, 2002, Volume: 89, Issue:1

    During the past two years, new molecular targets have been discovered which link cell cycle, cell proliferation and cellular growth. It has become more and more evident that whereas gain-of-function mutations in specific genes can lead to cancer, genomic instability plays also an important role in tumour progression. With examples taken from the recent literature, we describe in this short review crucial findings on the molecular mechanisms controlling cell cycle and proliferation. We illustrate how specific combinations of proto-oncogenes alterations can result in tissue-specific tumours. Finally, impairment of the interactions of a cancer cell with its surrounding neighbours is also shown to participate in the progression toward aggressive phenotypes.

    Topics: Animals; Antineoplastic Agents, Phytogenic; CDC2-CDC28 Kinases; Cell Cycle; Cell Division; Cell Transformation, Neoplastic; Cyclin D1; Cyclin E; Cyclin-Dependent Kinase 2; Cyclin-Dependent Kinase Inhibitor p16; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinases; Cyclins; Disease Progression; Drug Resistance; Fusion Proteins, bcr-abl; Humans; Mice; Mutation; Neoplasm Metastasis; Neoplasm Proteins; Neoplasms; Neovascularization, Pathologic; Neural Cell Adhesion Molecules; Paclitaxel; Protein Serine-Threonine Kinases; Radiation Tolerance; Rats; Thrombospondins; Transforming Growth Factor beta; Tuberous Sclerosis

2002
Transforming growth factor-beta in T-cell biology.
    Nature reviews. Immunology, 2002, Volume: 2, Issue:1

    Strict control of T-cell homeostasis is required to permit normal immune responses and prevent undesirable self-targeted responses. Transforming growth factor-beta (TGF-beta) has been shown to have an essential role in that regulation. Owing to its broad expression, and inhibitory effects on multiple cell types of the immune system, TGF-beta regulation is complex. Through advances in cell-specific targeting of TGF-beta signalling in vivo, the role of TGF-beta in T-cell regulation has become clearer. Recent in vitro studies provide a better understanding of how TGF-beta regulates T-cell homeostasis, through multiple mechanisms involving numerous cell types.

    Topics: Animals; Antigen-Presenting Cells; Apoptosis; Autoantigens; Autoimmunity; Cell Differentiation; Homeostasis; Humans; Lymphocyte Activation; Models, Immunological; Neoplasms; Signal Transduction; T-Lymphocytes; Transforming Growth Factor beta

2002
Human placental trophoblast as an in vitro model for tumor progression.
    Canadian journal of physiology and pharmacology, 2002, Volume: 80, Issue:2

    The human placenta is a highly invasive tumor-like structure in which a subpopulation of placental trophoblast cells known as the "extravillous trophoblast" (EVT) invades the uterine decidua and its vasculature to establish adequate fetal-maternal exchange of molecules. By utilizing in vitro-propagated short-lived EVT cell lines we found that molecular mechanisms responsible for their invasiveness are identical to those of cancer cells; however, unlike cancer cells, their proliferation, migration, and invasiveness in situ are stringently controlled by decidua-derived transforming growth factor (TGF)-beta. By SV40T antigen transfection of normal EVT cells followed by a forced crisis regimen in culture we produced an immortalized premalignant derivative that is hyperproliferative, hyperinvasive, and deficient in gap-junctional intercellular communication. Both premalignant and malignant EVT (JAR and JEG-3 choriocarcinoma) cell lines were found to be TGF-beta-resistant. Using these cell lines, we investigated genetic changes responsible for transition of the normal EVT cells to premalignant and malignant phenotype. Hyperinvasiveness in both cases resulted from a downregulation of tissue inhibitor of metalloprotease (TIMP)-1 and plasminogen activator inhibitor (PAI)-1 genes. In contrast to normal EVT cells, both cell types failed to upregulate these genes in response to TGF-beta. Loss of TGF-beta response in malignant EVT cells was explained by the loss of expression of Smad3 gene. Differential mRNA display of normal and premalignant EVT cells identified up- and down-regulation of numerous known or novel genes in premalignant EVT cells, with potential oncogenic and (or) tumor-suppressor functions, e.g., loss of fibronectin and insulin-like growth factor binding protein (IGFBP-5). Premalignant EVT cells also lost IGF receptor type 2 (IGFR-II). IGFBP-5 was shown to be a negative regulator of IGF-1-induced proliferation of premalignant EVT cells, so that loss of IGFBP-5 as well as IGFR-II permitted their unrestricted proliferation in an IGF-I-rich microenvironment of the fetal-maternal interface. The present model may be a good prototype for identifying genetic changes underlying epithelial tumor progression.

    Topics: Cell Line; Choriocarcinoma; Disease Progression; Female; Gene Expression; Humans; Neoplasm Invasiveness; Neoplasms; Placentation; Precancerous Conditions; Pregnancy; Signal Transduction; Transforming Growth Factor beta; Trophoblasts; Tumor Cells, Cultured

2002
Transforming growth factor beta signal transduction.
    Journal of leukocyte biology, 2002, Volume: 71, Issue:5

    Transforming growth factor beta1 (TGF-beta1) is the prototypic member of a large family of structurally related pleiotropic-secreted cytokines that play a pivotal role in the control of differentiation, proliferation, and state of activation of many different cell types including immune cells. TGF-beta family members have potent immunosuppressor activities in vitro and in vivo. These cytokines trigger their biological effects by inducing the formation of a heteromeric transmembrane serine/threonine kinase receptor complex. These receptors then initiate intracellular signaling through activation of Smad proteins, and specific Smads become phosphorylated and associate with other Smads. These heteromeric Smad complexes accumulate in the nucleus, where they modulate the expression of target genes. Recent data support the notion that Smads are important intracellular effectors of TGF-beta in immune cells. Here, we review recent advances in TGF-beta signal transduction in immune cells.

    Topics: Animals; Apoptosis; Cell Division; Cell Membrane; DNA-Binding Proteins; Humans; Immune System; Models, Biological; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Trans-Activators; Transcriptional Activation; Transforming Growth Factor beta

2002
Mechanisms of TGF-beta signaling in regulation of cell growth and differentiation.
    Immunology letters, 2002, Jun-03, Volume: 82, Issue:1-2

    Transforming growth factor beta (TGF-beta) is a secreted protein that regulates proliferation, differentiation and death of various cell types. All immune cell lineages, including B, T and dendritic cells as well as macrophages, secrete TGF-beta, which negatively regulates their proliferation, differentiation and activation by other cytokines. Thus, TGF-beta is a potent immunosuppressor and perturbation of TGF-beta signaling is linked to autoimmunity, inflammation and cancer. Regulation of cell proliferation and differentiation by TGF-beta is a topic of great basic and clinical importance. We summarize our work on the growth inhibitory pathway downstream of TGF-beta, which is triggered by receptor serine/threonine kinases at the cell surface and downstream effectors of the Smad family. Activated Smads regulate transcription of target genes, including cell cycle inhibitors such as p21, which mediate the anti-proliferative response and partially explain the tumor suppressive action of the TGF-beta pathway. We have described a molecular mechanism of regulation of the p21 gene by Smads and transcription factor Sp1. At late stages of tumor progression, TGF-beta promotes tumorigenesis via suppression of the immune system and changes in cell differentiation of epithelial tumor cells, a phenomenon termed epithelial to mesenchymal transdifferentiation (EMT). We review our work on the role of the Smad pathway in controlling EMT. In conclusion, the molecular pathways that describe the anti-proliferative and transdifferentiating effects of TGF-beta in epithelial cells have been uncovered to great molecular detail; a future challenge will be to test their generality in other systems, including the immune system.

    Topics: Cell Differentiation; Cell Division; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Epithelial Cells; Immune System; Mesoderm; Models, Biological; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2002
The transforming growth factor-beta signaling pathway in tumorigenesis.
    Current opinion in oncology, 2001, Volume: 13, Issue:1

    Transforming growth factor-beta is believed to play a dual role in carcinogenesis. Through its ability to inhibit cellular proliferation it suppresses tumor development in its early stages, but in the course of tumor progression malignant cells often acquire resistance to growth inhibition by transforming growth factor-beta and themselves secrete large amounts of this cytokine. Transforming growth factor-beta furthers malignant progression in two ways: for one, it acts on nontransformed cells present in the tumor mass to suppress antitumor immune responses and to augment angiogenesis. Secondly, it promotes invasion and the formation of metastases in a cell-autonomous manner that requires transforming growth factor-beta signaling activity, albeit at reduced levels, to be present in the tumor cells themselves.

    Topics: Cell Transformation, Neoplastic; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta

2001
Role of transforming growth factor beta in cancer.
    Journal of cellular physiology, 2001, Volume: 186, Issue:2

    Transforming growth factor beta (TGF-beta) is an effective and ubiquitous mediator of cell growth. The significance of this cytokine in cancer susceptibility, cancer development and progression has become apparent over the past few years. TGF-beta plays various roles in the process of malignant progression. It is a potent inhibitor of normal stromal, hematopoietic, and epithelial cell growth. However, at some point during cancer development the majority of transformed cells become either partly or completely resistant to TGF-beta growth inhibition. There is growing evidence that in the later stages of cancer development TGF-beta is actively secreted by tumor cells and not merely acts as a bystander but rather contributes to cell growth, invasion, and metastasis and decreases host-tumor immune responses. Subtle alteration of TGF-beta signaling may also contribute to the development of cancer. These various effects are tissue and tumor dependent. Identifying and understanding TGF-beta signaling pathway abnormalities in various malignancies is a promising avenue of study that may yield new modalities to both prevent and treat cancer. The nature, prevalence, and significance of TGF-beta signaling pathway alterations in various forms of human cancer as well as potential preventive and therapeutic interventions are discussed in this review.

    Topics: Animals; Cell Division; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2001
Transforming growth factor-beta signaling in cancer.
    Microscopy research and technique, 2001, Feb-15, Volume: 52, Issue:4

    Transforming growth factor (TGF-beta) is a multifunctional polypeptide implicated in the regulation of a variety of cellular processes including growth, differentiation, apoptosis, adhesion, and motility. Abnormal activation or inhibition of these TGF-beta regulated processes is implicated in many diseases, including cancer. Cancers can develop through selective exploitation of defects in TGF-beta signaling that occur at several different levels in the pathway. The TGF-beta signal transduction cascade is initiated when TGF-beta binds to transmembrane receptors. The TGF-beta receptors then phosphorylate and activate Smad proteins, which transduce the signal from the cytoplasm to the nucleus. In the nucleus, Smads can bind directly to DNA and cooperate with other transcription factors to induce transcription of TGF-beta target genes. Mutations in target genes, Smads, or the TGF-beta receptor are associated with certain human cancers.

    Topics: DNA-Binding Proteins; Humans; Mutation; Neoplasms; Proteins; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2001
Role of TGF-beta in immune-evasion of cancer.
    Microscopy research and technique, 2001, Feb-15, Volume: 52, Issue:4

    One of the major obstacles in tumor-immunology is the outgrowth of malignant tumors despite their immunogenicity and recognition by the immune-system. Multiple mechanisms for this phenomenon have been proposed. We review the possible involvement of transforming growth factor beta (TGF-beta) in this context. TGF-beta is a cytokine with pleiotropic functions, involved in multiple physiologic processes including immunoregulation. Immune elimination of most cancers ultimately depends on cytolytic T cells (CTL). We propose a mechanism of specific suppression of cytolytic T cell (CTL)-responses mediated through immunoglobulin-bound TGF-beta (IgG-TGF-beta), secreted by activated B cells, and a cell of myeloid origin. This mononuclear "Veto" cell presumably binds IgG-TGF-beta through Fc-receptors and activates latent TGF-beta. The suggestion that B cell responses can inhibit tumor rejection is supported by observations in B cell-deficient mice. Ways for enhancing effective cancer immunity by interfering with the network of interactions involving IgG-TGF-beta are discussed.

    Topics: Animals; B-Lymphocytes; Mice; Neoplasms; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Tumor Escape

2001
Clinical implications of circulating angiogenic factors in cancer patients.
    Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 2001, Feb-15, Volume: 19, Issue:4

    Angiogenesis, a process fundamental to tumor growth, is regulated by angiogenic factors. This article reviews prognostic and other clinical implications of circulating angiogenic factors in cancer patients.. A MEDLINE search of literature was performed using the names of various angiogenic factors as the key words. Studies pertaining to circulating angiogenic factors in cancer patients were reviewed. Pertinent literature regarding tumor expression of common angiogenic factors and their prognostic relevance in human cancers were also examined.. A substantial number of studies have demonstrated a strong association between elevated tumor expression of vascular endothelial growth factor (VEGF) and advanced disease or poor prognosis in various cancers. This supports the pivotal role of VEGF in regulating tumor angiogenesis. More recently, there is mounting evidence that the level of circulating VEGF in patients with different types of cancer may be predictive of tumor status and prognosis. Preliminary data also suggest that circulating VEGF may be useful in predicting and monitoring tumor response to anticancer therapies and in follow-up surveillance for tumor relapse. There are reports supporting the prognostic value of other circulating angiogenic factors such as basic fibroblast growth factor, platelet-derived endothelial cell growth factor, transforming growth factor-beta, and angiogenin, but their clinical significance is less conclusive because of limited data.. Circulating VEGF seems to be a reliable surrogate marker of angiogenic activity and tumor progression in cancer patients. Evaluation of circulating angiogenic factors is a promising novel approach of prognostication in cancer patients that has the advantages of being convenient and noninvasive, and it may provide new prognostic information that is not afforded by conventional clinicopathologic prognostic indicators.

    Topics: Angiogenesis Inducing Agents; Endothelial Growth Factors; Humans; Lymphokines; Neoplasms; Neovascularization, Pathologic; Prognosis; Thymidine Phosphorylase; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2001
[Molecular mechanism of inactivation of TGF-beta receptors during carcinogenesis].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 2001, Volume: 46, Issue:2

    Topics: Activin Receptors, Type I; Gene Deletion; Humans; Neoplasms; Oncogene Proteins, Fusion; Protein Serine-Threonine Kinases; Proto-Oncogene Protein c-fli-1; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA-Binding Protein EWS; Signal Transduction; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta

2001
Mechanisms of inhibin signal transduction.
    Recent progress in hormone research, 2001, Volume: 56

    Inhibin was first identified as a gonadal hormone that potently inhibits pituitary follicle-stimulating hormone (FSH) synthesis and secretion. Although the notion of a nonsteroidal, gonadally derived inhibitory substance was realized in the early 1930s (McCullagh, 1932), identification of the hormone was not accomplished until more than 50 years later. At that time, inhibin was purified from bovine and porcine follicular fluid and was shown to be produced in two forms through dimeric assembly of an alpha subunit (18 kDa) and one of two closely related beta subunits (betaA and betaB, approximately 14 kDa) (Ling et al., 1985; Miyamoto et al., 1985; Rivier et al., 1985; Robertson et al., 1985). Dimers of alpha and betaA and alpha and betaB subunits form inhibin A and inhibin B, respectively. In the process of purifying inhibin, two groups also identified homo- and heterodimers of the inhibin beta subunits (Ling et al., 1986; Vale et al., 1986). These hormones, the activins, were shown to potently stimulate FSH secretion from primary pituitary cultures and are now known to play important roles in growth and development (Woodruff, 1998; Pangas and Woodruff, 2000). Inhibins and activins are considered members of the transforming growth factor-beta (TGF-beta) superfamily of growth and differentiation factors, based on a pattern of conserved cysteine residues in the alpha and beta subunits, similar to other ligands in the family. Identification of the subunit proteins led to the cloning of their cDNAs and subsequently to their chromosomal mapping in several species (Mason et al, 1985,1986; Forage et al., 1986; Mayo et al., 1986; Esch et al., 1987; Woodruff et al., 1987; Barton et al., 1989; Hiendleder et al., 2000). Three additional activin-related beta subunits (betaC and betaE in mammals and betaD in Xenopus laevis) also have been identified but do not appear to play a role in FSH regulation (Hotten et al., 1995; Oda et al., 1995; Fang et al., 1996, 1997; Loveland et al., 1996; Schmitt el al., 1996; O'Bryan et al., 2000; Lau et al., 2000). To date, only one alpha subunit has been reported. The inhibin subunits are expressed in various tissues (Meunier et al., 1988a, 1988b) but the gonads are clearly the primary source of circulating inhibins (Woodruff et al., 1996). While inhibins act in a paracrine role in some tissues (Hsueh et al., 1987), their best-understood roles are as endocrine regulators of pituitary FSH. Activins also were purified from follicular flu

    Topics: Activin Receptors; Activins; Animals; DNA, Complementary; Female; Follicle Stimulating Hormone; Gene Expression Regulation; Humans; Inhibins; Ligands; Models, Biological; Neoplasms; Prostatic Secretory Proteins; Protein Binding; Protein Isoforms; Receptors, Peptide; Reproduction; Signal Transduction; Transforming Growth Factor beta

2001
TGFbeta signaling and cancer.
    Experimental cell research, 2001, Mar-10, Volume: 264, Issue:1

    Topics: Animals; Cell Division; DNA-Binding Proteins; Humans; Mice; Models, Biological; Mutation; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2001
Synthesis and degradation of basement membranes and extracellular matrix and their regulation by TGF-beta in invasive carcinomas (Review).
    International journal of oncology, 2001, Volume: 18, Issue:4

    The proper structure of the extracellular matrix, in particular of the basement membrane and the adjacent interstitial matrix, are essential prerequisites for a proper function of tissues. Invasive growth in malignant tumors is associated with a destruction of various matrix structures. Due to extensive recent analyses significant advances have been made in the knowledge of the structure of the extracellular matrix, the composition of its most important constituents, their metabolism and that of matrix degrading enzymes. This information provides insight into the pathophysiology of malignant growth. Thereby, it has been shown that malignant tumor growth is associated with a loss of basement membrane (BM) material which, however, disappears not homogeneously, but affects various BM components to different degree. The loss of an intact BM as the first barrier is therefore the initial step of tumor invasion. Despite this loss there is evidence that the de novo synthesis of BM constituents in tumor and adjacent stromal cells is enhanced. Thus, it is obvious that BM material is degraded during the invasion process to significant degree. In addition, since there is a positive correlation between the amount of retained peritumoral BM and a higher degree of tumor cell differentiation the amount of retained BM material seems to represent a marker for the biological behaviour of the tumor cells. The loss of BM material is well explained by a significant expression of major matrix degrading enzymes, the matrix metalloproteinases (MMPs) both on the mRNA and protein level. Here again, there is considerable data indicating that both tumor and stroma cells are involved in the MMP synthesis. In addition to the loss of BM substances, the interstitial extracellular matrix (ECM) is disarranged. This disarrangement may comprise enhanced de novo synthesis ("desmoplasia") or dissolution by distinct MMPs (collagenases, such as MMP-1) reflecting obviously different reaction statuses of the stromal cells. Finally, significant work has been done on the elucidation of the role of regulating cytokine systems. To this regard, particular attention has been paid to the TGF-beta system and it has been shown that the major three isoforms of TGF-betas are upregulated both in tumor and stroma cells. Since the TGF-beta-effect is mainly mediated by a particular signalling system via the TGF-beta-receptors (TBRs), the investigation of this system has provided considerable insight into the rol

    Topics: Basement Membrane; Collagen; Extracellular Matrix; Gene Expression Regulation; Humans; Matrix Metalloproteinases; Neoplasm Invasiveness; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2001
The smad proteins and TGFbeta signalling: uncovering a pathway critical in cancer.
    Pathology, 2001, Volume: 33, Issue:1

    The critical role of TGFbeta in development and growth control is well established but the signalling pathway has only recently been elucidated. Identification of the Smads as TGFbeta's intracellular signalling mediators has led to an explosion of information on a novel signalling network that links the cell surface to the nucleus. Many cancers develop resistance to the growth-inhibitory effects of TGFbeta and mutations in signalling pathway components have been discovered that may underly tumour progression.

    Topics: DNA-Binding Proteins; Humans; Models, Biological; Neoplasms; Signal Transduction; Smad2 Protein; Smad3 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2001
The role of TGFbeta in human cancers.
    Pathology, 2001, Volume: 33, Issue:1

    Transforming growth factor beta (TGFbeta) is secreted as a large latent precursor from both normal and transformed cells which needs to be activated for biological activity. The active TGFbeta binds either directly to TbetaR-II or indirectly by binding to beta-glycan which then presents the TGFbeta to TbetaR-II. Formation of the TGFbeta-TbetaR-II complex rapidly leads to phosphorylation of TbetaR-I. TbetaR-I, in turn, phosphorylates receptor-specific Smads and induces their translocation into the nucleus. TGFbeta is able to act as a growth stimulator or inhibitor and elicits a broad spectrum of biological effects on various cell types. However, these cells may lose their sensitivity and responsiveness to TGFbeta. Down-regulation or loss of functional receptors, aberrant signal transduction pathways due to Smad mutations, loss of the cell's ability to activate latent TGFbeta, loss of the peptide itself or functional genes that control the transcription and translation of TGFbeta may contribute to development of cancer.

    Topics: DNA-Binding Proteins; Down-Regulation; Female; Humans; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad2 Protein; Smad3 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2001
Endoglin: An accessory component of the TGF-beta-binding receptor-complex with diagnostic, prognostic, and bioimmunotherapeutic potential in human malignancies.
    Journal of cellular physiology, 2001, Volume: 188, Issue:1

    Endoglin (CD105) is a cell membrane glycoprotein over-expressed on highly proliferating endothelial cells in culture, and on endothelial cells of angiogenetic blood vessels within benign and malignant tissues. CD105 binds several factors of the Transforming Growth Factor (TGF)-beta superfamily, and its over-expression modulates cellular responses to TGF-beta1. The complex of experimental findings accumulated in the last few years strongly indicate that CD105 is a powerful marker of angiogenesis, and that it might play a critical role in the pathogenesis of vascular diseases and in tumor progression. In this paper, we will review the structural, biological and functional features of CD105, as well as its distribution within normal and neoplastic tissues, emphasizing its foreseeable role as a molecular target for new diagnostic and bioimmunotherapeutic approaches in human malignancies.

    Topics: Animals; Antigens, CD; Biomarkers, Tumor; Endoglin; Endothelium, Vascular; Humans; Macromolecular Substances; Neoplasms; Neovascularization, Pathologic; Neovascularization, Physiologic; Receptors, Cell Surface; Transforming Growth Factor beta; Vascular Cell Adhesion Molecule-1

2001
Negative regulation of Janus kinases.
    Cell biochemistry and biophysics, 2001, Volume: 34, Issue:1

    The precise regulation of both the magnitude and the duration of Janus kinase (JAK) catalytic activity is essential for the cytokine orchestration of many biological processes, and the dysregulation of JAK activity has pathological implications. Immunosuppressive disease states, such as X-linked severe combined immunodeficiency, arise from inappropriate JAK inhibition. In contrast, a limited number of cancers, primarily leukemias, result from constitutive or enhanced activation of JAK activity. JAKs are no longer implicated only in classic cytokine receptor-mediated signaling pathways, but are now also known to integrate indirectly into other receptor-mediated signal transduction processes. Therefore, an increasing number of therapeutic applications exist for biological-response modifiers that can restore aberrant JAK activity to normal levels. Exciting breakthroughs in both physiological and pharmacological methods of selective inhibition of cytokine-JAK-signal transducers and activators of transcription pathways have recently emerged in the form of suppressors of cytokine signaling (also known as cytokine-inducible SH2 protein, JAK-binding protein, or STAT-induced STAT inhibitor) proteins and novel dimethoxyquinazoline derivatives, respectively. The basis of these and other mechanisms of negative regulation of JAK activity, including the suppression of jak expression levels caused by tumor- or pathogen-derived agents, the complex interactions of JAKs with phosphatases, and the redox regulation of JAK catalytic activity, is the focus of this review.

    Topics: Animals; Cell Transformation, Neoplastic; Cytokines; Enzyme Activation; Humans; Immune Tolerance; Janus Kinase 1; Janus Kinase 2; Janus Kinase 3; Neoplasms; Oxidation-Reduction; Protein-Tyrosine Kinases; Proteins; Proto-Oncogene Proteins; Signal Transduction; Transforming Growth Factor beta

2001
The role of thrombospondin-1 in tumor progression.
    Experimental biology and medicine (Maywood, N.J.), 2001, Volume: 226, Issue:8

    The role of thrombospondin-1 (TSP-1) in tumor progression is both complex and controversial. It is clear from the literature that the function of TSP-1 in malignancy depends on the presence of other factors and the level of TSP-1 expression in the tumor tissue. High levels of TSP-1 secreted by tumors, which were engineered to overexpress TSP-1, inhibit tumor growth, while anti-sense inhibition of TSP-1 production in certain tumors also inhibits growth. Clearly, the presence of other factors in these experimental systems must be important. The role of TSP-1 in angiogenesis also depends on the levels of TSP-1, the presence and level of angiogenic stimulators such as basic fibroblast growth factor (bFGF), and the localization of TSP-1 in the tissue. Matrix-bound TSP-1 promotes capillary tube formation in the rat aorta model of angiogenesis, while TSP-1 inhibits bFGF- induced angiogenesis in the rat cornea model. The inhibitory effect also depends on the proteolytic state of TSP-1 since the amino terminus promotes angiogenesis in the cornea model, while the remaining 140-kDa fragment inhibits bFGF-induced angiogenesis. Both the stimulatory and inhibitory effects of TSP-1 are likely due to upregulation of matrix-degrading enzymes and their inhibitors. These enzymes are critical for maintaining optimal matrix turnover during angiogenesis. These varied TSP-1-dependent mechanisms offer new targets for the development of anti-angiogenic therapeutics for the treatment of a variety of cancers, as well as other pathologies involving inappropriate angiogenesis such as diabetic retinopathy.

    Topics: Animals; Cell Adhesion; Cell Division; Cell Movement; Disease Progression; Humans; Mice; Models, Biological; Neoplasms; Neovascularization, Pathologic; Thrombospondin 1; Transforming Growth Factor beta; Transforming Growth Factor beta1

2001
TGF-beta signaling in tumor suppression and cancer progression.
    Nature genetics, 2001, Volume: 29, Issue:2

    Epithelial and hematopoietic cells have a high turnover and their progenitor cells divide continuously, making them prime targets for genetic and epigenetic changes that lead to cell transformation and tumorigenesis. The consequent changes in cell behavior and responsiveness result not only from genetic alterations such as activation of oncogenes or inactivation of tumor suppressor genes, but also from altered production of, or responsiveness to, stimulatory or inhibitory growth and differentiation factors. Among these, transforming growth factor beta (TGF-beta) and its signaling effectors act as key determinants of carcinoma cell behavior. The autocrine and paracrine effects of TGF-beta on tumor cells and the tumor micro-environment exert both positive and negative influences on cancer development. Accordingly, the TGF-beta signaling pathway has been considered as both a tumor suppressor pathway and a promoter of tumor progression and invasion. Here we evaluate the role of TGF-beta in tumor development and attempt to reconcile the positive and negative effects of TGF-beta in carcinogenesis.

    Topics: Animals; Disease Progression; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2001
Suppressor and oncogenic roles of transforming growth factor-beta and its signaling pathways in tumorigenesis.
    Advances in cancer research, 2001, Volume: 83

    Transforming growth factor-beta (TGF-beta) has been implicated in oncogenesis since the time of its discovery almost 20 years ago. The complex, multifunctional activities of TGF-beta endow it with both tumor suppressor and tumor promoting activities, depending on the stage of carcinogenesis and the responsivity of the tumor cell. Dysregulation or alteration of TGF-beta signaling in tumorigenesis can occur at many different levels, including activation of the ligand, mutation or transcriptional suppression of the receptors, or alteration of downstream signal transduction pathways resulting from mutation or changes in expression patterns of signaling intermediates or from changes in expression of other proteins which modulate signaling. New insights into signaling from the TGF-beta receptors, including the identification of Smad signaling pathways and their interaction with mitogen-activated protein (MAP) kinase pathways, are providing an understanding of the changes involved in the change from tumor suppressor to tumor promoting activities of TGF-beta. It is now appreciated that loss of sensitivity to inhibition of growth by TGF-beta by most tumor cells is not synonymous with complete loss of TGF-beta signaling but rather suggests that tumor cells gain advantage by selective inactivation of the tumor suppressor activities of TGF-beta with retention of its tumor promoting activities, especially those dependent on cross talk with MAP kinase pathways and AP-1.

    Topics: Animals; Cell Differentiation; Genes, Tumor Suppressor; Humans; Models, Biological; Mutation; Neoplasms; Protein Isoforms; Signal Transduction; Transforming Growth Factor beta

2001
TGF-beta signaling in cancer--a double-edged sword.
    Trends in cell biology, 2001, Volume: 11, Issue:11

    Transforming growth factor (TGF) beta1 is a potent growth inhibitor, with tumor-suppressing activity. Cancers are often refractile to this growth inhibition either because of genetic loss of TGF-beta signaling components or, more commonly, because of downstream perturbation of the signaling pathway, such as by Ras activation. Carcinomas often secrete excess TGF-beta1 and respond to it by enhanced invasion and metastasis. Therapeutic approaches should aim to inhibit the TGF-beta-induced invasive phenotype, but also to retain its growth-inhibitory and apoptosis-inducing effects.

    Topics: Animals; Disease Progression; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta

2001
Malignant cells, directors of the malignant process: role of transforming growth factor-beta.
    Cancer metastasis reviews, 2001, Volume: 20, Issue:1-2

    Malignant cells survive and thrive by expressing growth and invasion 'programs' that many normal cell types recognize and respond to in 'programmed' patterns. An early event in the molecular evolution of many malignancies loss of response to growth control by transforming growth factor-beta (TGF-beta) frequently due to mutation in the type I or type II TGF-beta receptor or a Smad protein. The malignant cells secrete TFG-beta that acts on the host to suppress antitumor immune responses, to enhance extracellular matrix production and to augment angiogenesis. These activities resemble those induced by TGF-beta during embryonic development and account in part for the 'de-differentiated' nature of malignant disease. Clinically, TGF-beta1 is often elevated in the plasma of breast cancer patients, lung cancer patients, hepatocellular carcinoma patients, and prostate cancer patients. Preclinically, several breast cancer models and prostate cancer models in vivo have demonstrated a connection between TGF-beta expression and increased tumorigenicity, increased invasion and drug resistance. In other diseases such as colon, gastric, endometrial, ovarian, and cervical cancers and gliomas and melanoma, loss of response to TGF-beta as a growth inhibitor and increased expression of TGF-beta have been associated with malignant conversion and progression. Elevated levels of TGF-beta are measurable in nude mice bearing a wide variety of human tumor xenografts; thus, these tumor models may serve as useful mimics of the human disease with respect to the TGF-beta pathway. Cancer cure may be approached by blocking several of the major normal pathways used for tumor growth and survival in combination with cytotoxic therapies.

    Topics: Animals; Cell Differentiation; Cell Division; Humans; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2001
Histone acetylation/deacetylation and cancer: an "open" and "shut" case?
    Current molecular medicine, 2001, Volume: 1, Issue:4

    DNA in eukaryotic cells is packaged into chromatin. The main packaging component of chromatin is the nucleosome, and this is composed of proteins known as histones. Histones can be reversibly modified in several ways, and the best characterized of these modifications is histone acetylation. This is a reversible modification, which is carried out by two families of enzymes, the histone acetyltransferases (HATs), and the histone deacetylases (HDACs). These enzymes have important activities in many cellular processes including transcription, DNA replication and cell cycle progression. The mechanisms underlying tumor formation are multifaceted, and often involve mutations or alterations of genes involved with the regulation and control of the cell cycle or cell death. Because of their important roles in the regulation of such events, enzymes that affect histone acetylation status are increasingly being associated with tumors. This article describes some of the current knowledge about histone acetyltransferases and histone deacetylases, and how their multitudinal roles in cellular events may have important roles in tumorigensis.

    Topics: Acetylation; Acetyltransferases; Animals; Ataxia Telangiectasia Mutated Proteins; Cell Cycle Proteins; DNA Methylation; DNA Repair; DNA-Binding Proteins; DNA, Neoplasm; Genes, Tumor Suppressor; Histone Acetyltransferases; Histone Deacetylases; Histones; Humans; Hypoxia; Neoplasms; Oncogenes; Oncogenic Viruses; Protein Serine-Threonine Kinases; Retinoblastoma Protein; Saccharomyces cerevisiae Proteins; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

2001
Atherosclerosis: a cancer of the blood vessels?
    American journal of clinical pathology, 2001, Volume: 116 Suppl

    A series of molecular pathways have in common a significant role in the pathogenesis and progression of atherosclerosis and cancer. Shared mechanisms implicated for both diseases include oxidative stress and the cellular damage that results from it, toxic metabolites produced by cigarette smoking, and increased dietary fat intake. Atherosclerosis may begin when an injury or infection mutates or transforms a single arterial smooth muscle cell in the progenitor of a proliferative clone, similar to the most widely held carcinogenesis theory. Cell proliferation regulatory pathways have been associated with plaque progression, stenosis, and restenosis after angioplasty and with cancer progression. Alterations in cell adhesion molecules have been linked to plaque formation and thrombosis and to tumor invasion and metastasis. Altered expression of proteases associated with thrombolysis has been implicated in atherosclerotic plaque expansion and hemorrhage and in the invasion and metastasis of malignant neoplasms. Ligand-growth factor receptor interactions have been associated with early atherosclerotic lesions and with cancer development and spread. Nuclear transcription factors have been associated with progression of both diseases. Angiogenesis modulators have been linked to plaque expansion and restenosis of atherosclerotic lesions and to local and metastatic tumor expansion.

    Topics: Arteriosclerosis; Cell Adhesion Molecules; Cell Division; Dietary Fats; Genetic Predisposition to Disease; Humans; Neoplasms; Neovascularization, Pathologic; NF-kappa B; Oxidative Stress; Receptors, Growth Factor; Smoking; Transcription Factors; Transforming Growth Factor beta

2001
[PEBP2/CBF/AML1: master regulators for tissue specific gene expression].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 2000, Volume: 45, Issue:1

    Topics: Animals; Bone Morphogenetic Proteins; Core Binding Factor Alpha 1 Subunit; Core Binding Factor Alpha 2 Subunit; Core Binding Factors; DNA-Binding Proteins; Gene Expression Regulation, Developmental; Humans; Neoplasm Proteins; Neoplasms; Organ Specificity; Proto-Oncogene Proteins; Signal Transduction; Transcription Factor AP-2; Transcription Factors; Transforming Growth Factor beta

2000
Stem cells: the intestinal stem cell as a paradigm.
    Carcinogenesis, 2000, Volume: 21, Issue:3

    Stem cell research provides a foundation for therapeutic advancement in oncology, clinical genetics and a diverse array of degenerative disorders. For example, the elucidation of pathways governing proliferative regulation and differentiation within cellular systems will result in medical strategies aimed at the root cause of cancer. At present the characterization of reliable stem cell markers is the immediate aim in this particular field. Over the past 30 years investigators have determined many of the physical and functional properties of stem cells through careful and imaginative experimentation. Intestinal stem cells reside at the crypt base and give rise to all cell types found within the crypt. They readily undergo altruistic apoptosis in response to toxic stimuli although their progeny are hardier and will regain stem cell function to repopulate the tissue compartment, giving rise to the concept of a proliferative hierarchy. Contention exists when deciding whether the full complement of cells within a crypt is derived from either a single or multiple stems. Evidence has also arisen to challenge the long held view that colorectal tumours arise from a single mutated stem cell, as early adenomas from a human XO/XY mosaic contained distinct clones. Mechanisms governing the stem cell cycle and subsequent proliferative activity largely remain obscure. The adenomatous polyposis coli gene product has, however, been shown to promote the degradation of beta-catenin, an enhancer of cell proliferation, thereby downregulating this activity in healthy individuals.

    Topics: Adenomatous Polyposis Coli; Animals; Cell Differentiation; Cell Division; Colorectal Neoplasms; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Cyclooxygenase 2; Glucagon-Like Peptides; Humans; Intestinal Mucosa; Intestines; Isoenzymes; Membrane Proteins; Neoplasms; Peptides; Prostaglandin-Endoperoxide Synthases; Proto-Oncogene Proteins c-bcl-2; Signal Transduction; Stem Cells; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2000
Role of transforming growth factor beta in human disease.
    The New England journal of medicine, 2000, May-04, Volume: 342, Issue:18

    Topics: Animals; Arteriosclerosis; Cell Cycle; Embryonic and Fetal Development; Fibrosis; Humans; Immune Tolerance; Neoplasm Metastasis; Neoplasms; Signal Transduction; Telangiectasia, Hereditary Hemorrhagic; Transforming Growth Factor beta

2000
TGF-beta signaling by Smad proteins.
    Advances in immunology, 2000, Volume: 75

    Members of the transforming growth factor-beta (TGF-beta) family bind to type II and type I serine/threonine kinase receptors, which initiate intracellular signals through activation of Smad proteins. Receptor-regulated Smads (R-Smads) are anchored to the cell membrane by interaction with membrane-bound proteins, including Smad anchor for receptor activation (SARA). Upon ligand stimulation, R-Smads are phosphorylated by the receptors and form oligomeric complexes with common-partner Smads (Co-Smads). The oligomeric Smad complexes then translocate into the nucleus, where they regulate the transcription of target genes by direct binding to DNA, interaction with various DNA-binding proteins, and recruitment of transcriptional coactivators or corepressors. A third class of Smads, inhibitory Smads (I-Smads), inhibits the signals from the serine/threonine kinase receptors. Since the expression of I-Smads is induced by the TGF-beta superfamily proteins, Smads constitute an autoinhibitory signaling pathway. The functions of Smads are regulated by other signaling pathways, such as the MAP kinase pathway. Moreover, Smads interact with and modulate the functions of various transcription factors which are downstream targets of other signaling pathways. Loss of function of certain Smads is involved in tumorigenesis, e.g., pancreatic and colorectal cancers. Analyses by gene targeting revealed pivotal roles of Smads in early embryogenesis, angiogenesis, and immune functions in vivo.

    Topics: Animals; Caenorhabditis elegans; Cell Membrane; Cell Nucleus; DNA-Binding Proteins; Drosophila melanogaster; Embryonic and Fetal Development; Fetal Proteins; Gene Expression Regulation; Helminth Proteins; Humans; Insect Proteins; Macromolecular Substances; MAP Kinase Signaling System; Models, Biological; Multigene Family; Neoplasm Proteins; Neoplasms; Phosphorylation; Protein Isoforms; Protein Processing, Post-Translational; Protein Serine-Threonine Kinases; Protein Structure, Tertiary; Receptor Cross-Talk; Receptors, Transforming Growth Factor beta; Signal Transduction; Structure-Activity Relationship; Trans-Activators; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Xenopus

2000
The influence of the microenvironment on the malignant phenotype.
    Molecular medicine today, 2000, Volume: 6, Issue:8

    Normal tissue homeostasis is maintained by dynamic interactions between epithelial cells and their microenvironment. As tissue becomes cancerous, there are reciprocal interactions between neoplastic cells, adjacent normal cells such as stroma and endothelium, and their microenvironments. The current dominant paradigm wherein multiple genetic lesions provide both the impetus for, and the Achilles heel of, cancer might be inadequate to understand cancer as a disease process. In the following brief review, we will use selected examples to illustrate the influence of the microenvironment in the evolution of the malignant phenotype. We will also discuss recent studies that suggest novel therapeutic interventions might be derived from focusing on microenvironment and tumor cells interactions.

    Topics: Animals; Cell Transformation, Neoplastic; Extracellular Matrix; Homeostasis; Humans; Integrins; Neoplasms; Phenotype; Radiation, Ionizing; Transforming Growth Factor beta; Wounds and Injuries

2000
TGF-beta-based immunotherapy for cancer: breaching the tumor firewall.
    The Prostate, 2000, Oct-01, Volume: 45, Issue:2

    Many malignant cells secrete transforming growth factor-beta (TGF-beta), a potent immunosuppresant, suggesting that TGF-beta production may represent a significant tumor escape mechanism from host immunosurveillance. Establishment of a leukocyte subpopulation with disrupted TGF-beta signaling in the tumor-bearing host offers a potential means for immunotherapy of cancer. Downregulation of TGF-beta secretion in tumor cells results in restoration of immunogenicity in the host, while T-cell insensitivity to TGF-beta results in accelerated differentiation and autoimmunity, elements of which may be required in order to combat self-antigen-expressing tumors in a tolerized host. The rationale, approaches, and potential pitfalls of this strategy will be discussed.

    Topics: Animals; Autoimmunity; Genetic Therapy; Humans; Immune Tolerance; Immunotherapy; Neoplasms; Transforming Growth Factor beta

2000
TGF-beta/SMAD signaling and its involvement in tumor progression.
    Biological & pharmaceutical bulletin, 2000, Volume: 23, Issue:10

    Cytokines of the transforming growth factor-beta (TGF-beta) superfamily are multifunctional peptides that regulate growth and differentiation of various types of cells. Members of the TGF-beta superfamily bind to type 11 and type I serine/threonine kinase receptors, which mediate intracellular signals through SMAD proteins. Of 3 subtypes of SMADs, receptor-regulated SMADs are phosphorylated by the serine/threonine kinase receptors, form complexes with common-mediator SMAD, and move into the nucleus, where they act as components of transcription factor complexes. Abnormalities of the TGF-beta receptors and SMADs have been detected in various tumors, including colorectal cancers and pancreatic cancers. Inhibitory SMADs and transcriptional co-repressors, including c-Ski and SnoN, repress the TGF-beta/SMAD signaling. Perturbation of the TGF-beta/SMAD signaling pathway may result in progression of tumors through resistance of the cells to the growth inhibition induced by TGF-beta.

    Topics: Animals; Disease Progression; Humans; Neoplasms; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2000
TGFbeta signaling in growth control, cancer, and heritable disorders.
    Cell, 2000, Oct-13, Volume: 103, Issue:2

    Topics: Cell Differentiation; Cell Division; Genetic Diseases, Inborn; Humans; Models, Biological; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2000
The latent transforming growth factor beta binding protein (LTBP) family.
    The Biochemical journal, 2000, Dec-15, Volume: 352 Pt 3

    The transforming growth factor beta (TGFbeta) cytokines are a multi-functional family that exert a wide variety of effects on both normal and transformed mammalian cells. The secretion and activation of TGFbetas is regulated by their association with latency-associated proteins and latent TGFbeta binding proteins (LTBPs). Over the past few years, three members of the LTBP family have been identified, in addition to the protoype LTBP1 first sequenced in 1990. Three of the LTBP family are expressed in a variety of isoforms as a consequence of alternative splicing. This review summarizes the differences between the isoforms in terms of the effects on domain structure and hence possible function. The close identity between LTBPs and members of the fibrillin family, mutations in which have been linked directly to Marfan's syndrome, suggests that anomalous expression of LTBPs may be associated with disease. Recent data indicating that differential expression of LTBP1 isoforms occurs during the development of coronary heart disease is considered, together with evidence that modulation of LTBP function, and hence of TGFbeta activity, is associated with a variety of cancers.

    Topics: Alternative Splicing; Animals; Arteriosclerosis; Carrier Proteins; Humans; Intracellular Signaling Peptides and Proteins; Latent TGF-beta Binding Proteins; Multigene Family; Neoplasms; Organ Specificity; Transforming Growth Factor beta

2000
Signaling of transforming growth factor-beta family members through Smad proteins.
    European journal of biochemistry, 2000, Volume: 267, Issue:24

    Smads are pivotal intracellular nuclear effectors of transforming growth factor-beta (TGF-beta) family members. Ligand-induced activation of TGF-beta family receptors with intrinsic serine/threonine kinase activity trigger phosphorylation of receptor-regulated Smads (R-Smads), whereas Smad2 and Smad3 are phosphorylated by TGF-beta, and activin type I receptors, Smad1, Smad5 and Smad8, act downstream of BMP type I receptors. Activated R-Smads form heteromeric complexes with common-partner Smads (Co-Smads), e.g. Smad4, which translocate efficiently to the nucleus, where they regulate, in co-operation with other transcription factors, coactivators and corepressors, the transcription of target genes. Inhibitory Smads act in most cases in an opposite manner from R- and Co-Smads. Like other components in the TGF-beta family signaling cascade, Smad activity is intricately regulated. The multifunctional and context dependency of TGF-beta family responses are reflected in the function of Smads as signal integrators. Certain Smads are somatically mutated at high frequency in particular types of human cancers. Gene ablation of Smads in the mouse has revealed their critical roles during embryonic development. Here we review the latest advances in our understanding of the Smad mechanism of action and their in vivo functions.

    Topics: DNA-Binding Proteins; Gene Targeting; Humans; Neoplasms; Signal Transduction; Subcellular Fractions; Trans-Activators; Transforming Growth Factor beta; Ubiquitins

2000
[Transforming growth factor-beta signaling and cancer].
    Human cell, 2000, Volume: 13, Issue:3

    Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that inhibits the growth of most types of cells and induces fibrosis of various tissues. TGF-beta binds to two different types of serine/threonine kinase receptors, and activate Smad proteins. Perturbation of the TGF-beta signaling pathway results in tumorigenesis of various tissues. TGF-beta/Smad signaling is regulated by several regulatory proteins, including inhibitory Smads and transcriptional corepressors. Abnormalities in these negative regulators of TGF-beta signaling may also involved in tumorigenesis of certain tissues.

    Topics: Animals; DNA-Binding Proteins; Humans; Neoplasms; Nuclear Proteins; Proto-Oncogene Proteins; Receptors, Transforming Growth Factor beta; Signal Transduction; Trans-Activators; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta

2000
Innovative treatment programs against cancer: II. Nuclear factor-kappaB (NF-kappaB) as a molecular target.
    Biochemical pharmacology, 1999, Jan-01, Volume: 57, Issue:1

    Nuclear factor-kappaB (NF-kappaB) activity affects cell survival and determines the sensitivity of cancer cells to cytotoxic agents as well as to ionizing radiation. Preventing the protective function of NF-kappaB may result in chemo- and radio-sensitization of cancer cells. Therefore, NF-kappaB has emerged as one of the most promising molecular targets in rational drug design efforts of translational cancer research programs.

    Topics: Animals; Cell Survival; Drug Design; Gene Expression Regulation; Gene Expression Regulation, Viral; Humans; Models, Biological; Neoplasms; NF-kappa B; Radiation, Ionizing; Signal Transduction; Transforming Growth Factor beta

1999
Development of TGF-beta resistance during malignant progression.
    Archives of pharmacal research, 1999, Volume: 22, Issue:1

    Transforming growth factor-beta (TGF-beta) is the prototypical multifunctional cytokine, participating in the regulation of vital cellular activities such as proliferation and differentiation as well as a number of basic physiological functions. The effects of TGF-beta are critically dependent on the expression and distribution of a family of TGF-beta receptors, the TGF-beta types I, II, and III. It is now known that a wide variety of human pathology can be caused by aberrant expression and function of these receptors. The coding sequence of the type II receptor (RII) appears to render it uniquely susceptible to DNA replication errors in the course of normal cell division. By virtue of its key role in the regulation of cell proliferation, TGF-beta RII should be considered as a tumor suppressor gene. High levels of mutation in the TGF-beta RII gene have been observed in a wide range of primarily epithelial malignancies, including colon and gastric cancer. It appears likely that mutation of the TGF-beta RII gene may be a very critical step in the pathway of carcinogenesis.

    Topics: Animals; Disease Progression; ErbB Receptors; Humans; Mutation; Neoplasms; Transforming Growth Factor beta

1999
Translational control of growth factor and proto-oncogene expression.
    The international journal of biochemistry & cell biology, 1999, Volume: 31, Issue:1

    Control of translation is now understood to be one of the major regulatory events in eukaryotic gene expression. Moreover there is evidence which suggests that aberrant expression of growth-related genes by translational mechanisms makes a significant contribution to cell transformation. However, the mechanisms which regulate translation of specific growth-related mRNAs have yet to be fully elucidated. The majority of these mRNAs have long 5' untranslated regions (UTRs) and three features which are important in translational control have been identified, namely (i) structured regions which inhibit the scanning mechanisms of translation, (ii) regulatory upstream open reading frames and (iii) internal ribosome entry segments which are capable of initiating cap-independent translation. In this review the translational regulation of specific mRNAs encoding growth factors and proto-oncogenes by these three mechanisms will be discussed, together with examples of altered translational regulation in neoplasia.

    Topics: 5' Untranslated Regions; Animals; Fibroblast Growth Factor 2; Gene Expression Regulation, Neoplastic; Genes, mos; Growth Substances; Humans; Neoplasms; Nuclear Proteins; Platelet-Derived Growth Factor; Protein Biosynthesis; Proto-Oncogene Mas; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-bcl-2; Proto-Oncogene Proteins c-mdm2; Proto-Oncogenes; Ribosomes; RNA, Messenger; Transforming Growth Factor beta

1999
Bisphosphonates as cancer drugs.
    Hospital practice (1995), 1999, May-15, Volume: 34, Issue:5

    Bone appears to act like fertilizer for many tumors, including myeloma and metastatic breast cancer. The explanation must lie in interactions between tumor cells and the bone-tissue microenvironment. At this level, too, must lie the explanation of how bisphosphonates address not only cancer osteolysis but also the tumor burden. By inhibiting osteoclasts, the drugs may block a cancer-related vicious cycle.

    Topics: Antineoplastic Agents; Clodronic Acid; Diphosphonates; Etidronic Acid; Humans; Neoplasm Proteins; Neoplasms; Osteolysis; Pamidronate; Parathyroid Hormone-Related Protein; Proteins; Risedronic Acid; Transforming Growth Factor beta

1999
Transforming growth factor-beta and p-21: multiple molecular targets of decorin-mediated suppression of neoplastic growth.
    Cell and tissue research, 1999, Volume: 296, Issue:2

    Decorin is a member of the small leucine-rich proteoglycan (SLRP) gene family that has recently become a focus in various areas of cancer research. The decorin protein consists of a core protein and a covalently linked glycosaminoglycan chain. Decorin binds to collagens type I, II and IV in vivo and promotes the formation of fibers with increased stability and changes in solubility. Further, the decorin core protein binds to growth factors, including transforming growth factor-beta (TGF-beta), to other intercellular matrix molecules such as fibronectin and thrombospondin, and to the decorin endocytosis receptor. Decorin may directly interfere with the cell cycle via the induction of p21WAF1/CIP1 (p21), a potent inhibitor of cyclin-dependent kinases (CDKs). Here, we discuss interactions of decorin with TGF-beta and with p21, both of which are relevant to carcinogenesis and tumor progression. TGF-beta is released by tumors of various histogenetic origins and promotes immunosuppression in the host and tumor immune escape by induction of growth arrest and apoptosis in immune cells, by downregulation of MHC II antigen expression and by changes in the cytokine release profiles of immune and tumor cells. Moreover, TGF-beta may modulate tumor growth in an autocrine and paracrine fashion, may mediate drug resistance, and may facilitate tumor angiogenesis. Decorin binds to TGF-beta, thus inhibiting its bioactivity, and is a direct or indirect negative modulator of TGF-beta synthesis. Ectopic expression of decorin results in the regression of rat C6 gliomas, an antineoplastic effect attributed to the reversal of TGF-beta-induced immunosuppression. On the other hand, de novo expression of decorin in colon cancer cells and some other tumor cells, even though not in glioma cells, results in an upregulation of p21 expression and a cell cycle arrest, presumably in a TGF-beta-independent manner. Decorin expression is downregulated in many tumors but upregulated in the peritumoral stroma. By virtue of its growth regulatory and immunomodulatory properties, decorin promises to become a novel target for the experimental therapy of human cancers.

    Topics: Animals; Cell Cycle; Cell Division; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Decorin; Enzyme Inhibitors; Extracellular Matrix Proteins; Gene Expression Regulation; Humans; Models, Biological; Neoplasms; Proteoglycans; Rats; Transforming Growth Factor beta

1999
Effects of TGF-beta on the immune system: implications for cancer immunotherapy.
    Leukemia, 1999, Volume: 13, Issue:8

    Transforming growth factor-beta (TGF-beta) is a potent regulator of numerous processes including hematopoiesis, cell proliferation, differentiation and activation. TGF-beta has pleiotropic and profound effects on the immune system and on hematologic malignancies, ie leukemia. It is the most potent immunosuppressor described to date. Evidence exists that the immunosuppressive potential of TGF-beta is an important promoter of malignant cell growth. This is partly caused by TGF-beta-induced interference with the generation of tumor-specific cytotoxic T lymphocytes, but also by TGF-beta-induced promotion of angiogenesis and tumor stroma formation. Until now, significant clinical responses have not been achieved with the current cancer immunotherapeutic approaches. One of the possible explanations for this failure is immunosuppression induced by tumor-derived TGF-beta. Here, several strategies to counteract the immunosuppressive effects of TGF-beta and the current limitations of these strategies will be discussed. Knowledge of the mechanisms by which TGF-beta interferes with the development of an anti-tumor response and of the strategies to counteract these immunosuppressive activities is crucial to improve the current cancer immunotherapeutic approaches.

    Topics: Animals; Cytotoxicity, Immunologic; Humans; Immunotherapy; Neoplasms; Neoplasms, Experimental; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta

1999
Regulation of megakaryocytopoiesis.
    Haemostasis, 1999, Volume: 29, Issue:1

    After 35 years of research, a physiological regulator of platelet production has been identified and the recombinant protein is available. With the discovery of thrombopoietin (TPO), its potential use in a wide variety of clinical megakaryocytic and platelet disorders has been expected and clinical trials have been undertaken. To date, the reported encouraging pre-clinical studies indicate that, as with erythropoietin or G-CSF, minimal toxicity can be expected. A potential limiting side-effect of TPO could be the induction of thrombosis. Nevertheless, it is too early to know whether this cytokine will be of major therapeutic importance for patients with life-threatening thrombocytopenia, such as patients undergoing bone marrow transplantation or subjected to a high dose of chemotherapy. Several experimental and clinical studies are still needed to determine the efficacy of TPO in the prevention or the amelioration of bleeding, which is the ultimate goal for the appropriate use of cytokines with haemostatic benefit. Basic and clinical studies on regulators of megakaryocytopoiesis have rapidly progressed. Now, there is no doubt that some of these regulators are effective in correcting haematopoietic disorders of various aetiologies. Studies on negative regulators not only are important to understand the regulation of megakaryocytopoiesis in normal and pathological states but also have a potential clinical application. Some of these regulators have been shown to be effective in the treatment of essential thrombocythaemia and other myeloproliferative disorders. Platelet factor 4 (PF4) and some other chemokines are also capable of protecting progenitor cells from the cytotoxicity of chemotherapeutic drugs. However, detailed investigations are still required to determine the precise mechanism(s) of action of these regulators and to establish the optimal clinical protocols of negative regulators alone or in association with positive regulators for the treatment of various haematological diseases and cancer.

    Topics: Animals; Chemokines; Clinical Trials as Topic; Cytokines; Hematopoiesis; Hematopoietic Cell Growth Factors; Humans; Interferon-alpha; Macaca mulatta; Megakaryocytes; Neoplasm Proteins; Neoplasms; Platelet Factor 4; Proto-Oncogene Proteins; Quinazolines; Radiation Injuries, Experimental; Receptors, Cytokine; Receptors, Thrombopoietin; Recombinant Proteins; Thrombin; Thrombocytopenia; Thrombopoietin; Transforming Growth Factor beta

1999
Immune privilege, tumors, and the eye.
    Chemical immunology, 1999, Volume: 73

    Topics: Animals; Antigen Presentation; Antigens, Differentiation; Antigens, Neoplasm; Apoptosis; Endothelial Growth Factors; Eye; Fas Ligand Protein; Gene Expression Regulation, Neoplastic; Genes, MHC Class I; Graft Survival; Histocompatibility Antigens Class I; Humans; Immune Tolerance; Immunization; Immunotherapy; Interleukin-10; Killer Cells, Natural; Lymphokines; Membrane Glycoproteins; Mice; Neoplasm Transplantation; Neoplasms; Receptors, Immunologic; Receptors, KIR; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

1999
The Smads.
    The international journal of biochemistry & cell biology, 1999, Volume: 31, Issue:11

    The Smads are a family of intracellular signalling molecules that act downstream of receptors for the transforming growth factor (TGF)-beta family of ligands. Three classes of Smads have been identified. The receptor-regulated Smads are direct substrates for the type I receptors, which are serine/threonine kinases. Once phosphorylated and activated, these Smads form hetero-oligomeric complexes with a second class of Smad, the common mediator Smads. These Smad complexes translocate to the nucleus, where they are recruited to DNA primarily by site-specific DNA binding transcription factors, and participate in regulating the transcription of target genes. Inhibitory Smads are the third identified class which antagonise the activity of the receptor-regulated Smads. Aberrant TGF-beta signalling has been associated with several human diseases such as cancer and fibrosis. The identification of the Smads as primary transducers of TGF-beta signals raises the possibility that agents directed at modulating Smad activity would have therapeutic applications.

    Topics: Animals; DNA-Binding Proteins; Fibrosis; Humans; Neoplasms; Phylogeny; Protein Structure, Tertiary; Signal Transduction; Trans-Activators; Transforming Growth Factor beta

1999
TGF-beta and cancer.
    Microbes and infection, 1999, Volume: 1, Issue:15

    The relationships between transforming growth factor-beta (TGF-beta) and cancer are varied and complex. The paradigm that is emerging from the experimental evidence accumulated over the past decade or so is that TGF-beta can play two different and opposite roles with respect to the process of malignant progression. During early stages of carcinogenesis, TGF-beta acts predominantly as a potent tumor suppressor and may mediate the actions of chemopreventive agents such as retinoids and nonsteroidal anti-estrogens. However, at some point during the development and progression of malignant neoplasms, bioactive TGF-betas make their appearance in the tumor microenvironment and the tumor cells escape from TGF-beta-dependent growth arrest. In many cases, this resistance to TGF-beta is the consequence of loss or mutational inactivation of the genes that encode signaling intermediates. These include the types I and II TGF-beta receptors, as well as receptor-associated and common-mediator Smads. The stage of tumor development or progression at which TGF-beta-resistant clones come to dominate the tumor cell population in different types of neoplasm remains to be defined. The phenotypic switch from TGF-beta-sensitivity to TGF-beta-resistance that occurs during carcinogenesis has several important implications for cancer prevention and treatment.

    Topics: Animals; Humans; Mice; Neoplasms; Signal Transduction; Trans-Activators; Transforming Growth Factor beta

1999
The role for transforming growth factor-beta (TGF-beta) in human cancer.
    Critical reviews in oncogenesis, 1999, Volume: 10, Issue:4

    Uncontrolled cellular proliferation is a hallmark of cancer. Thus, a relevant and important question is how cancer cells have escaped from normal growth regulatory mechanisms to become malignant and, further, what events favor progression and metastasis. Growth regulatory proteins of the transforming growth factor-beta family (TGF-beta) are one of the few classes of endogenous inhibitors of cell growth. Contrary to the first notion that these proteins may be downregulated in cancer cells to promote their growth, generally it has been otherwise found that there is a marked increase in the expression of TGF-beta mRNA and protein in human cancers (in vivo), including those of the pancreas, colon, stomach, lung, endometrium, prostate, breast, brain, and bone. Furthermore, in many of these cancers high expression correlates with more advanced stages of malignancy and decreased survival. The increased expression of TGF-beta is usually accompanied by a loss in the growth inhibitory response to TGF-beta. For example, certain tumor cells in culture (i.e., colon carcinoma and glioblastoma multiforme) demonstrate a progressive loss of the growth inhibitory response to TGF-beta that varies directly with the malignant stage of the original tumor, and the most aggressive forms actually switch to being autocrine and/or paracrine growth stimulated by TGF-beta. The study of the molecular events associated with the escape of tumor cells from growth regulation by TGF-beta has provided insight into mechanisms underlying carcinogenesis. The mechanisms for upregulation of TGF-beta are unknown. However, once malignant cells lose their growth inhibitory response to TGF-beta and produce massive amounts of these proteins, the increased expression of TGF-beta provides a selective advantage for tumor cell survival as TGF-betas are also angiogenic and have potent immunosuppressive effects, including specifically inhibiting tumoricidal natural and lymphocyte-activated killer cells. In light of the significant role for TGF-betas in regulating cell growth, it is not surprising that in more recent years studies have shown that specific genetic alterations involved in the signaling pathway for TGF-beta-mediated growth inhibition have occurred in many human cancers. Specific defects in TGF-beta receptors, TGF-beta-related-signal transduction/gene activation, and TGF-beta-regulated cell cycle proteins, have all been implicated in the oncogenesis of many human cancers. In this context, compo

    Topics: Animals; Humans; Neoplasms; Transforming Growth Factor beta

1999
TGFbeta signaling pathways and human diseases.
    Cancer metastasis reviews, 1999, Volume: 18, Issue:2

    Recent progress in deciphering the TGFbeta pathway has uncovered a new signaling molecule, the Smads, and with this finding now gives us insights into how TGFbeta-like signals are transmitted from outside the cell to the nucleus. As we learn more about how TGFbeta regulates normal development, we also are gaining insights into diseases that are caused by mis-regulation or mutation of various components of the signaling pathways.

    Topics: Animals; Caenorhabditis elegans; DNA-Binding Proteins; Drosophila; Humans; Ligands; Neoplasms; Signal Transduction; Transforming Growth Factor beta

1999
TGF-beta receptors and signalling mechanisms.
    Mineral and electrolyte metabolism, 1998, Volume: 24, Issue:2-3

    Transforming growth factor-beta (TGF-beta) is the founding member of a large superfamily of related growth and differentiation factors that include bone morphogenetic proteins and activins. TGF-beta signals through two related transmembrane ser/thr kinase receptors, the type I and type II receptors. Signalling is initiated when the ligand binds to the type II receptor which is followed by recruitment of the type I receptor into a heteromeric complex. Within the complex the type II receptor transphosphorylates and activates the type I receptor kinase which targets downstream signalling components of the pathway. Proteins related to the Drosophila gene Mothers against dpp (MAD) are critical downstream substrates of the type I kinase. The vertebrate members of the MAD-related family, termed Smad2 and Smad3, interact specifically with the TGF-beta type I receptor and are phosphorylated on the last two serines of a conserved C-terminal SSXS motif. This phosphorylation induces association between these receptor-regulated Smads and Smad4 followed by translocation of the heteromeric complex to the nucleus. In the nucleus, heteromeric complexes of Smads can interact with DNA and with specific DNA binding transcription factors to elicit gene responses to TGF-beta. Thus TGF-beta signalling involves a direct pathway from the cell surface receptors to the nucleus. Recently, a novel mechanism to negatively regulate TGF-beta signalling was described that involves another class of MADR proteins. These anti-MADR proteins potently inhibit TGF-beta signalling by functioning as direct antagonists of the TGF-beta receptor type I kinase domain.

    Topics: Animals; Cell Nucleus; Genes, Tumor Suppressor; Humans; Insect Proteins; Macromolecular Substances; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Trans-Activators; Transforming Growth Factor beta

1998
Smads are the central component in transforming growth factor-beta signaling.
    Pharmacology & therapeutics, 1998, Volume: 78, Issue:1

    Until recently, little was known about how transforming growth factor (TGF)-beta signals are transduced to the nucleus. With the discovery of the Smad proteins initially in Drosophila and C. elegans, the unraveling of the pathway has begun. Nine different vertebrate members also have been reported, indicating that Smads are a conserved component of the TGF-beta pathway. Currently, there are three functional classes of Smads. Class I Smads are phosphorylated by TGF-beta receptors and move to the nucleus. The Class II Smads function with Class I Smads, while Class III Smads antagonize the function of Class I Smads. New evidence shows that Smads bind specific DNA sequences and induce transcription of downstream target genes, thus placing the Smads at the center of the TGF-beta signaling pathway.

    Topics: Animals; Caenorhabditis elegans; DNA-Binding Proteins; Drosophila; Evolution, Molecular; Mutation; Neoplasms; Phosphorylation; Promoter Regions, Genetic; Receptors, Transforming Growth Factor beta; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

1998
Molecular mechanisms of transforming growth factor-beta signaling.
    Endocrine reviews, 1998, Volume: 19, Issue:3

    Topics: Animals; Cell Cycle; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

1998
TGF-beta signaling and cancer: structural and functional consequences of mutations in Smads.
    Molecular medicine today, 1998, Volume: 4, Issue:6

    Transforming growth factor-beta (TGF-beta) and related cytokines control the development and homeostasis of many tissues by regulating the expression of genes that determine cell phenotype. Recent progress has elucidated the way in which members of the TGF-beta family initiate their signal through transmembrane receptors and transmit it to target genes via the Smad family of signal-transducing proteins. This review describes TGF-beta signaling pathways as currently understood and mutations of the genes that encode Smads that disrupt the function of these proteins and cause various forms of cancer.

    Topics: Animals; DNA-Binding Proteins; Humans; Neoplasms; Neoplasms, Experimental; Signal Transduction; Smad Proteins; Structure-Activity Relationship; Trans-Activators; Transforming Growth Factor beta

1998
Extracellular matrix-associated transforming growth factor-beta: role in cancer cell growth and invasion.
    Advances in cancer research, 1998, Volume: 75

    Growth factors of the transforming growth factor-beta (TGF-beta) family inhibit the proliferation of epithelial, endothelial, and hematopoietic cells, and stimulate the synthesis of extracellular matrix components. TGF-beta s are secreted from cells in high-molecular-mass protein complexes that are composed of three proteins, the mature TGF-beta-dimer, the TGF-beta propeptide dimer, or latency-associated protein (LAP), and the latent TGF-beta binding protein (LTBP). Mature TGF-beta is cleaved from its propeptide during secretion, but the proteins remain associated by noncovalent interactions. LTBP is required for efficient secretion and processing of latent TGF-beta and it binds to LAP via disulfide bond(s). LTBP is a component of extracellular matrix microfibrils, and it targets the latent TGF-beta complex to the extracellular matrix. TGF-beta signaling is initiated by proteolytic cleavage of LTBP that results in the release of the latent TGF-beta complex from the extracellular matrix. TGF-beta is activated by dissociation of LAP from the mature TGF-beta. Subsequent signaling involves binding of active TGF-beta to its type II cell surface receptors, which phosphorylate and activate type I TGF-beta receptors. Type I receptors, in turn, phosphorylate cytoplasmic transcriptional activator proteins Smad2 and Smad3, inducing their translocation to the nucleus. Recent evidence suggests that acquisition of resistance to TGF-beta growth inhibition plays a major role in the progression of epithelial and hematopoietic cell malignancies. The role of secretion of TGF-beta in tumorigenesis is more complex. The secretion of TGF-beta s by tumor cells may contribute to autocrine growth inhibition, but on the other hand, it may also promote invasion, metastasis, angiogenesis, and even immunosuppression. Tumor cells may also fail to deposit LTBP:TGF-beta complexes to the extracellular matrix. The elucidation of the mechanisms of the release of TGF-beta from the matrix and its subsequent activation aids the understanding of the pathophysiologic roles of TGF-beta in malignant growth, and allows the development of therapeutic agents that regulate the activity of TGF-beta.

    Topics: Cell Division; Extracellular Matrix; Gene Expression Regulation, Neoplastic; Humans; Neoplasm Invasiveness; Neoplasms; Signal Transduction; Transforming Growth Factor beta

1998
[Cyclin A: a good markers for the study of cell cycle control and tumor progression?].
    Comptes rendus des seances de la Societe de biologie et de ses filiales, 1998, Volume: 192, Issue:2

    Cyclin A is a positive regulatory component of kinases required for the progression through S phase and for the transition between the G2 and M phases of the cell division cycle. Previous studies conducted in established cell lines and in primary human T lymphocytes, have demonstrated that the promoter of its gene is under negative transcriptional control in quiescent cells. The DNA sequences mediating this repression have been delineated through in vitro mutagenesis as well as in vivo genomic footprinting experiments. Indirect observations suggest the involvement of proteins related to the retinoblastoma tumor suppressor protein (pRb). Using primary fibroblasts from either pRb(-/-), p107(-/-), p130(-/-) or p107(-/-)/p130(-/-) mice, we show in this work that mutation of the pRb gene has the more profound effect on cyclin A transcription. Finally, normal fibroblasts cultured in suspension fail to express cyclin A and can no longer enter S phase and proliferate, revealing thus a dependence of cyclin A expression on cell anchorage. Our work suggests the existence of at least two sets of regulators controlling cell cycle progression. On the one hand, proteins like cyclin D1, whose expression is a direct consequence of the activation of the ras signalling pathway and on the other hand, proteins like cyclin A which are secondary response effectors. As a result, growth factor stimulation leads to a transcriptional activation of the former set, while the transcription of the latter set is under the control of a repressor whose effect is alleviated after triggering the ras cascade. The status of pRb thus dictates whether cells continue their progression through the cell cycle when ras is mutated, probably by allowing the uncontrolled expression of critical genes like cyclin A.

    Topics: Animals; Biomarkers; Cell Cycle; Cell Transformation, Neoplastic; Cyclin A; Disease Progression; Genes, Retinoblastoma; Humans; Mice; Mice, Knockout; Mutagenesis; Neoplasm Invasiveness; Neoplasms; Transforming Growth Factor beta

1998
TGF-beta signal transduction.
    Annual review of biochemistry, 1998, Volume: 67

    The transforming growth factor beta (TGF-beta) family of growth factors control the development and homeostasis of most tissues in metazoan organisms. Work over the past few years has led to the elucidation of a TGF-beta signal transduction network. This network involves receptor serine/threonine kinases at the cell surface and their substrates, the SMAD proteins, which move into the nucleus, where they activate target gene transcription in association with DNA-binding partners. Distinct repertoires of receptors, SMAD proteins, and DNA-binding partners seemingly underlie, in a cell-specific manner, the multifunctional nature of TGF-beta and related factors. Mutations in these pathways are the cause of various forms of human cancer and developmental disorders.

    Topics: Biological Transport; DNA-Binding Proteins; Gene Expression Regulation; Genetic Diseases, Inborn; Humans; Neoplasms; Protein Serine-Threonine Kinases; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad1 Protein; Trans-Activators; Transforming Growth Factor beta

1998
Transforming growth factor-beta protection of cancer cells against tumor necrosis factor cytotoxicity is counteracted by hyaluronidase (review).
    International journal of molecular medicine, 1998, Volume: 2, Issue:6

    Numerous cancer cells, when exposed to transforming growth factor beta (TGF-beta), become resistant to tumor necrosis factor (TNF) cytotoxicity. Pretreatment of L929 fibroblasts, for example, with TGF-beta isoforms (beta 1, beta 2 and beta 3) for at least 0.5-1 h results in resistance to TNF killing. TGF-beta 1 mediates the following sequential events in L929 cells: i) rapid induction of protein tyrosine-phosphorylation (< 30 min), ii) stimulation of protective protein synthesis and acquisition of TNF resistance (approximately 0.5-1 h), and iii) suppression of I kappa B-alpha expression (1-2 h). Two protective proteins induced by TGF-beta 1 are a 46 kDa extracellular matrix TNF-resistance triggering (TRT) protein and a putative transmembrane anti-apoptotic adhesion protein TIF2 (containing and RGD motif in the extracellular region). Both proteins enable L929 cells to resist TNF killing. Notably, testicular hyaluronidase increases TNF sensitivity in several types of cancer cells, counteracts TGF-beta-mediated TNF-resistance, and suppresses TGF-beta 1 gene expression in L929 cells in a serum-dependent manner. Moreover, hyaluronidase antagonizes TGF-beta-mediated inhibition of epithelial cell growth. Both TGF-beta and hyaluronidase are essential for the progression and invasiveness of breast, prostate and other cancers. Conceivably, a stage-dependent expression, as well as a balanced production, of these proteins is essential for cancer development and self protection against TNF cytotoxicity.

    Topics: Animals; Extracellular Matrix Proteins; Humans; Hyaluronoglucosaminidase; Male; Neoplasms; NF-kappa B; Nuclear Receptor Coactivator 2; Testis; Transcription Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

1998
Smad4 (DPC4)--a potent tumour suppressor?
    British journal of cancer, 1998, Volume: 78, Issue:12

    The recently described family of Smad molecules are essential mediators of transforming growth factor beta (TGF-beta) signalling. To date, seven members of this family have been identified, each of which plays a specific and separate role in mediating TGF-beta superfamily gene transcription. At least two different Smads, Smad2 and Smad4 (DPC4), have been implicated in human cancer and appear to have tumour-suppressor functions. Loss of function of Smad4 is most strongly associated with human pancreatic and colorectal malignancy. Furthermore, work from several different groups has suggested associations between Smad4 loss and malignancy in a number of other tissues. Here, we present a review of the current state of the literature implicating the central Smad mediator, Smad4, in the development of cancer.

    Topics: Animals; Chromosomes, Human, Pair 18; Genes, Tumor Suppressor; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

1998
Cancer as a complex developmental disorder--nineteenth Cornelius P. Rhoads Memorial Award Lecture.
    Cancer research, 1998, Dec-15, Volume: 58, Issue:24

    The processes of differentiation and tumorigenesis have been long thought to be connected. The recent identification of Patched, a gene essential for Drosophila embryonic development, as a tumor suppressor has focused attention on the concept that tumorigenesis involves abnormalities of development. In fact, a large number of genes in the signalling pathway of the Patched gene are either tumor suppressors or oncogenes. This supports the concept that growth control is a critical requirement of differentiation, and that aberrant cellular development can contribute to malignancy. Whereas the identification of genes that result in dominantly inherited cancer syndromes has played a vital role in understanding cancer, the vast majority of "sporadic" cancers have properties of a complex genetic disease. Approaches to identify common alleles in cancer-associated genes promise to increase our understanding of the disease and aid the rational design of preventative and therapeutic strategies.

    Topics: Animals; Cell Differentiation; Cell Division; DNA Mutational Analysis; Embryonic and Fetal Development; Gene Expression Regulation, Developmental; Genes; Genes, Tumor Suppressor; Humans; Neoplasms; Polymorphism, Genetic; Risk; Transforming Growth Factor beta

1998
Cyclin-dependent kinase regulation during G1 phase and cell cycle regulation by TGF-beta.
    Advances in cancer research, 1997, Volume: 71

    The aim of this review is to provide insight into the molecular mechanisms by which transforming growth factor-beta (TGF-beta) modulates cell cycle progression in different cell types. Particular attention is focused on the differences between these mechanisms in cells of epithelial origin and in mesenchymally derived cells. This is important because many transformed epithelial cells lose responsiveness to the growth-inhibitory effects of TGF-beta, thus generating a more fibroblast-like phenotype. Loss of negative growth control, including a lack of response to growth-inhibitory factors, is a common feature of many tumor cells. G1 phase cyclin-dependent kinases (cdks) and their inhibitors (ckis) are central to the pathways that regulate commitment to cellular division in response to positive as well as negative growth effectors. Many checkpoints are deregulated in oncogenesis, and this is often due to alterations in cyclin-cdk complexes. The loss of R-point regulation, in particular, can allow cell growth and division to proceed autonomously of external signals. This may occur due to either the aberrant expression of positive regulators, such as the cyclins and cdks, or the loss of negative regulators, such as the ckis. Beginning with a survey of the role of the cdks in the mammalian cell cycle, the review examines how cdk activity is modulated by cyclin binding, phosphorylation, and ckis, including the Ink4 proteins and the closely related inhibitors p21Cip1 and p27Kip1. Particular attention is paid to the role of p27Kip1 and p21Cip1 in the mechanisms of TGF-beta-induced suppression or stimulation of the cell cycle and how these mechanisms contrast between epithelial cells and cells of mesenchymal origin. Other aspects of TGF-beta signal transduction are discussed, including its effects on cyclin and cdk expression in various cell types, and the downstream targets of cdks and their modulation by TGF-beta and other growth factors are also discussed. These include proteins of the retinoblastoma family, and the related modulation of the transcriptional activity of the E2F family members. Finally, the role of cell cycle regulatory proteins in oncogenesis is review in view of the findings described here.

    Topics: Animals; Carrier Proteins; Cell Cycle; Cell Cycle Proteins; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p16; Cyclin-Dependent Kinase Inhibitor p27; Cyclin-Dependent Kinases; Cyclins; G1 Phase; Humans; Microtubule-Associated Proteins; Neoplasms; Phosphorylation; Retinoblastoma Protein; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Proteins

1997
Towards a unified model of tumor suppression: lessons learned from the human patched gene.
    Biochimica et biophysica acta, 1997, Apr-18, Volume: 1332, Issue:2

    Topics: Animals; Carcinoma, Basal Cell; Drosophila Proteins; Genes, Tumor Suppressor; Insect Proteins; Membrane Proteins; Neoplasms; Oncogenes; Patched Receptors; Receptors, Cell Surface; Signal Transduction; Transforming Growth Factor beta

1997
Reversal of tumor-induced immunosuppression: a new approach to cancer therapy.
    Journal of immunotherapy (Hagerstown, Md. : 1997), 1997, Volume: 20, Issue:3

    Many studies show defective immune responses in patients diagnosed with cancer. Most of the diverse nonspecific approaches used to stimulate the immune system to recognize and destroy abnormal tumor cells have limited clinical utility. Attempts to identify tumor-specific antigens and to improve the antigen presentation were equally disappointing. It appears that some of these failures can be explained by tumor-induced immunosuppression. A large number of cytokines, hormones, and other molecules secreted by tumors were demonstrated to have immunomodulating properties. The most extensively studied immunosuppressive molecules secreted by tumors are transforming growth factor-beta (TGF beta), interleukin 10 (IL-10), and prostaglandin E2 (PGE2). TGF beta in particular may play a key role in tumor-induced immunosuppression. It is the most potent immunosuppressor described to date, and it has been consistently isolated from variety of tumor cell lines and detected in plasma of tumor-bearing hosts. Level of TGF beta production by tumor cells correlates with their metastatic potential, and TGF beta neutralization not only prevents development of metastases, but also inhibits growth or completely eradicates tumors as diverse as breast cancer, melanoma, and malignant gliosarcoma in animal models. PGE2 may play significant role in early stages of tumor development, promoting the process of tumorigenesis in some tumors. Research on reversal of tumor-induced immunosuppression promises new, more powerful, and less toxic approaches to cancer therapy. Existence of molecule(s) consistently secreted by different types of tumors and responsible for tumor progression raises the possibility of a single, universal assay to monitor progression and recurrence in many malignancies, including those that currently do not have reliable plasma markers.

    Topics: Animals; Cytokines; Dinoprostone; Humans; Immune Tolerance; Immunotherapy; Interleukin-10; Neoplasms; Neoplasms, Experimental; Transforming Growth Factor beta

1997
Transforming growth factor-beta: the breaking open of a black box.
    BioEssays : news and reviews in molecular, cellular and developmental biology, 1997, Volume: 19, Issue:7

    Transforming growth factor-beta (TGF-beta) and its related proteins regulate broad aspects of body development, including cell proliferation, differentiation, apoptosis and gene expression, in various organisms. Deregulated TGF-beta function has been causally implicated in the generation of human fibrotic disorders and in tumor progression. Nevertheless, the molecular mechanisms of TGF-beta action remained essentially unknown until recently. Here, we discuss recent progress in our understanding of the mechanism of TGF-beta signal transduction with respect to the regulation of gene expression, the control of cell phenotype and the potential usage of TGF-beta for the treatment of human diseases.

    Topics: Animals; Cell Communication; Cell Cycle; Cell Differentiation; Gene Expression Regulation; Humans; Immunologic Factors; Multigene Family; Neoplasms; Organ Specificity; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta; Vertebrates

1997
The multiple roles of tumour stroma.
    Virchows Archiv : an international journal of pathology, 1997, Volume: 430, Issue:6

    Since the work of Judah Folkman in the 1970s demonstrating the importance of vascularization on tumour growth the many roles played by tumour stroma have been demonstrated. Vascular endothelial growth factor/vascular permeability factor appears to be the main in vivo inducer of both stromal blood vessels and other components of the tumour stroma. Its action is probably mediated through its strong and long-lasting effect on microvascular permeability leading to fibrin extravasation and organisation ("tumours are wounds that do not heal"). During tumour invasion, stromal fibroblasts participate in the degradation of the extracellular matrix (ECM) by secreting matrix degrading proteases as well as their downstream-activators. Stroma derived factors such as scatter factor/hepatocyte growth factor as well as interactions between neoplastic cells and the ECM can play a role in both tumour cell migration and proliferation. The ECM may also act as a reservoir for growth factors. A novel transcription factor encoded by the c-ets 1 proto-oncogene is likely to be involved in the transcriptional regulation of both tumour vascularization and invasion. This contribution summarizes recent developments in the tumour stroma field.

    Topics: Animals; Capillary Permeability; Cell Division; Extracellular Matrix; Humans; Neoplasm Invasiveness; Neoplasms; Proto-Oncogene Mas; Stromal Cells; Transforming Growth Factor beta

1997
Diet, apoptosis, and carcinogenesis.
    Advances in experimental medicine and biology, 1997, Volume: 422

    It is known that long-term withdrawal of choline from the diet induces hepatocellular carcinomas in animal models in the absence of known carcinogens. We hypothesize that a choline deficient diet (CD) alters the balance of cell growth and cell death in hepatocytes and thus promotes the survival of clones of cells capable of malignant transformation. When grown in CD medium (5 microM or 0 microM choline) CWSV-1 rat hepatocytes immortalized with SV40 large T-antigen underwent p53-independent apoptosis (terminal dUTP end-labeling of fragmented DNA; laddering of DNA in agarose gel). CWSV-1 cells which were adapted to survive in 5 microM choline acquired resistance to CD-induced apoptosis and were able to form hepatocellular carcinomas in nude mice. These adapted CWSV-1 cells express higher amounts of both the 32 kDa membrane-bound and 6 kDa mature form of TGF alpha compared to cells made acutely CD. Control (70 microM choline) and adapted cells, but not acutely deficient hepatocytes, could be induced to undergo apoptosis by neutralization of secreted TGF alpha. Protein tyrosine phosphorylation is known to protect against apoptosis. We found decreased EGF receptor tyrosine phosphorylation in acutely choline deficient CWSV-1 cells. TGF beta 1 is an important growth-regulator in the liver. CWSV-1 cells express TGF beta 1 receptors and this peptide induced cell detachment and death in control and acutely deficient cells. Hepatocytes adapted to survive in low choline were also resistant to TGF beta 1, although TGF beta 1 receptors and protein could be detected in the cytoplasm of these cells. The non-essential nutrient choline is important in maintaining plasma membrane structure and function, and in intracellular signaling. Our results indicate that acute withdrawal of choline induces p53-independent programmed cell death in hepatocytes, whereas cells adapted to survive in low choline are resistant to this form of apoptosis, as well as to cell death induced by TGF beta 1. Our results also suggest that CD may induce alterations (mutations?) in growth factor signaling pathways which may enhance cell survival and malignant transformation.

    Topics: Animals; Apoptosis; Choline; Choline Deficiency; Diet; Epidermal Growth Factor; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Suppressor Protein p53

1997
Transforming growth factor-beta signaling in epithelial cells.
    Pharmacology & therapeutics, 1997, Volume: 75, Issue:1

    Transforming growth factor (TGF)-beta is a potent growth suppressor of epithelial cells. Resistance to TGF-beta, however, occurs frequently in solid tumors of epithelial origin and contributes to the uncontrolled growth of these tumors. Although mutant receptor proteins contribute to TGF-beta insensitivity, deregulation of TGF-beta signaling cascades represents an equally important mechanism underlying TGF-beta resistance. Identification of abnormal regulation of signaling components in tumor epithelial cells will lead to the development of selective therapeutic approaches to repair the relevant signaling cascade(s) and reverse the growth anomaly. Within the past few years, great strides have been made in defining signaling pathways for TGF-beta. For example, our laboratory has demonstrated a direct correlation between TGF-beta-mediated growth inhibition of epithelial cells and activation of Ras and three members of the mitogen-activated protein kinase (MAPK) superfamily. The TGF-beta signaling events were sustained, dose-dependent, and absent in TGF-beta-resistant cells. Further, up-regulation of both p27Kip1 and p21Cip1, nuclear events important for the growth inhibitory effect of TGF-beta, are completely dependent upon the activation of Ras. However, Ras-independent pathways are also activated simultaneously with the Ras/MAPK pathways to mediate the final TGF-beta growth inhibitory outcome. One such pathway includes the SMAD signaling components that control TGF-beta-mediated gene transcription, currently under active study by a number of laboratories, including our own. Future efforts in this field will focus on defining the significance of these signaling proteins and pathways in mediating specific TGF-beta responses. Moreover, additional novel signaling proteins are sure to be identified.

    Topics: Alkyl and Aryl Transferases; Carrier Proteins; Cell Nucleus; Cytoplasm; DNA-Binding Proteins; Drosophila Proteins; Epithelial Cells; Heat-Shock Proteins; Humans; Insect Proteins; Neoplasms; Protein Sorting Signals; Receptors, Transforming Growth Factor beta; Signal Transduction; Tacrolimus Binding Proteins; Transforming Growth Factor beta

1997
TGF-beta receptor signaling.
    Biochimica et biophysica acta, 1997, Oct-24, Volume: 1333, Issue:2

    Topics: Animals; Antigens, CD; Cell Division; DNA-Binding Proteins; Drosophila; Drosophila Proteins; Endoglin; Models, Biological; Neoplasms; Phosphoric Monoester Hydrolases; Phosphotransferases; Proteoglycans; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Receptors, Tumor Necrosis Factor; Receptors, Tumor Necrosis Factor, Type I; Repressor Proteins; Signal Transduction; Transcription Factors; Transforming Growth Factor beta; Vascular Cell Adhesion Molecule-1

1997
Tumor suppressor genes in the TGF-beta signaling pathway?
    Nature medicine, 1996, Volume: 2, Issue:4

    Topics: Animals; Genes, Tumor Suppressor; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

1996
Elimination of transformed cells by normal cells: a novel concept for the control of carcinogenesis.
    Histology and histopathology, 1996, Volume: 11, Issue:1

    Control of transformed cells by neighbouring normal cells is known since the beginning of transformation studies in vitro. The classical explanation for this phenomenon is based on proliferation inhibition of transformed cells by normal cells. We extend this model by presenting data that show that TGF-beta-treated normal cells can eliminate transformed cells by induction of apoptosis. Both the TGF-beta-induced signal pathway in normal cells, leading to the production of a short-lived apoptosis-inducing factor, as well as the specific interaction of this factor with transformed cells depend on the action of reactive oxygen species. Sensitivity to induction of apoptosis seems to be a common feature associated with the transformed state, independent of the originally transforming principle. Therefore, tumor development should require either interference with the process of elimination or acquisition of resistance against it. We discuss experimental evidence for interfering substances, such as antioxidants, as well as for genetic systems that protect transformed cells from the negative effects of their cellular environment, such as Bcl-2 or papilloma viruses. These findings, as well as the general resistance of exvivo tumor cells against induction of apoptosis are in line with the novel model of control of tumor progression presented by us in this review.

    Topics: Animals; Apoptosis; Carcinogens; Cell Transformation, Neoplastic; Humans; Neoplasms; Transforming Growth Factor beta

1996
SPARC and the extracellular matrix: implications for cancer and wound repair.
    Current topics in microbiology and immunology, 1996, Volume: 213 ( Pt 1)

    Topics: Cell Adhesion; Cell Division; Extracellular Matrix; Gene Expression Regulation; Growth Substances; Metalloendopeptidases; Neoplasm Proteins; Neoplasms; Neovascularization, Pathologic; Osteonectin; Plasminogen Activator Inhibitor 1; Transforming Growth Factor beta; Wound Healing

1996
Tumor suppressor activity of the TGF-beta pathway in human cancers.
    Cytokine & growth factor reviews, 1996, Volume: 7, Issue:1

    The TGF-betas are a family of cytokines with antiproliferative activity on many cell types. Recent findings demonstrate that the TGF-beta receptor complex functions as a tumor suppressor gene in human malignancy. Somatic mutations of the type II subunit of the TGF-beta receptor (RII) have been demonstrated in several different tumor types. RII frameshift mutations within a short coding region polyadenine repeat are particularly characteristic of colon and gastric cancers that also demonstrate the phenotype of microsatellite instability (RER cancers). These and other mutations as in the type I receptor (RI) are associated with both loss of cell surface TGF beta receptors and with resistance of the cancer cells to TGF-beta-induced growth inhibition. Restoration of receptor expression by gene transfection reverses the transformed phenotype in cancer cells that lacked functional RII or RI. These receptors and, by implication, TGF-beta as well as its complete signalling pathway, are thus new and novel additions to the family of human tumor suppressor genes.

    Topics: Cell Division; Colonic Neoplasms; Genes, Tumor Suppressor; Humans; Mutation; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

1996
Transforming growth factor-beta: a general review.
    European cytokine network, 1996, Volume: 7, Issue:3

    Three isoforms of Transforming Growth Factor-beta (TGF-beta 1, beta 2 and beta 3) exist in mammals. They play critical roles in growth regulation and development. Each isoform is encoded by a unique gene on different chromosomes. All three of these growth factors are secreted by most cell types, generally in a latent form, requiring activation before they can exert biological activity. This activation of latent TGF-beta, which may involve plasmin, thrombospondin and possibly acidic microenvironments, appears to be a crucial regulatory step in controlling their effects. The TGF-betas possess three major activities: they inhibit proliferation of most cells, but can stimulate the growth of some mesenchymal cells; they exert immunosuppressive effects; and they enhance the formation of extracellular matrix. Two types of membrane receptors (type I and type II) possessing a serine/threonine kinase activity within their cytoplasmic domains are involved in signal transduction. Inhibition of growth by the TGF-betas stems from a blockage of the cell cycle in late G1 phase. Among the molecular participants concerned in G1-arrest are the Retinoblastoma (Rb) protein and members of the Cyclin/Cyclin-dependent kinase/Cyclin dependent kinase inhibitor families. In the intact organism the TGF-betas are involved in wound repair processes and in starting inflammatory reactions and then in their resolution. The latter effects of the TGF-betas derive in part from their chemotactic attraction of inflammatory cells and of fibroblasts. From gene knockout and from overexpression studies it has been shown that precise regulation of each isoform is essential for survival, at least in the long term. Several clinical applications for certain isoforms have already shown their efficacy and they have been implicated in numerous other pathological situations.

    Topics: Animals; Cell Differentiation; Cell Division; CHO Cells; Cricetinae; Humans; Inflammation; Mice; Mice, Knockout; Neoplasms; Rats; Receptors, Transforming Growth Factor beta; Recombinant Proteins; RNA, Messenger; Signal Transduction; Transforming Growth Factor beta; Wound Healing

1996
Cancer gets Mad: DPC4 and other TGFbeta pathway genes in human cancer.
    Biochimica et biophysica acta, 1996, Dec-09, Volume: 1288, Issue:3

    Topics: Animals; DNA-Binding Proteins; Drosophila; Genes, Tumor Suppressor; Humans; Neoplasms; Nerve Growth Factors; Smad Proteins; Smad1 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta; Xenopus Proteins

1996
EGR-1, the reluctant suppression factor: EGR-1 is known to function in the regulation of growth, differentiation, and also has significant tumor suppressor activity and a mechanism involving the induction of TGF-beta1 is postulated to account for this sup
    Critical reviews in oncogenesis, 1996, Volume: 7, Issue:1-2

    The transcription factor EGR-1 is a potential regulator of over 30 genes and plays a role in growth, development, and differentiation and, in addition, has significant transformation suppression activity. The regulatory properties are reviewed and a hypothesis for the transformation suppression activity is proposed. EGR-1 contains three "zinc-finger" motifs in the C-terminal portion of the molecule that constitute the DNA-binding domain and interact with the promoters by virtue of two classes of GC-rich elements: single GC-elements (GCEs) with the consensus 5'-T-G-C-G-T/g-G/A-G-G-C/a/t-G-G/T-3' and overlapping sites consisting of an Sp-1 binding site and the GCE consensus or close homolog of these sequences. The Wilm's tumor suppressor gene product WT1 interacts with the same GCE and, owing in part to four alternate splice products, interacts with a broader range of GC-rich elements with the consensus 5'-GNGNGGGNG-3' and 5'-TCCTCCTCCTCCTC-3'. WT1 commonly but not invariably acts as repressor of transcription, whereas EGR-1, in the absence of overlapping Sp-1 binding sequences, is often an activator. The well-known rapid response of the EGR-1 gene following mitogenic stimulation together with the occurrence of GCEs in the promoters of many growth factors and protooncogenes suggests a role of EGR-1 in growth. Moreover, EGR-1 is constitutively expressed in several viral-transformed systems. On the other hand, studies of model and human tumor lines reveal that EGR-1 has significant growth and transformation suppression roles. Recent studies show that this effect can be accounted for by the ability of EGR-1 to induce the expression and secretion of TGF-beta1, a potent growth suppressor of many cell types, by binding to a single GCE of the TGF-beta1 promoter. Although the effects of EGR- at overlapping Sp1/EGR-1 DNA binding sites are not predictable, known cases fall into two loose groups. Sp1 is usually activating and increasing concentrations of EGR-1 lead to displacement that results in either inhibition of transactivation or EGR-1-dependent transactivation. Moreover, recent studies suggest that displaced Sp1 binds to and activates the endogenous Egr-1 gene, thereby leading to "facilitated inhibition" of Sp1 function by the resulting increased EGR-1. This effect may augment the growth suppressive function of EGR-1 based on induction of TGF-beta1.

    Topics: Animals; Cell Differentiation; Cell Division; Cell Transformation, Viral; DNA-Binding Proteins; Early Growth Response Protein 1; Humans; Immediate-Early Proteins; Neoplasms; Transcription Factors; Transforming Growth Factor beta

1996
[Transforming growth factors and cancer].
    Sheng li ke xue jin zhan [Progress in physiology], 1996, Volume: 27, Issue:4

    Topics: Animals; Humans; Neoplasm Invasiveness; Neoplasms; Prognosis; Transforming Growth Factor alpha; Transforming Growth Factor beta

1996
Signaling by the transforming growth factor-beta receptors.
    Biochimica et biophysica acta, 1995, Dec-18, Volume: 1242, Issue:2

    Topics: Animals; Antigens, CD; Cell Cycle; Endoglin; Humans; Neoplasms; Protein Serine-Threonine Kinases; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Signal Transduction; Telangiectasia, Hereditary Hemorrhagic; Transforming Growth Factor beta; Vascular Cell Adhesion Molecule-1

1995
Transforming growth factor beta: relevance to radiotherapy.
    Clinical oncology (Royal College of Radiologists (Great Britain)), 1995, Volume: 7, Issue:3

    Topics: Animals; Fibrosis; Humans; Mice; Mice, Transgenic; Neoplasms; Radiotherapy; Transforming Growth Factor beta

1995
Transforming growth factor beta and cancer.
    Cancer treatment reviews, 1995, Volume: 21, Issue:4

    Topics: Breast Neoplasms; Cell Adhesion; Cell Adhesion Molecules; Cell Cycle; Cell Division; Extracellular Matrix; Female; Gastrointestinal Neoplasms; Gene Expression Regulation; Gene Expression Regulation, Neoplastic; Humans; Lung Neoplasms; Male; Neoplasms; Prostatic Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

1995
Nutrition, immunology and cancer: an overview.
    Advances in experimental medicine and biology, 1995, Volume: 369

    There is epidemiological, laboratory and some clinical evidence that certain dietary factors play a role in either promoting or inhibiting cancer development. An understanding of the mechanisms whereby specific nutrients are having effects in the promotion or prevention of cancer is beginning to take shape. Research into nutrient effects on the expression of specific genes, especially cytokine and cytokine receptor expression, will help increase our basic knowledge of cancer biology. Since cytokines can either enhance or suppress both immune defense and cancer growth, increased understanding of nutrient effects on the cytokine network will be beneficial. The regulation of specific gene expression by specific nutrients, indeed, identifies a major frontier for present and future nutritional biologists.

    Topics: Allergy and Immunology; Cell Division; Cytokines; Gene Expression Regulation; Humans; Neoplasms; Nutritional Physiological Phenomena; Transforming Growth Factor beta; Tumor Cells, Cultured; Vitamin E

1995
[Transforming growth factor-beta. An interesting candidate for clinical use].
    Lakartidningen, 1995, Apr-12, Volume: 92, Issue:15

    Transforming growth factor-beta (TGF-beta) is the prototype of a large family of growth regulatory factors affecting the growth and differentiation of many cell types. Their action is mediated by complex formation of type I and type II receptors, both of which are serine/threonine kinases. TGF-beta inhibitors inhibit the growth of most cell types are potent immunosuppressive agents. They also stimulate the formation of connective tissue, thus promoting wound healing. TGF-beta and TGF-beta antagonists may have potential clinical uses in the treatment of various disorders.

    Topics: Fibrosis; Humans; Immune System; Immunosuppressive Agents; Neoplasms; Transforming Growth Factor beta; Wound Healing

1995
Role of active cell death (apoptosis) in multi-stage carcinogenesis.
    Toxicology letters, 1995, Volume: 82-83

    Active cell death is a genetically encoded self-destruction of a cell. There occur morphologically different types of active cell death, e.g. apoptosis in the liver or autophagic cell death in human mammary carcinoma cells after tamoxifen treatment (Pre)neoplastic lesions in rat liver exhibit enhanced rates of apoptosis, which tend to increase with increasing malignancy. Tumor promoters and non-genotoxic carcinogens inhibit active cell death, thereby increasing the accumulation of (pre)neoplastic cells and accelerating the development of cancer. On the other hand promoter withdrawal, fasting or application of negative growth signals such as transforming growth factor beta 1 (TGF beta 1) enhance apoptosis and can lead to selective regression of preneoplastic lesions or tumors.

    Topics: Animals; Apoptosis; Breast Neoplasms; Humans; Liver Neoplasms; Neoplasms; Rats; Transforming Growth Factor beta

1995
The tumor-promoting effect of wounding: a possible role for TGF-beta-induced stromal alterations.
    Critical reviews in oncogenesis, 1994, Volume: 5, Issue:2-3

    From clinical, chemical carcinogenesis and transgenic animal studies, it is evident that wounding has a tumor-promoting effect. We discuss the role of TGF-beta (with special emphasis on TGF-beta 1) in this process and suggest that stromal alterations during wound healing, induced by TGF-beta, can be an important determinant of tumor growth. A tumor and a wound both require similar stromal microenvironments. Thus, a chemically initiated or an oncogene-expressing cell could be complemented to grow into a tumor if it finds itself in a hospitable wound-healing stroma.

    Topics: Animals; Cell Transformation, Neoplastic; Cocarcinogenesis; Humans; Mice; Neoplasms; Transforming Growth Factor beta; Wound Healing

1994
Transforming growth factor-beta: expression in normal and pathological conditions.
    Hormone research, 1994, Volume: 42, Issue:1-2

    Expression of the various isoforms of transforming growth factor-beta (TGF-beta) is differentially controlled both in vivo and in vitro. Characterization of the molecular mechanisms governing expression of TGF-beta isoforms now provides a basis for understanding the selective regulation of expression of the TGF-beta s by a variety of factors including oncogenes and tumor suppressor genes. In addition to transcriptional control, data suggest that expression of TGF-beta s is also regulated posttranscriptionally. Regulation of TGF-beta s by steroids and retinoids appears to involve predominantly posttranscriptional mechanisms. Identification of these mechanisms may contribute to the understanding of the regulatory events controlling cellular proliferation and differentiation by TGF-beta s and steroids/retinoids.

    Topics: Animals; Gene Expression; Genes, Tumor Suppressor; Humans; Isomerism; Neoplasms; Promoter Regions, Genetic; Protein Processing, Post-Translational; Reference Values; Transforming Growth Factor beta

1994
Growth factor balance and tumor progression.
    Current opinion in oncology, 1994, Volume: 6, Issue:1

    The role of balance of negative and positive autocrine growth factors in malignant progression is reviewed with an emphasis on transforming growth factor alpha (TGF-alpha) as a stimulating factor and transforming growth factor beta (TGF-beta) as an inhibiting factor. Evidence suggests that in normal cells TGF-alpha is down regulated in non-dividing or quiescent states in vitro. Tumor cells which have early stage characteristics as represented by poor clonal growth and poor tumorigenicity in athymic mice also show repression of TGF-alpha in non-dividing states. Progression of this phenotype is induced by uncontrolled low level expression of TGF-alpha by transfection with a constitutive expression vector for the polypeptide. Transfection of the unprogressed phenotype with a constitutive anti-sense vector for TGF-beta, also leads to tumor progression by repressing the autocrine negative TGF-beta activity normally expressed by these cells. Both the upregulation of TGF-alpha and the repression of TGF-beta generated in vivo progression without changing growth rates in vitro. Instead, clonality and ability to reenter the cell cycle from quiescence were increased. Thus, it is concluded that an autocrine balance of positive and negative factors is important for maintaining controlled re-entry into dividing states from non-dividing states and that disruption of this balance leads to malignant progression characterized by greater independence of the malignant cells from the control of exogenous growth factors.

    Topics: Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Transforming Growth Factor alpha; Transforming Growth Factor beta

1994
Downregulation in vivo of the invasion-suppressor molecule E-cadherin in experimental and clinical cancer.
    Princess Takamatsu symposia, 1994, Volume: 24

    The invasion-suppressor molecule E-cadherin (E-CAD) can be regulated at multiple levels: synthesis, processing and stability of mRNA; synthesis, processing and stability of protein; localization and posttranslational modification of protein; binding to catenins (E-CAD-associated proteins); and size and charge of cell surface glycosaminoglycans. Loss of E-CAD antigen and of E-CAD function in vivo has been observed with cell lines that homogeneously expressed functional E-CAD in vitro. These observations led to the idea that factors in the host may downmodulate E-CAD on the cancer cells, thereby promoting cell invasion. Nude mouse cancers that were homogeneously E-CAD-positive and noninvasive in vitro, formed by epithelioid MDCK or NMuMG cells, stained heterogeneously for E-CAD; such cancers were invasive and metastatic. The in vivo downmodulation appeared to be transient. Ex vivo cultures from primary cancers, as well as from metastases, produced homogeneously E-CAD-positive and noninvasive cells. Downmodulation did not occur when cells were micro-encapsulated and then implanted in the mouse, suggesting a role for immediate cancer cell-host cell contact. Similar in vitro/in vivo/ex vivo experiments with mouse MO4 fibrosarcoma cells, transfected with E-CAD cDNA under the control of a b-actin promotor, showed downregulation at the transcriptional or mRNA stability level. This downregulation was rapidly reversible upon ex vivo culture of the tumor cells. TGF-bl and IGF-I were found, respectively, to downregulate and upregulate the expression or the function of E-CAD. We speculate that IGF-1 restores the function of E-CAD through interaction of the IGF-I tyrosine kinase receptor with the catenin-actin cytoskeletal complex. In human cancers, immunohistochemistry has revealed changes in E-cadherin that agree with the experimental data on transient downmodulation of the invasion-suppressor function of E-cadherin by host factors.

    Topics: alpha Catenin; Animals; beta Catenin; Cadherins; Cytoskeletal Proteins; Gene Expression Regulation, Neoplastic; Humans; Insulin-Like Growth Factor I; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasm Proteins; Neoplasms; Neoplasms, Experimental; Phosphorylation; Protein Kinases; Protein Processing, Post-Translational; RNA, Messenger; RNA, Neoplasm; Trans-Activators; Transforming Growth Factor beta; Tumor Cells, Cultured

1994
The role of beta 1 integrins in tumors.
    Seminars in cancer biology, 1993, Volume: 4, Issue:5

    Members of the beta 1 subfamily of integrins contribute to cell adhesion, cytoskeletal organization and signal transduction processes. In some transformed cell lines and tumors, a correlation has been established between the level of expression of the alpha 5/beta 1 fibronectin receptor and neoplastic behavior. In other instances, normal and neoplastic tissue differ in beta 1 integrin expression or sub-cellular distribution. The level of expression of beta 1 integrins in tumor cells may affect tumor growth properties in several ways, including: (a) effects on anchorage dependence of growth; (b) direct signaling processes; (c) organization of the extracellular matrix and presentation of matrix bound growth factors; (d) effects on the functions of host defense cells. Thus the interplay between integrin expression and tumor behavior is complex and might be viewed as a series of interactive feedback loops rather than in terms of a straightforward cause and effect relationship.

    Topics: Animals; Cell Transformation, Neoplastic; Humans; Integrin beta1; Integrins; Neoplasm Metastasis; Neoplasms; Transforming Growth Factor beta

1993
Transforming growth factor beta and fibroblast growth factor as promoters of tumor progression to malignancy.
    Critical reviews in oncogenesis, 1993, Volume: 4, Issue:5

    Polypeptide growth factors are a diverse group of biological regulators. Because they are fundamentally involved in the cellular processes that are important for transformation and progression to malignancy, alterations in growth factor control and in their signal pathways are often observed in tumor cells. In this review, we consider the participation of growth factors and the mechanisms by which they effect tumor progression, using as examples members of the transforming growth factor beta (TGF-beta) and fibroblast growth factor (FGF) families. We explore the hypothesis that although abrogation of TGF-beta negative growth regulation is necessary for transformation, in the later stages of tumor progression, TGF-beta plays a direct role in the enhancement of invasion and metastasis as an autocrine stimulator of these processes. In addition, we present evidence that demonstrates both the potential and the importance of members of the FGF family in transformation and induction of metastasis. Several models of growth factor regulation of malignancy are presented in which we demonstrate (1) a link between TGF-beta 1 mitogenic stimulation of malignant cells and alterations in the expression of ribonucleotide reductase, a key rate-limiting step in the synthesis of DNA and in cell proliferation; (2) autocrine and/or intracrine FGF mitogenic stimulation of malignant cell proliferation and metastasis; and (3) autocrine TGF-beta regulation of malignant cell locomotion and invasion through elevated proteolytic activity and increased synthesis of hyaluronan and RHAMM, a novel hyaluronan cell surface receptor.

    Topics: Animals; Cell Division; Cell Movement; Fibroblast Growth Factors; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neoplasms, Experimental; Transforming Growth Factor beta

1993
Resistance to inhibition of cell growth by transforming growth factor-beta and its role in oncogenesis.
    Critical reviews in oncogenesis, 1993, Volume: 4, Issue:5

    Transforming growth factor-beta (TGF beta) was first described as a soluble factor capable of transforming normal rat kidney cells in the presence of transforming growth factor-alpha (TGF alpha) or epidermal growth factor (EGF). TGF beta is now known to have a wide spectrum of effects, depending on cell type, culture conditions, and the presence of other growth factors. Whereas picomolar concentrations of TGF beta completely inhibit the growth of most nonmalignant epithelial cells and early hematopoietic progenitor cells, a review of the published English language literature reveals that virtually all malignant cells of epithelial or hematopoietic origin are refractory to the antiproliferative effects of TGF beta in vitro. When compared with normal epithelial cells, malignant carcinoma cells require significantly higher doses of TGF beta to achieve a similar degree of growth suppression or they are only partially suppressed by large doses of TGF beta. This loss of sensitivity to TGF beta is associated with the development of invasive properties. More than one genetic event appears to be required to convert a sensitive cell into one that is completely refractory to TGF beta. In conclusion, resistance to the growth-inhibiting effect of TGF beta appears to be a late but consistent event in the process of malignant transformation in vitro. Identification of the genetic lesions that result in the loss of response to TGF beta may lead to the design of new therapeutic modalities to arrest tumor growth.

    Topics: Animals; Carcinoma; Cell Division; Growth Inhibitors; Humans; Neoplasms; Neoplasms, Experimental; Transforming Growth Factor beta

1993
Transforming growth factor beta and the cell surface in tumor progression.
    Cancer metastasis reviews, 1993, Volume: 12, Issue:3-4

    Type 1 transforming growth beta (TGF-beta 1) is a multifunctional regulator of cellular differentiation, motility and growth. It is capable of inhibiting or stimulating these processes depending on cell type, cell density, culture conditions and TGF-beta 1 concentration. TGF-beta 1 regulates growth, in part, by inducing the expression and secretion of various types of collagen, which participate in the control of cell adhesion and migration, as well as growth. TGF-beta 1 also regulates cell growth by controlling the response to epidermal growth factor (EGF) and other growth factors, in ways that can either decrease or increase their growth-promoting effects. Alterations in both negative and positive growth responses to TGF-beta 1 play important roles in tumor progression. Loss of sensitivity to growth inhibition by TGF-beta 1 can occur as a result of decreased expression of collagen. Acquisition of sensitivity to growth stimulation, and autocrine transformation by TGF-beta 1, are associated with aberrant EGF receptor regulation. Aberrant growth factor receptor regulation by TGF-beta 1 may be mediated by a protein kinase C (PKC)-dependent pathway which inhibits degradation of growth factor receptor/ligand complexes. The evidence reviewed is consistent with a minimal two-step mechanism for autocrine transformation, which involves production of growth factor and enhanced cellular response as a result of aberrant membrane traffic. Defects in membrane traffic regulation may provide an explanation for common alterations in tumor cell response to both multiple growth inhibitors and growth stimulators, and may also suggest novel approaches to cancer chemotherapy.

    Topics: Animals; Cell Division; Cell Membrane; Cell Transformation, Neoplastic; Epithelial Cells; Fibroblasts; Humans; Neoplasms; Transforming Growth Factor beta

1993
Antiinflammatory effects of neoplasms.
    Research in immunology, 1993, Volume: 144, Issue:4

    Topics: Animals; Anti-Inflammatory Agents; Humans; L-Lactate Dehydrogenase; Macrophages; Neoplasm Proteins; Neoplasms; Retroviridae Proteins; Transforming Growth Factor beta; Viral Envelope Proteins

1993
Development of resistance mechanisms to the growth-inhibitory effects of transforming growth factor-beta during tumor progression.
    Current opinion in oncology, 1993, Volume: 5, Issue:1

    Members of the transforming growth factor-beta family, especially transforming growth factor-beta type 1, are among the most potent growth-inhibitory factors for epithelial, lymphohematopoietic, and neuroectodermal cells. Resistance to transforming growth factor-beta-mediated growth inhibition is frequently observed in cancers derived from these cell types. We review two important aspects of cancer cell resistance to transforming growth factor-beta: 1) It occurs progressively during the multistep process of tumor progression, and 2) it may encompass a potentially wide range of mechanisms involving transforming growth factor-beta-receptor alterations and cell-signaling defects. Stepwise increases in resistance to the growth-inhibitory action of transforming growth factor-beta may therefore sometimes be attributed to not one but several defects that are involved in transforming growth factor-beta activation, binding, and signaling. These factors accumulate within expanding tumor subclones during disease progression.

    Topics: Cell Division; Drug Resistance; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

1993
Physiological actions and clinical applications of transforming growth factor-beta (TGF-beta).
    Growth factors (Chur, Switzerland), 1993, Volume: 8, Issue:1

    Topics: Animals; Bone and Bones; Humans; Immunity; Inflammation; Ischemia; Neoplasms; Transforming Growth Factor beta

1993
Active cell death in hormone-dependent tissues.
    Cancer metastasis reviews, 1992, Volume: 11, Issue:2

    Active cell death (ACD) in hormone-dependent tissues such as the prostate and mammary gland is readily induced by hormone ablation and by treatment with anti-androgens or anti-estrogens, calcium channel agonists and TGF beta. These agents induce a variety of genes within the hormone-dependent epithelial cells including TRPM-2, transglutaminase, poly(ADP-ribose) polymerase, Hsp27 and several other unidentified genes. Not all epithelial cells in the glands are equally sensitive to the induction of ACD. In the prostate, the secretory epithelial cells that are sensitive to hormone ablation are localized in the distal region of the prostatic ducts, and are in direct contact with the neighboring stroma. In contrast, the epithelial cells in the proximal regions of the ducts are more resistant to hormone ablation, probably because the permissive effects of the stroma are attenuated by the presence of the basal epithelial cells, which are intercalated between the epithelium and stroma. The underlying biology of ACD in prostate and mammary glands, and its relevance to hormone resistance, is discussed in this review.

    Topics: Animals; Apoptosis; Breast; Calcium; Cell Communication; Clusterin; Extracellular Matrix; Female; Gene Expression; Glycoproteins; Heat-Shock Proteins; Humans; Male; Molecular Chaperones; Neoplasms; Poly(ADP-ribose) Polymerases; Prostate; Transforming Growth Factor beta; Transglutaminases

1992
Programmed cell death and its protective role with particular reference to apoptosis.
    Toxicology letters, 1992, Volume: 64-65 Spec No

    Apoptosis is a type of programmed cell death involved in growth control of tissues. It is considered as a cellular suicide functionally opposite to mitosis. It may serve to remove "unwanted" damaged or dangerous, e.g. precancerous, cells. Chemical compounds can interfere with the regulatory network which controls apoptosis and can thereby stimulate or prevent cell death. Both induction or inhibition of apoptosis may result in various diseases such as of the immune system, malformation or tumor development. The protective role of apoptosis against carcinogenesis is described in some detail. Tumor formation seems to occur through several stages, namely initiation, promotion, progression, and involves formation and growth of premalignant cell populations. At least in some model systems initiated cells and premalignant cell populations have been found to exhibit enhanced cell replication, but also enhanced apoptotic activity as compared to the normal tissue. Therefore, initiated cells may be eliminated by apoptosis. Tumor promoters can inhibit apoptosis in putative preneoplastic cells and thereby accelerate tumor development. Furthermore, in hormone-dependent cancers malignant cells may undergo massive apoptosis in response to hormone withdrawal or antihormone treatment. Finally, the regulation of apoptosis will be addressed. Our results suggest that transforming growth factor beta 1, a negative regulator of epithelial tissue growth, is a signal inducing apoptosis of liver cells.

    Topics: Apoptosis; Humans; Neoplasms; Transforming Growth Factor beta

1992
Growth hormones and factors in surgical patients.
    Advances in surgery, 1992, Volume: 25

    Topics: Animals; Clinical Trials as Topic; Colony-Stimulating Factors; Erythropoietin; Fibroblast Growth Factors; Growth Hormone; Growth Substances; Humans; Insulin-Like Growth Factor I; Interleukins; Neoplasms; Platelet-Derived Growth Factor; Recombinant Proteins; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Wound Healing

1992
The role of TGF-beta s in mammalian development and neoplasia.
    Molecular reproduction and development, 1992, Volume: 32, Issue:2

    To date, three mammalian TGF-beta isoforms have been identified, each encoded by different genetic loci. Through each is very similar in primary amino acid structure, there are clear differences both in the mature bioactive peptide region and in the latency-associated peptide, which could potentially confer differential biological specificity. As one route to investigate differential biological function in vivo we have used gene specific probes for in situ hybridization studies to examine the distribution of RNA transcripts during mammalian embryogenesis. Mouse embryos from 6 to 14.5 gestational age and human embryos from 32 to 57 days post-fertilization have been probed. A general conclusion from these studies is that each TGF-beta gene has a distinct, through overlapping, pattern of transcript distribution and that this pattern, in most cases, is conserved between mouse and man. We have focused on the biological function the TGF-betas play in certain epithelia and in cardiogenesis, which will be discussed in this presentation.

    Topics: Animals; Embryonic and Fetal Development; Epithelium; Heart; Humans; Neoplasms; Skin Neoplasms; Skin Physiological Phenomena; Transforming Growth Factor beta

1992
Transforming growth factor-beta.
    Cancer surveys, 1992, Volume: 12

    This chapter has described some of the most salient features of the biology of the TGF-beta s. The TGF-beta s are of great interest as growth inhibitors, regulators of cell phenotype and regulators of cell adhesion. The various TGF-beta isoforms are highly conserved and display a complex pattern of interactions with multiple membrane receptor components. Activation of these receptors leads to inhibition of epithelial cell proliferation by a mechanism that may involve proteins related to the growth suppressor, RB. TGF-beta receptors are also coupled to mechanisms that control expression of differentiation commitment genes and differentiated cell functions. TGF-beta can affect cell proliferation and differentiation through indirect mechanisms involving regulation of expression of cytokines, extracellular matrix molecules and their respective receptors. These responses strongly influence the growth and phenotype of an array of cell types. Excess or reduced TGF-beta activity may contribute to the pathogenesis of certain fibrotic disorders and certain hyperproliferative disorders including cancer, respectively.

    Topics: Cell Differentiation; Cell Division; Growth Inhibitors; Humans; Neoplasms; Protein Binding; Protein Kinases; Retinoblastoma Protein; Signal Transduction; Transforming Growth Factor beta

1992
Transforming growth factor-beta and the immune system.
    Progress in growth factor research, 1991, Volume: 3, Issue:2

    It is now apparent that the transforming growth factor beta (TGF-beta) family of proteins has potent immunoregulatory properties ranging from effects on the growth and differentiation of primitive stem cells to the differentiated functions of immune effector cells. Although most reports have described the immunosuppressive activities of TGF-beta, recent evidence supports the concept that TGF-beta can have both inhibitory and stimulatory actions on these systems. Recently, it has been found that TGF-beta can have autocrine as well as paracrine effects on the immune system, indicating that immune cells can activate the inactive secreted form of TGF-beta. Furthermore, TGF-beta has differential intracellular effects on cell surface receptor modulation, tyrosine phosphorylation, and cytokine gene transcription as well as cell-mediated cytotoxicity. Importantly, the administration of TGF-beta has proven beneficial in several animal disease models such as septic shock, allograft rejection, and autoimmunity. Moreover, the increased levels of TGF-beta found in several disease states associated with immunosuppression such as different forms of malignancy, chronic degenerative diseases, and AIDS implicate the involvement of TGF-beta in the pathogenesis of some diseases. Ultimately, well designed clinical trials will determine whether the exciting potential of TGF-beta can be used to treat or prevent disease.

    Topics: Animals; Autoimmunity; Humans; Immune Tolerance; Immunity; Inflammation; Lymphoma; Neoplasms; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Retroviridae Infections; Transforming Growth Factor beta

1991
Polypeptide growth factors in the regulation of human tumor cell proliferation.
    Current opinion in oncology, 1991, Volume: 3, Issue:1

    The capacity of growth factors to activate receptors through autocrine and paracrine pathways continues to be a major focus of cancer biology research. This review of growth factors for solid tumor cells complements our summary of hematopoietic growth factors.

    Topics: Amphiregulin; Cell Differentiation; Cell Division; EGF Family of Proteins; Epidermal Growth Factor; ErbB Receptors; Glycoproteins; Growth Substances; Humans; Intercellular Signaling Peptides and Proteins; Neoplasms; Platelet-Derived Growth Factor; Proto-Oncogene Proteins; Receptor, ErbB-2; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Sequence Homology, Nucleic Acid; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Cells, Cultured

1991
Interactions of retinoids and transforming growth factor-beta in regulation of cell differentiation and proliferation.
    Molecular endocrinology (Baltimore, Md.), 1991, Volume: 5, Issue:1

    Topics: Animals; Cell Differentiation; Cell Division; Genes, Suppressor; Humans; Neoplasms; Oncogenes; Retinoids; Transforming Growth Factor beta

1991
Suppression of carcinogenesis: a role for TGF-beta and related molecules in prevention of cancer.
    Immunology series, 1990, Volume: 51

    Topics: Animals; Cell Communication; Cell Differentiation; Cell Division; Cells, Cultured; Depression, Chemical; Epithelium; Extracellular Matrix; Gene Expression Regulation; Genes, Retinoblastoma; Hematopoietic Stem Cells; Humans; Intercellular Junctions; Interphase; Mice; Neoplasms; Precancerous Conditions; Retinoids; Steroids; Transforming Growth Factor beta

1990
[Transforming growth factor (TGF)-beta].
    Sheng li ke xue jin zhan [Progress in physiology], 1990, Volume: 21, Issue:4

    Topics: Animals; Humans; Neoplasms; Transforming Growth Factor beta

1990

Trials

7 trial(s) available for transforming-growth-factor-beta and Neoplasms

ArticleYear
Phase 1 first-in-human study of dalutrafusp alfa, an anti-CD73-TGF-β-trap bifunctional antibody, in patients with advanced solid tumors.
    Journal for immunotherapy of cancer, 2023, Volume: 11, Issue:2

    Cluster of differentiation (CD)73-adenosine and transforming growth factor (TGF)-β pathways are involved in abrogated antitumor immune responses and can lead to protumor conditions. This Phase 1 study (NCT03954704) evaluated the safety, pharmacokinetics, pharmacodynamics, and efficacy of dalutrafusp alfa (also known as GS-1423 and AGEN1423), a bifunctional, humanized, aglycosylated immunoglobulin G1 kappa antibody that selectively inhibits CD73-adenosine production and neutralizes active TGF-β signaling in patients with advanced solid tumors.. Dose escalation started with an accelerated titration followed by a 3+3 design. Patients received dalutrafusp alfa (0.3, 1, 3, 10, 20, 30, or 45 mg/kg) intravenously every 2 weeks (Q2W) up to 1 year or until progressive disease (PD) or unacceptable toxicity.. In total, 21/22 patients received at least one dose of dalutrafusp alfa. The median number of dalutrafusp alfa doses administered was 3 (range 1-14). All patients had at least one adverse event (AE), most commonly fatigue (47.6%), nausea (33.3%), diarrhea (28.6%), and vomiting (28.6%). Nine (42.9%) patients had a Grade 3 or 4 AE; two had Grade 5 AEs of pulmonary embolism and PD, both unrelated to dalutrafusp alfa. Target-mediated drug disposition appears to be saturated at dalutrafusp alfa doses above 20 mg/kg. Complete CD73 target occupancy on B cells and CD8+ T cells was observed, and TGF-β 1/2/3 levels were undetectable at dalutrafusp alfa doses of 20 mg/kg and higher. Free soluble (s)CD73 levels and sCD73 activity increased with dalutrafusp alfa treatment. Seventeen patients reached the first response assessment, with complete response, partial response, stable disease, and PD in 0, 1 (4.8%), 7 (33.3%), and 9 (42.9%) patients, respectively.. Dalutrafusp alfa doses up to 45 mg/kg Q2W were well tolerated in patients with advanced solid tumors. Additional evaluation of dalutrafusp alfa could further elucidate the clinical utility of targeting CD73-adenosine and TGF-β pathways in oncology.

    Topics: Antibodies, Bispecific; Antibodies, Monoclonal, Humanized; Humans; Immunoglobulin G; Neoplasms; Transforming Growth Factor beta; Treatment Outcome

2023
Bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in patients with human papillomavirus-associated malignancies.
    Journal for immunotherapy of cancer, 2020, Volume: 8, Issue:2

    Bintrafusp alfa is a first-in-class bifunctional fusion protein composed of the extracellular domain of transforming growth factor (TGF)-βRII (a TGF-β 'trap') fused to a human IgG1 mAb blocking programmed cell death ligand 1. This is the largest analysis of patients with advanced, pretreated human papillomavirus (HPV)-associated malignancies treated with bintrafusp alfa.. In these phase 1 (NCT02517398) and phase 2 trials (NCT03427411), 59 patients with advanced, pretreated, checkpoint inhibitor-naive HPV-associated cancers received bintrafusp alfa intravenously every 2 weeks until progressive disease, unacceptable toxicity, or withdrawal. Primary endpoint was best overall response per Response Evaluation Criteria in Solid Tumors (RECIST) V.1.1; other endpoints included safety.. As of April 17, 2019 (phase 1), and October 4, 2019 (phase 2), the confirmed objective response rate per RECIST V.1.1 in the checkpoint inhibitor-naive, full-analysis population was 30.5% (95% CI, 19.2% to 43.9%; five complete responses); eight patients had stable disease (disease control rate, 44.1% (95% CI, 31.2% to 57.6%)). In addition, three patients experienced a delayed partial response after initial disease progression, for a total clinical response rate of 35.6% (95% CI, 23.6% to 49.1%). An additional patient with vulvar cancer had an unconfirmed response. Forty-nine patients (83.1%) experienced treatment-related adverse events, which were grade 3/4 in 16 patients (27.1%). No treatment-related deaths occurred.. Bintrafusp alfa showed clinical activity and manageable safety and is a promising treatment in HPV-associated cancers. These findings support further investigation of bintrafusp alfa in patients with advanced, pretreated HPV-associated cancers.

    Topics: B7-H1 Antigen; Female; Humans; Male; Middle Aged; Neoplasms; Papillomaviridae; Papillomavirus Infections; Transforming Growth Factor beta

2020
Safety, Antitumor Activity, and Immune Activation of Pegylated Recombinant Human Interleukin-10 (AM0010) in Patients With Advanced Solid Tumors.
    Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 2016, 10-10, Volume: 34, Issue:29

    Purpose Interleukin-10 (IL-10) stimulates the expansion and cytotoxicity of tumor-infiltrating CD8+ T cells and inhibits inflammatory CD4+ T cells. Pegylation prolongs the serum concentration of IL-10 without changing the immunologic profile. This phase I study sought to determine the safety and antitumor activity of AM0010. Patients and Methods Patients with selected advanced solid tumors were treated with AM0010 in a dose-escalation study, which was followed by a renal cell cancer (RCC) dose-expansion cohort. AM0010 was self-administered subcutaneously at doses of 1 to 40 μg/kg once per day. Primary end points were safety and tolerability; clinical activity and immune activation were secondary end points. Results In the dose-escalation and -expansion cohorts, 33 and 18 patients, respectively, were treated with daily subcutaneous injection of AM0010. AM0010 was tolerated in a heavily pretreated patient population. Treatment-related adverse events (AEs) included anemia, fatigue, thrombocytopenia, fever, and injection site reactions. Grade 3 to 4 nonhematopoietic treatment-related AEs, including rash (n = 2) and transaminitis (n = 1), were observed in five of 33 patients. Grade 3 to 4 anemia or thrombocytopenia was observed in five patients. Most treatment-related AEs were transient or reversible. AM0010 led to systemic immune activation with elevated immune-stimulatory cytokines and reduced transforming growth factor beta in the serum. Partial responses were observed in one patient with uveal melanoma and four of 15 evaluable patients with RCC treated at 20 μg/kg (overall response rate, 27%). Prolonged stable disease of at least 4 months was observed in four patients, including one with colorectal cancer with disease stabilization for 20 months. Conclusion AM0010 has an acceptable toxicity profile with early evidence of antitumor activity, particularly in RCC. These data support the further evaluation of AM0010 both alone and in combination with other immune therapies and chemotherapies.

    Topics: Adult; Aged; Aged, 80 and over; Anemia; Carcinoma, Renal Cell; Cytokines; Drug Eruptions; Exanthema; Fatigue; Female; Fever; Humans; Injections, Subcutaneous; Interferon-gamma; Interleukin-10; Interleukin-4; Interleukin-8; Kidney Neoplasms; Male; Melanoma; Middle Aged; Neoplasms; Polyethylene Glycols; Recombinant Proteins; Thrombocytopenia; Transforming Growth Factor beta; Uveal Neoplasms; Young Adult

2016
Vitamin D favorably alters the cancer promoting prostaglandin cascade.
    Anticancer research, 2013, Volume: 33, Issue:9

    Preclinical studies suggest that 1,25-dihydroxyvitamin D [1,25(OH)2D] and celecoxib inhibit prostaglandins (PGs) associated with cancer through different mechanisms. We determined if there was synergy in their use.. A total of 36 healthy women received daily for one month/menstrual cycle: placebo, 400 international units (IU) vitamin D-3, 2,000 IU vitamin D-3, or 2,000 IU vitamin D-3 plus 400 mg celecoxib. Serum and nipple aspirate fluid (NAF) were analyzed for PGE2 and transforming growth factor (TGF)β1 and -2; serum for 25(OH)D (total, -D-2, -D-3), plasma for celecoxib; and mammary duct RNA for cyclooxygenase (COX)2.. 25(OH)D-3 increased (p<0.01) only in the groups receiving 2,000 IU vitamin D-3. PGE2 decreased in the breast (p=0.01) only after receiving 2,000 IU vitamin D-3; 2,000 IU vitamin D-3 alone was more effective in decreasing PGE2 than 2,000 IU vitamin D-3 plus celecoxib (p=0.018). COX2 expression decreased only in the breasts of women taking 2,000 IU vitamin D-3. Change in circulating 25(OH)D-3 correlated with change in TGFβ2 in the breast.. Vitamin D-3 reduces the PG cascade and increases TGFβ2 in a dose-dependent fashion. Adding celecoxib did not provide synergy.

    Topics: Adult; Celecoxib; Cholecalciferol; Cyclooxygenase 2; Dinoprostone; Double-Blind Method; Female; Humans; Middle Aged; Neoplasms; Placebos; Prostaglandin Antagonists; Pyrazoles; Reference Values; RNA; Sulfonamides; Transforming Growth Factor beta; Vitamin D

2013
Effect of infliximab on the levels of TNF-alpha and TGF-beta in the whole blood cultures of irradiated patients.
    Folia histochemica et cytobiologica, 2008, Volume: 46, Issue:3

    TGF-beta is supposed to be the major cytokine responsible for post-radiation fibrosis of healthy tissues and actively modifies post-radiation changes. The growth of TGF-beta level induces the expression of collagen synthesis gene which triggers off the production of fibrosis of hyaline membranes. The main purpose of this study was to discover the way and methods of reducing post-radiation damage of normal tissues and provide an adequate scientific justification for using Infliximab as an effective radio protector in the neoplasm radiotherapy. A group of 97 patients were subjected to the experiment. Randomly selected patients were assigned to 3 groups according to the radiation exposure. The samples of whole blood were suspended in RPMI 1640 growth medium standardized according to the number of leukocytes. Two milliliters of whole blood was taken from each patient immediately before irradiation and 100 microl sample of the blood was placed in wells with 0.8 mg/ml of Infliximab or without the preparation. TGF-beta levels in blood culture without cA2 before irradiation showed continuous rise from 3978 to 8950 pg/ml at the 96th h. In the post irradiated group without cA2, a continuous growth was recorded till the 48th h (from 4758 to 13324 pg/ml at the 24th h) and then a slight decline to 11950 pg/ml at 96th h, respectively. In the cultures with cA2, TGF-beta levels before irradiation showed also the peak value at the 48th h (from 4050 to 7340 pg/ml at the 48th h) and then started to go down (6500 pg/ml at the 72nd h and 5720 pg/ml at the 96th h). In the post-irradiated group, during the first 6 hours, there was a growth from 4717 pg/ml to 7462 pg/ml, and then a paradoxical increase to 16885 pg/ml at the 12th h. From the 12th h the values started to decrease to 6895 pg/ml at the 96th h. The obtained results confirmed the hypothesis of decreasing the TGF-beta expression by inactivating TNF-alpha with a monoclonal antibody (Infliximab) in the patients' whole blood culture in vitro. These observations are a good starting point for further experiments in vitro and in vivo, whose main objective is to reduce post radiation fibrosis.

    Topics: Adolescent; Adult; Aged; Antibodies, Monoclonal; Female; Humans; Infliximab; Male; Middle Aged; Neoplasms; Radiation Pneumonitis; Radiation-Protective Agents; Radiation, Ionizing; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Young Adult

2008
Elevated transforming growth factor beta levels in the plasma of cytokine-treated cancer patients and normal allogeneic stem cell donors.
    Cytotherapy, 2001, Volume: 3, Issue:5

    Allogeneic blood stem-cell donors demonstrate more vigorous mobilization of CD34(+) cells to the circulation in response to cytokine administration than do autologous donors. Transforming growth factor (TGF-beta1) has been implicated as a mobilization inhibitor. A study was designed to determine whether plasma TGF-beta1 levels are elevated in cytokine-mobilized autologous cancer donors compared with cytokine-mobilized normal donors.. Plasma collected from 29 autologous cancer donors and 33 normal allogeneic stem-cell donors following administration of mobilizing cytokines just prior to the first collection was assayed for TGF-beta1 using a sandwich-type ELISA. Plasma from three volunteers not treated with cytokine was also analyzed. Comparisons were made using the Student's t test on log-transformed data.. Average TGF-beta1 levels in the plasma of cancer patients were significantly higher than in allogeneic stem-cell donors (4.4 ng/mL versus 7.2 ng/mL; p = 0.038). The allogeneic donors required fewer collections to harvest greater numbers of CD34(+) cells and colony-forming unit granulocyte-macrophage (CFU-GM) than autologous donors. Plasma from three untreated volunteers had mean TGF-beta1 levels of 0.36 ng/mL, with all three levels below the 25th percentile for allogeneic donors and the 5th percentile for cancer patients.. Cytokine administration was associated with increased plasma TGF-beta1 levels. The levels were higher in cancer patients being mobilized for stem-cell collection than in allogeneic blood stem-cell donors. These differences could underlie the increased number of apheresis procedures required to harvest autologous graft products from cancer patients.

    Topics: Antigens, CD34; Blood Donors; Breast Neoplasms; Enzyme-Linked Immunosorbent Assay; Granulocyte Colony-Stimulating Factor; Hematopoietic Stem Cell Mobilization; Hematopoietic Stem Cells; Humans; Lymphoma, Non-Hodgkin; Neoplasms; Transforming Growth Factor beta; Transplantation, Autologous; Transplantation, Homologous

2001
Increase of plasma transforming growth factor beta (TGF beta) during immunotherapy with IL-2.
    Cancer investigation, 1995, Volume: 13, Issue:6

    Interleukin-2 (IL-2) is a lymphokine with pleiotropic activities on the immune system. When administered in vivo, besides inducing unrestricted tumor cytotoxicity, it is also responsible for the secondary release of other lymphokines, such as IL-1, TNF, and marrow growth factors, which may mediate some of the clinical toxicities (as well as therapeutic effects) seen during IL-2 immunotherapy. Among the clinical effects of IL-2, we previously reported thrombocytopenia and IL-2-induced in vitro inhibition of platelet aggregation accompanied by rapid secretion of alpha-granule components such as platelet factor 4 (PF4) and beta-thromboglobulin. Platelets constitute one of the largest storage forms of TGF beta. Preliminary evaluation of this factor in patients receiving IL-2 had indicated that plasma TGF beta activity increased in cancer patients following IL-2 therapy. We report a more detailed study of the quantitation of TGF beta activity in the plasma of 23 cancer patients treated with IL-2 immunotherapy. Of interest, we found that although elevation of the bioactive form of TGF beta occurred in most patients during IL-2 therapy, it was significantly higher in patients with clinical regression of tumor (p = .004). In the first 2 weeks of therapy increase of plasma TGF beta activity appeared to correlate with a decrease of platelet counts, suggesting that the factor may derive from the storage form of TGF beta contained therein.

    Topics: Adult; Aged; Blood Platelets; Female; Humans; Immunotherapy; Interleukin-2; Male; Middle Aged; Neoplasms; Platelet Count; Transforming Growth Factor beta

1995

Other Studies

475 other study(ies) available for transforming-growth-factor-beta and Neoplasms

ArticleYear
TGFβ signaling activation correlates with immune-inflamed tumor microenvironment across human cancers and predicts response to immunotherapy.
    Cell cycle (Georgetown, Tex.), 2023, Volume: 22, Issue:1

    Considering the determining role of TGFβ signaling in the tumor microenvironment (TME) on immune evasion, the inhibition of signaling is expected to enhance the therapeutic efficacy of immunotherapies, especially immune checkpoint blockade (ICB), which is confirmed in preclinical data. However, successive failures in clinical translation occur at the initial stage. To provide a better understanding of TGFβ signaling within the TME and its relation to the individual immunological status, we performed a pan-cancer analysis comparing the activation of TGFβ pathway among different TMEs based on multi-omics data. Compared with non-inflamed tumors, increased TGFβ signaling activity appeared in four non-cancer cell types within TME in inflamed tumors. Significant correlations were revealed between TGFβ signaling and reliable biomarkers for ICB therapy, as well as between TGFβ signaling and HPV status. Our findings contribute to explain the inconsistency between preclinical and clinical research, and are crucial to optimizing upcoming clinical trial design and improving patient stratification for personalized prediction.

    Topics: Humans; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2023
Galunisertib synergistically potentiates the doxorubicin-mediated antitumor effect and kickstarts the immune system against aggressive lymphoma.
    International immunopharmacology, 2023, Volume: 114

    In clinical practice, major efforts are underway to identify appropriate drug combinations to boost anticancer activity while suppressing unwanted adverse effects. In this regard, we evaluated the efficacy of combination treatment with the widely used chemotherapeutic drug doxorubicin along with the TGFβRI inhibitor galunisertib (LY2157299) in aggressive B-cell non-Hodgkin lymphoma (B-NHL). The antiproliferative effects of these drugs as single agents or in combination against several B-NHL cell lines and the synergism of the drug combination were evaluated by calculating the combination index. To understand the putative molecular mechanism of drug synergism, the TGF-β and stress signaling pathways were analyzed after combination treatment. An aggressive lymphoma model was used to evaluate the anticancer activity and post-therapeutic immune response of the drug combination in vivo. Galunisertib sensitized various B-NHL cells to doxorubicin and in combination synergistically increased apoptosis. The antitumor activity of the drug combinations involved upregulation of p-P38 MAPK and inhibition of the TGF-β/Smad2/3 and PI3K/AKT signaling pathways. Combined drug treatment significantly reduced tumor growth and enhanced survival, indicating that the synergism between galunisertib and Dox observed in vitro was most likely retained in vivo. Based on the tumor-draining lymph node analysis, combination therapy results in better prognosis, including disappearance of disease-exacerbating regulatory T cells and prevention of CD8

    Topics: Apoptosis; Cell Line, Tumor; Doxorubicin; Drug Synergism; Humans; Immune System; Lymphoma; Neoplasms; Phosphatidylinositol 3-Kinases; Transforming Growth Factor beta

2023
Molecular Pathways and Mechanisms of TGFβ in Cancer Therapy.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2023, 06-01, Volume: 29, Issue:11

    Even though the number of agents that inhibit TGFβ being tested in patients with cancer has grown substantially, clinical benefit from TGFβ inhibition has not yet been achieved. The myriad mechanisms in which TGFβ is protumorigenic may be a key obstacle to its effective deployment; cancer cells frequently employ TGFβ-regulated programs that engender plasticity, enable a permissive tumor microenvironment, and profoundly suppress immune recognition, which is the target of most current early-phase trials of TGFβ inhibitors. Here we discuss the implications of a less well-recognized aspect of TGFβ biology regulating DNA repair that mediates responses to radiation and chemotherapy. In cancers that are TGFβ signaling competent, TGFβ promotes effective DNA repair and suppresses error-prone repair, thus conferring resistance to genotoxic therapies and limiting tumor control. Cancers in which TGFβ signaling is intrinsically compromised are more responsive to standard genotoxic therapy. Recognition that TGFβ is a key moderator of both DNA repair and immunosuppression might be used to synergize combinations of genotoxic therapy and immunotherapy to benefit patients with cancer.

    Topics: DNA Damage; DNA Repair; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2023
TLE3 Sustains Luminal Breast Cancer Lineage Fidelity to Suppress Metastasis.
    Cancer research, 2023, 04-04, Volume: 83, Issue:7

    Breast cancer subtypes and their phenotypes parallel different stages of the mammary epithelial cell developmental hierarchy. Discovering mechanisms that control lineage identity could provide novel avenues for mitigating disease progression. Here we report that the transcriptional corepressor TLE3 is a guardian of luminal cell fate in breast cancer and operates independently of the estrogen receptor. In luminal breast cancer, TLE3 actively repressed the gene-expression signature associated with highly aggressive basal-like breast cancers (BLBC). Moreover, maintenance of the luminal lineage depended on the appropriate localization of TLE3 to its transcriptional targets, a process mediated by interactions with FOXA1. By repressing genes that drive BLBC phenotypes, including SOX9 and TGFβ2, TLE3 prevented the acquisition of a hybrid epithelial-mesenchymal state and reduced metastatic capacity and aggressive cellular behaviors. These results establish TLE3 as an essential transcriptional repressor that sustains the more differentiated and less metastatic nature of luminal breast cancers. Approaches to induce TLE3 expression could promote the acquisition of less aggressive, more treatable disease states to extend patient survival.. Transcriptional corepressor TLE3 actively suppresses SOX9 and TGFβ transcriptional programs to sustain the luminal lineage identity of breast cancer cells and to inhibit metastatic progression.

    Topics: Breast Neoplasms; Cell Differentiation; Co-Repressor Proteins; Humans; Neoplasms; Receptors, Estrogen; Transcription Factors; Transforming Growth Factor beta

2023
The paracaspase MALT1 is a downstream target of Smad3 and potentiates the crosstalk between TGF-β and NF-kB signaling pathways in cancer cells.
    Cellular signalling, 2023, Volume: 105

    TGF-β signaling mediates its biological effects by engaging canonical Smad proteins and crosstalking extensively with other signaling networks, including the NF-kB pathway. The paracaspase MALT1 is an intracellular signaling molecule essential for NF-kB activation downstream of several key cell surface receptors. Despite intensive research on TGF-β and NF-kB interactions, the significance of MALT1 in this context remains undecoded. Here we provide experimental evidence supporting that MALT1 functions to converge these pathways. Using A549 and Huh7 cancer cell line models, we report that TGF-β stimulation enhances MALT1 protein and transcript levels in a time- and dose-dependent manner. Systematic and selective perturbation of TGF-β signaling components identifies MALT1 as a downstream target of Smad3. Rescue experiments in SMAD3 knockout cells confirm that C-terminal phosphorylation of Smad3 is central to MALT1 induction. Corroborating these data, we document that the expression of SMAD3 and MALT1 genes are positively correlated in TCGA cohorts, and we trace the molecular basis of MALT1 elevation to promoter activation. Functional studies in parental as well as NF-kB p65 signaling reporter engineered cells conclusively reveal that MALT1 is paramount for TGF-β-stimulated nuclear translocation and transcriptional activation of NF-kB p65. Furthermore, we find that BCL10 is also implicated in TGF-β activation of NF-kB target genes, potentially coupling the TGF-β-MALT1-NF-kB signaling axis to the CARMA-BCL10-MALT1 (CBM) signalosome. The novel findings of this study indicate that MALT1 is a downstream target of the canonical TGF-β/Smad3 pathway and plays a critical role in modulating TGF-β and NF-kB crosstalk in cancer.

    Topics: CARD Signaling Adaptor Proteins; Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein; Neoplasms; NF-kappa B; Signal Transduction; Transforming Growth Factor beta

2023
A Perspective on Bone Morphogenetic Proteins: Dilemma behind Cancer- related Responses.
    Current drug targets, 2023, Volume: 24, Issue:5

    Bone morphogenetic proteins are a center of serious concern and are known to execute various cancer-related issues. The BMP signaling cascades have become more unpredictable as a result of their pleiotropic and risky characteristics, particularly when it comes to cancer responses. This perspective discusses the current therapeutic implications, emphasizes different cellular aspects that impact the failures of the current drug treatments, and speculates on future research avenues that include novel strategies like metabolomic studies and bio-mimetic peptide therapeutics to mitigate cancerous outcomes.

    Topics: Bone Morphogenetic Proteins; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2023
Acidity-mediated induction of FoxP3
    European journal of immunology, 2023, Volume: 53, Issue:6

    Glucose limitation and increased lactic acid levels are consequences of the elevated glycolytic activity of tumor cells, and constitute a metabolic barrier for the function of tumor infiltrating effector immune cells. The immune-suppressive functions of regulatory T cells (Tregs) are unobstructed in lactic-acid rich environments. However, the impact of lactic acid on the induction of Tregs remains unknown. We observed increased TGFβ-mediated induction of Forkhead box P3

    Topics: Forkhead Transcription Factors; Humans; Immunosuppression Therapy; Neoplasms; T-Lymphocytes, Regulatory; Transcription Factors; Transforming Growth Factor beta; Tumor Microenvironment

2023
Anterior gradient-2 regulates cell communication by coordinating cytokine-chemokine signaling and immune infiltration in breast cancer.
    Cancer science, 2023, Volume: 114, Issue:6

    Anterior gradient-2 (AGR2) is crucial to breast cancer progression. However, its role in the tumor immune microenvironment remains unclear. RNA sequencing expression profiles and associated clinical information were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases, respectively. The AGR2 expression patterns were verified using clinical samples of breast cancer. Based on single-cell transcriptomic data, AGR2 expression patterns were identified and cell communication analysis was carried out. Furthermore, the roles of AGR2 in breast tumor progression were explored by a series of functional experiments. We found that DNA methylation was an important mechanism for regulating the expression patterns of AGR2. Patients with AGR2 low expression displayed an immune "hot" and immunosuppressive phenotype characterized by high abundance of tumor immune cell infiltration and increased enrichment scores for transforming growth factor-β (TGF-β) and epithelial-mesenchymal transition pathways, whereas patients with AGR2 high expression showed an opposite immunologic feature with a lack of immune cell infiltration, suggestive of an immune "cold" and desert phenotype. Moreover, single-cell analysis further revealed that AGR2 in malignant cells alters cell-cell interactions by coordinating cytokine-chemokine signaling and immune infiltration. Notably, two immunotherapy cohorts revealed that AGR2-coexpressed genes could serve as prognostic indicators of patient survival. In conclusion, AGR2 could promote breast cancer progression by affecting the tumor immune microenvironment. Patients with AGR2 low expression could be suitable for combination treatment with immune checkpoint inhibitor agents and TGF-β blockers. Therefore, this study provides a theoretical foundation for developing a strategy for personalized immunotherapy to patients with breast cancer.

    Topics: Cell Communication; Chemokines; Cytokines; Mucoproteins; Neoplasms; Oncogene Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2023
Vaccination with celecoxib-treated dendritic cells improved cellular immune responses in an animal breast cancer model.
    Advances in medical sciences, 2023, Volume: 68, Issue:1

    Prostaglandin E2 (PGE2), a product of cyclooxygenase (COX) pathway of arachidonic acid, exerts inhibitory impacts on dendritic cell (DC) activity to repress anti-tumor immune responses. Therefore, targeting COX during DC vaccine generation may enhance DC-mediated antitumor responses. We aimed to investigate the impacts of DC vaccine treated with celecoxib (CXB), a selective COX2 inhibitor, on some T cell-related parameters.. Breast cancer (BC) was induced in BALB/c mice, and then they received DC vaccine treated with lipopolysaccharide (LPS-mDCs), LPS with a 5 ​μM dose of CXB (LPS/CXB5-mDCs) and LPS with a 10 ​μM dose of CXB (LPS/CXB10-mDCs). The frequency of splenic Th1 and Treg cells and amounts of IFN-γ, IL-12 and TGF-β production by splenocytes, as well as, the expression of Granzyme-B, T-bet and FOXP3 in tumors were determined using flow cytometry, ELISA, and real-time PCR, respectively.. Compared with untreated tumor group (T-control), treatment with LPS/CXB5-mDCs and LPS/CXB10-mDCs decreased tumor growth (P ​= ​0.009 and P ​< ​0.0001), escalated survival rate (P ​= ​0.002), increased the frequency of splenic Th1 cells (P ​= ​0.0872, and P ​= ​0.0155), increased the IFN-γ (P ​= ​0.0003 and P ​= ​0.0061) and IL-12 (P ​= ​0.001 and P ​= ​0.0009) production by splenocytes, upregulated T-bet (P ​= ​0.062 and P ​< ​0.0001) and Granzyme-B (P ​= ​0.0448 and P ​= ​0.4485), whereas decreased the number of Treg cells (P ​= ​0.0014, and P ​= ​0.0219), reduced the amounts of TGF-β production by splenocytes (P ​= ​0.0535 and P ​= ​0.0169), and reduced the expression of FOXP3 (P ​= ​0.0006 and P ​= ​0.0057) in comparison with T-control group.. Our findings show that LPS/CXB-treated DC vaccine potently modulated antitumor immune responses in a mouse BC model.

    Topics: Animals; Celecoxib; Dendritic Cells; Forkhead Transcription Factors; Granzymes; Immunity, Cellular; Interleukin-12; Lipopolysaccharides; Mice; Neoplasms; Transforming Growth Factor beta; Vaccination; Vaccines

2023
ABCB1 and ABCC1 Function during TGF-β-Induced Epithelial-Mesenchymal Transition: Relationship between Multidrug Resistance and Tumor Progression.
    International journal of molecular sciences, 2023, Mar-23, Volume: 24, Issue:7

    Multidrug resistance (MDR) and induction of metastasis are some of the puzzles encountered during cancer chemotherapy. The MDR phenotype is associated with overexpression of ABC transporters, involved in drug efflux. Metastasis originates from the epithelial-mesenchymal transition (EMT), in which cells acquire a migratory phenotype, invading new tissues. ABC transporters' role during EMT is still elusive, though cells undergoing EMT exhibit enhanced ABCB1 expression. We demonstrated increased ABCB1 expression but no change in activity after TGF-β-induced EMT in A549 cells. Moreover, ABCB1 inhibition by verapamil increased snail and fibronectin expression, an event associated with upregulation of ABCB1, evidencing coincident cell signaling pathways leading to ABCB1 and EMT-related markers transcription, rather than a direct effect of transport. Additionally, for the first time, increased ABCC1 expression and activity was observed after EMT, and use of ABCC1 inhibitors partially inhibited EMT-marker snail, although increased ABCC1 function translated into collateral sensibility to daunorubicin. More investigations must be done to evaluate the real benefits that the gain of ABC transporters might have on the process of metastasis. Considering ABCC1 is involved in the stress response, affecting intracellular GSH content and drug detoxification, this transporter could be used as a therapeutic target in cancer cells undergoing EMT.

    Topics: ATP Binding Cassette Transporter, Subfamily B; ATP-Binding Cassette Transporters; Cell Line, Tumor; Drug Resistance, Multiple; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Humans; Multidrug Resistance-Associated Proteins; Neoplasms; Transforming Growth Factor beta

2023
Evaluation of the interaction between tumor growth factor-β and interferon type I pathways in patients with COVID-19: focusing on ages 1 to 90 years.
    BMC infectious diseases, 2023, Apr-18, Volume: 23, Issue:1

    Evidence revealed that age could affect immune responses in patients with the acute respiratory syndrome of coronavirus 2 (SARS-CoV-2) infection. This study investigated the impact of age on immune responses, especially on the interaction between the tumor growth factor-β (TGF-β) and interferon type-I (IFN-I) axes in the pathogenesis of novel coronavirus disease 2019 (COVID-19).. This age-matched case-control investigation enrolled 41 COVID-19 patients and 40 healthy controls categorized into four groups, including group 1 (up to 20 years), group 2 (20-40 years), group 3 (40-60 years), and group 4 (over 60 years). Blood samples were collected at the time of admission. The expression of TGF-βRI, TGF-βRII, IFNARI, IFNARII, interferon regulatory factor 9 (IRF9), and SMAD family member 3 (SMAD3) was measured using the real-time PCR technique. In addition, serum levels of TGF-β, IFN-α, and SERPINE1 were measured by the enzyme-linked immunosorbent assay (ELISA) technique. All biomarkers were measured and analyzed in the four age studies groups.. The expression of TGF-βRI, TGF-βRII, IFNARI, IFNARII, IRF9, and SMAD3 was markedly upregulated in all age groups of patients compared with the matched control groups. Serum levels of IFN-α and SERPINE1 were significantly higher in patient groups than in control groups. While TGF-β serum levels were only significantly elevated in the 20 to 40 and over 60 years patient group than in matched control groups.. These data showed that the age of patients, at least at the time of admission, may not significantly affect TGF-β- and IFN-I-associated immune responses. However, it is possible that the severity of the disease affects these pathway-mediated responses, and more studies with a larger sample size are needed to verify it.

    Topics: Adolescent; Adult; Aged; Aged, 80 and over; Child; Child, Preschool; COVID-19; Humans; Infant; Interferon Type I; Middle Aged; Neoplasms; SARS-CoV-2; Transforming Growth Factor beta; Young Adult

2023
BCA101 Is a Tumor-Targeted Bifunctional Fusion Antibody That Simultaneously Inhibits EGFR and TGFβ Signaling to Durably Suppress Tumor Growth.
    Cancer research, 2023, 06-02, Volume: 83, Issue:11

    The EGFR and TGFβ signaling pathways are important mediators of tumorigenesis, and cross-talk between them contributes to cancer progression and drug resistance. Therapies capable of simultaneously targeting EGFR and TGFβ could help improve patient outcomes across various cancer types. Here, we developed BCA101, an anti-EGFR IgG1 mAb linked to an extracellular domain of human TGFβRII. The TGFβ "trap" fused to the light chain in BCA101 did not sterically interfere with its ability to bind EGFR, inhibit cell proliferation, or mediate antibody-dependent cellular cytotoxicity. Functional neutralization of TGFβ by BCA101 was demonstrated by several in vitro assays. BCA101 increased production of proinflammatory cytokines and key markers associated with T-cell and natural killer-cell activation, while suppressing VEGF secretion. In addition, BCA101 inhibited differentiation of naïve CD4+ T cells to inducible regulatory T cells (iTreg) more strongly than the anti-EGFR antibody cetuximab. BCA101 localized to tumor tissues in xenograft mouse models with comparable kinetics to cetuximab, both having better tumor tissue retention over TGFβ "trap." TGFβ in tumors was neutralized by approximately 90% in animals dosed with 10 mg/kg of BCA101 compared with 54% in animals dosed with equimolar TGFβRII-Fc. In patient-derived xenograft mouse models of head and neck squamous cell carcinoma, BCA101 showed durable response after dose cessation. The combination of BCA101 and anti-PD1 antibody improved tumor inhibition in both B16-hEGFR-expressing syngeneic mouse models and in humanized HuNOG-EXL mice bearing human PC-3 xenografts. Together, these results support the clinical development of BCA101 as a monotherapy and in combination with immune checkpoint therapy.. The bifunctional mAb fusion design of BCA101 targets it to the tumor microenvironment where it inhibits EGFR and neutralizes TGFβ to induce immune activation and to suppress tumor growth.

    Topics: Animals; Antibodies, Monoclonal, Humanized; Carcinoma, Squamous Cell; Cell Line, Tumor; Cetuximab; ErbB Receptors; Head and Neck Neoplasms; Humans; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment; Xenograft Model Antitumor Assays

2023
Analogies between the periphery of cancer and the leading edge of pulmonary fibrosis.
    Journal of translational medicine, 2023, 04-21, Volume: 21, Issue:1

    The periphery of malignant tumors and the leading edge of fibrotic tissue have analogous metabolic pathways. Both use glycolysis as the primary source of energy to produce biomass with consequential acidification of the microenvironment. A low PH has been shown to increase the ability of cancer cells to invade the surrounding tissue in both in vitro and in vivo studies. The pH-dependent activation of TGF-B leading to myofibroblast activation is an important step in the initiation and progression of fibrosis. Markers of accelerated cell proliferation have also been reported in the periphery of malignant tumors and the leading edge of fibrosis. Understanding the shared molecular and metabolic characteristics of these conditions may explain the increased prevalence of cancer among patients with fibrosis.

    Topics: Cell Differentiation; Fibroblasts; Fibrosis; Humans; Myofibroblasts; Neoplasms; Pulmonary Fibrosis; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Microenvironment

2023
Soluble dipeptidyl peptidase-4 induces epithelial-mesenchymal transition through tumor growth factor-β receptor.
    Pharmacological reports : PR, 2023, Volume: 75, Issue:4

    Kidney fibrosis is the final manifestation of chronic kidney disease, a condition mainly caused by diabetic nephropathy. Persistent tissue damage leads to chronic inflammation and excessive deposition of extracellular matrix (ECM) proteins. Epithelial-mesenchymal transition (EMT) is involved in a variety of tissue fibrosis and is a process during which epithelial cells transform into mesenchymal-like cells and lose their epithelial functionality and characteristics Dipeptidyl peptidase-4 (DPP4) is widely expressed in tissues, especially those of the kidney and small intestine. DPP4 exists in two forms: a plasma membrane-bound and a soluble form. Serum-soluble DPP4 (sDPP4) levels are altered in many pathophysiological conditions. Elevated circulating sDPP4 is correlated with metabolic syndrome. Because the role of sDPP4 in EMT remains unclear, we examined the effect of sDPP4 on renal epithelial cells.. The influences of sDPP4 on renal epithelial cells were demonstrated by measuring the expression of EMT markers and ECM proteins.. sDPP4 upregulated the EMT markers ACTA2 and COL1A1 and increased total collagen content. sDPP4 activated SMAD signaling in renal epithelial cells. Using genetic and pharmacological methods to target TGFBR, we observed that sDPP4 activated SMAD signaling through TGFBR in epithelial cells, whereas genetic ablation and treatment with TGFBR antagonist prevented SMAD signaling and EMT. Linagliptin, a clinically available DPP4 inhibitor, abrogated sDPP4-induced EMT.. This study indicated that sDPP4/TGFBR/SMAD axis leads to EMT in renal epithelial cells. Elevated circulating sDPP4 levels may contribute to mediators that induce renal fibrosis.

    Topics: Dipeptidyl Peptidase 4; Epithelial-Mesenchymal Transition; Fibrosis; Humans; Neoplasms; Transforming Growth Factor beta; Transforming Growth Factor beta1

2023
Nanovesicles loaded with a TGF-β receptor 1 inhibitor overcome immune resistance to potentiate cancer immunotherapy.
    Nature communications, 2023, 06-16, Volume: 14, Issue:1

    The immune-excluded tumors (IETs) show limited response to current immunotherapy due to intrinsic and adaptive immune resistance. In this study, it is identified that inhibition of transforming growth factor-β (TGF-β) receptor 1 can relieve tumor fibrosis, thus facilitating the recruitment of tumor-infiltrating T lymphocytes. Subsequently, a nanovesicle is constructed for tumor-specific co-delivery of a TGF-β inhibitor (LY2157299, LY) and the photosensitizer pyropheophorbide a (PPa). The LY-loaded nanovesicles suppress tumor fibrosis to promote intratumoral infiltration of T lymphocytes. Furthermore, PPa chelated with gadolinium ion is capable of fluorescence, photoacoustic and magnetic resonance triple-modal imaging-guided photodynamic therapy, to induce immunogenic death of tumor cells and elicit antitumor immunity in preclinical cancer models in female mice. These nanovesicles are further armored with a lipophilic prodrug of the bromodomain-containing protein 4 inhibitor (i.e., JQ1) to abolish programmed death ligand 1 expression of tumor cells and overcome adaptive immune resistance. This study may pave the way for nanomedicine-based immunotherapy of the IETs.

    Topics: Animals; Cell Line, Tumor; Female; Immunotherapy; Lymphocytes, Tumor-Infiltrating; Mice; Neoplasms; Photosensitizing Agents; Transforming Growth Factor beta; Tumor Microenvironment

2023
The lncRNA LETS1 promotes TGF-β-induced EMT and cancer cell migration by transcriptionally activating a TβR1-stabilizing mechanism.
    Science signaling, 2023, 06-20, Volume: 16, Issue:790

    Transforming growth factor-β (TGF-β) signaling is a critical driver of epithelial-to-mesenchymal transition (EMT) and cancer progression. In SMAD-dependent TGF-β signaling, activation of the TGF-β receptor complex stimulates the phosphorylation of the intracellular receptor-associated SMADs (SMAD2 and SMAD3), which translocate to the nucleus to promote target gene expression. SMAD7 inhibits signaling through the pathway by promoting the polyubiquitination of the TGF-β type I receptor (TβRI). We identified an unannotated nuclear long noncoding RNA (lncRNA) that we designated LETS1 (lncRNA enforcing TGF-β signaling 1) that was not only increased but also perpetuated by TGF-β signaling. Loss of LETS1 attenuated TGF-β-induced EMT and migration in breast and lung cancer cells in vitro and extravasation of the cells in a zebrafish xenograft model. LETS1 potentiated TGF-β-SMAD signaling by stabilizing cell surface TβRI, thereby forming a positive feedback loop. Specifically, LETS1 inhibited TβRI polyubiquitination by binding to nuclear factor of activated T cells (NFAT5) and inducing the expression of the gene encoding the orphan nuclear receptor 4A1 (NR4A1), a component of a destruction complex for SMAD7. Overall, our findings characterize LETS1 as an EMT-promoting lncRNA that potentiates signaling through TGF-β receptor complexes.

    Topics: Animals; Cell Movement; Humans; Neoplasms; Receptors, Transforming Growth Factor beta; RNA, Long Noncoding; Transforming Growth Factor beta; Zebrafish

2023
TGF-β1 mediates tumor immunosuppression aggravating at the late stage post-high-light-dose photodynamic therapy.
    Cancer immunology, immunotherapy : CII, 2023, Volume: 72, Issue:9

    Photodynamic therapy (PDT) is an emerging clinical treatment that is expected to become an important adjuvant strategy for the immunotherapeutic cancer treatment. Recently, numerous works have reported combination strategies. However, clinical data showed that the anti-tumor immune response of PDT was not lasting though existing. The immune activation effect will eventually turn to immunosuppressive effect and get aggravated at the late stage post-PDT. So far, the mechanism is still unclear, which limits the design of specific correction strategies and further development of PDT. Several lines of evidence suggest a role for TGF-β1 in the immunosuppression associated with PDT. Herein, this study systematically illustrated the dynamic changes of immune states post-PDT within the tumor microenvironment. The results clearly demonstrated that high-light-dose PDT, as a therapeutic dose, induced early immune activation followed by late immunosuppression, which was mediated by the activated TGF-β1 upregulation. Then, the mechanism of PDT-induced TGF-β1 accumulation and immunosuppression was elucidated, including the ROS/TGF-β1/MMP-9 positive feedback loop and CD44-mediated local amplification, which was further confirmed by spatial transcriptomics, as well as by the extensive immune inhibitory effect of local high concentration of TGF-β1. Finally, a TGF-β blockade treatment strategy was presented as a promising combinational strategy to reverse high-light-dose PDT-associated immunosuppression. The results of this study provide new insights for the biology mechanism and smart improvement approaches to enhance tumor photodynamic immunotherapy.

    Topics: Cell Line, Tumor; Humans; Immunosuppression Therapy; Neoplasms; Photochemotherapy; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Microenvironment

2023
An oncolytic virus-delivered TGFβ inhibitor overcomes the immunosuppressive tumor microenvironment.
    The Journal of experimental medicine, 2023, 10-02, Volume: 220, Issue:10

    While checkpoint blockade immunotherapies have widespread success, they rely on a responsive immune infiltrate; as such, treatments enhancing immune infiltration and preventing immunosuppression are of critical need. We previously generated αPD-1 resistant variants of the murine HNSCC model MEER. While entirely αPD-1 resistant, these tumors regress after single dose of oncolytic vaccinia virus (VV). We then generated a VV-resistant MEER line to dissect the immunologic features of sensitive and resistant tumors. While treatment of both tumor types induced immune infiltration and IFNγ, we found a defining feature of resistance was elevation of immunosuppressive cytokines like TGFβ, which blunted IFNγ signaling, especially in regulatory T cells. We engineered VV to express a genetically encoded TGFβRII inhibitor. Inhibitor-expressing VV produced regressions in resistant tumor models and showed impressive synergy with checkpoint blockade. Importantly, tumor-specific, viral delivery of TGFβ inhibition had no toxicities associated with systemic TGFβ/TGFβR inhibition. Our data suggest that aside from stimulating immune infiltration, oncolytic viruses are attractive means to deliver agents to limit immunosuppression in cancer.

    Topics: Animals; Cell Line, Tumor; Immunosuppressive Agents; Mice; Neoplasms; Oncolytic Virotherapy; Oncolytic Viruses; Transforming Growth Factor beta; Tumor Microenvironment; Vaccinia virus

2023
Tumor-mediated immunosuppression and cytokine spreading affects the relation between EMT and PD-L1 status.
    Frontiers in immunology, 2023, Volume: 14

    Epithelial-mesenchymal transition (EMT) and immune resistance mediated by Programmed Death-Ligand 1 (PD-L1) upregulation are established drivers of tumor progression. Their bi-directional crosstalk has been proposed to facilitate tumor immunoevasion, yet the impact of immunosuppression and spatial heterogeneity on the interplay between these processes remains to be characterized. Here we study the role of these factors using mathematical and spatial models. We first designed models incorporating immunosuppressive effects on T cells mediated

    Topics: B7-H1 Antigen; Cytokines; Epithelial-Mesenchymal Transition; Humans; Immunosuppression Therapy; Neoplasms; Transforming Growth Factor beta

2023
Mechanical microscopy of cancer cells: TGF-β induced epithelial to mesenchymal transition corresponds to low intracellular viscosity in cancer cells.
    The Journal of the Acoustical Society of America, 2023, 09-01, Volume: 154, Issue:3

    Viscosity is an essential parameter that regulates bio-molecular reaction rates of diffusion-driven cellular processes. Hence, abnormal viscosity levels are often associated with various diseases and malfunctions like cancer. For this reason, monitoring intracellular viscosity becomes vital. While several approaches have been developed for in vitro and in vivo measurement of viscosity, analysis of intracellular viscosity in live cells has not yet been well realized. Our research introduces a novel, natural frequency-based, non-invasive method to determine the intracellular viscosity in cells. This method can not only efficiently analyze the differences in intracellular viscosity post modulation with molecules like PEG or glucose but is sensitive enough to distinguish the difference in intra-cellular viscosity among various cancer cell lines such as Huh-7, MCF-7, and MDAMB-231. Interestingly, TGF-β a cytokine reported to induce epithelial to mesenchymal transition (EMT), a feature associated with cancer invasiveness resulted in reduced viscosity of cancer cells, as captured through our method. To corroborate our findings with existing methods of analysis, we analyzed intra-cellular viscosity with a previously described viscosity-sensitive molecular rotor-based fluorophore-TPSII. In parity with our position sensing device (PSD)-based approach, an increase in fluorescence intensity was observed with viscosity enhancers, while, TGF-β exposure resulted in its reduction in the cells studied. This is the first study of its kind that attempts to characterize differences in intracellular viscosity using a novel, non-invasive PSD-based method.

    Topics: Cytokines; Epithelial-Mesenchymal Transition; Microscopy; Neoplasms; Transforming Growth Factor beta; Viscosity

2023
BRD9-mediated control of the TGF-β/Activin/Nodal pathway regulates self-renewal and differentiation of human embryonic stem cells and progression of cancer cells.
    Nucleic acids research, 2023, Nov-27, Volume: 51, Issue:21

    Bromodomain-containing protein 9 (BRD9) is a specific subunit of the non-canonical SWI/SNF (ncBAF) chromatin-remodeling complex, whose function in human embryonic stem cells (hESCs) remains unclear. Here, we demonstrate that impaired BRD9 function reduces the self-renewal capacity of hESCs and alters their differentiation potential. Specifically, BRD9 depletion inhibits meso-endoderm differentiation while promoting neural ectoderm differentiation. Notably, supplementation of NODAL, TGF-β, Activin A or WNT3A rescues the differentiation defects caused by BRD9 loss. Mechanistically, BRD9 forms a complex with BRD4, SMAD2/3, β-CATENIN and P300, which regulates the expression of pluripotency genes and the activity of TGF-β/Nodal/Activin and Wnt signaling pathways. This is achieved by regulating the deposition of H3K27ac on associated genes, thus maintaining and directing hESC differentiation. BRD9-mediated regulation of the TGF-β/Activin/Nodal pathway is also demonstrated in the development of pancreatic and breast cancer cells. In summary, our study highlights the crucial role of BRD9 in the regulation of hESC self-renewal and differentiation, as well as its participation in the progression of pancreatic and breast cancers.

    Topics: Activins; Cell Cycle Proteins; Cell Differentiation; Embryonic Stem Cells; Human Embryonic Stem Cells; Humans; Neoplasms; Nuclear Proteins; Transcription Factors; Transforming Growth Factor beta; Wnt Signaling Pathway

2023
Monitoring TGFβ signaling in irradiated tumors.
    Methods in cell biology, 2023, Volume: 180

    Transforming growth factor β (TGFβ) is exquisitely regulated under physiological conditions but its activity is highly dysregulated in cancer. All cells make TGFβ and have receptors for the ligand, which is sequestered in the extracellular matrix in a latent form. Ionizing radiation elicits rapid release of TGFβ from these stores, so-called activation, over a wide range of doses and exposures, including low dose (<1Gy) whole-body irradiation, creating an extraordinarily potent signal in the irradiated tissue or tumor. Hence, accurate evaluation of TGFβ activity is complicated because of its ubiquitous distribution as a latent complex. Here we describe conditions for assays that reveal TGFβ activity in situ using either tissue preparations or functional imaging.

    Topics: Extracellular Matrix; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2023
Pharmacophore mapping approach to find anti-cancer phytochemicals with metformin-like activities against transforming growth factor (TGF)-beta receptor I kinase: An in silico study.
    PloS one, 2023, Volume: 18, Issue:11

    The most frequently prescribed first-line treatment for type II diabetes mellitus is metformin. Recent reports asserted that this diabetes medication can also shield users from cancer. Metformin induces cell cycle arrest in cancer cells. However, the exact mechanism by which this occurs in the cancer system is yet to be elucidated. Here, we investigated the impact of metformin on cell cycle arrest in cancer cells utilizing transforming growth factor (TGF)-beta pathway. TGF-ß pathway has significant effect on cell progression and growth. In order to gain an insight on the underlying molecular mechanism of metformin's effect on TGF beta receptor 1 kinase, molecular docking was performed. Metformin was predicted to interact with transforming growth factor (TGF)-beta receptor I kinase based on molecular docking and molecular dynamics simulations. Furthermore, pharmacophore was generated for metformin-TGF-ßR1 complex to hunt for novel compounds having similar pharmacophore as metformin with enhanced anti-cancer potentials. Virtual screening with 29,000 natural compounds from NPASS database was conducted separately for the generated pharmacophores in Ligandscout® software. Pharmacophore mapping showed 60 lead compounds for metformin-TGF-ßR1 complex. Molecular docking, molecular dynamics simulation for 100 ns and ADMET analysis were performed on these compounds. Compounds with CID 72473, 10316977 and 45140078 showed promising binding affinities and formed stable complexes during dynamics simulation with aforementioned protein and thus have potentiality to be developed into anti-cancer medicaments.

    Topics: Diabetes Mellitus, Type 2; Humans; Ligands; Metformin; Molecular Docking Simulation; Molecular Dynamics Simulation; Neoplasms; Pharmacophore; Transforming Growth Factor beta

2023
Convergent insulin and TGF-β signalling drives cancer cachexia by promoting aberrant fat body ECM accumulation in a Drosophila tumour model.
    EMBO reports, 2023, Dec-06, Volume: 24, Issue:12

    In this study, we found that in the adipose tissue of wildtype animals, insulin and TGF-β signalling converge via a BMP antagonist short gastrulation (sog) to regulate ECM remodelling. In tumour bearing animals, Sog also modulates TGF-β signalling to regulate ECM accumulation in the fat body. TGF-β signalling causes ECM retention in the fat body and subsequently depletes muscles of fat body-derived ECM proteins. Activation of insulin signalling, inhibition of TGF-β signalling, or modulation of ECM levels via SPARC, Rab10 or Collagen IV in the fat body, is able to rescue tissue wasting in the presence of tumour. Together, our study highlights the importance of adipose ECM remodelling in the context of cancer cachexia.

    Topics: Adipose Tissue; Animals; Cachexia; Drosophila; Fat Body; Insulin; Neoplasms; Transforming Growth Factor beta

2023
Setting up clinical trials for success: Applying preclinical advances in combined TGFβ/PD-L1 inhibition to ongoing clinical studies.
    Molecular carcinogenesis, 2022, Volume: 61, Issue:2

    Combined TGFβ/PD-L1 inhibition is currently undergoing clinical trials in multiple cancer types. The early reported clinical trials of bintrafusp alfa, a bifunctional fusion protein targeting both of these pathways, have had mixed results. Here, we briefly review recent preclinical advances that can be used to refine these ongoing clinical trials and improve their outcomes.

    Topics: B7-H1 Antigen; Clinical Trials as Topic; Humans; Immunotherapy; Longitudinal Studies; Neoplasms; Transforming Growth Factor beta

2022
Transcriptomic Determinants of Response to Pembrolizumab Monotherapy across Solid Tumor Types.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2022, 04-14, Volume: 28, Issue:8

    To explore relationships between biological gene expression signatures and pembrolizumab response.. RNA-sequencing data on baseline tumor tissue from 1,188 patients across seven tumor types treated with pembrolizumab monotherapy in nine clinical trials were used. A total of 11 prespecified gene expression signatures [18-gene T-cell-inflamed gene expression profile (TcellinfGEP), angiogenesis, hypoxia, glycolysis, proliferation, MYC, RAS, granulocytic myeloid-derived suppressor cell (gMDSC), monocytic myeloid-derived suppressor cell (mMDSC), stroma/epithelial-to-mesenchymal transition (EMT)/TGFβ, and WNT] were evaluated for their relationship to objective response rate (per RECIST, version 1.1). Logistic regression analysis of response for consensus signatures was adjusted for tumor type, Eastern Cooperative Oncology Group performance status, and TcellinfGEP, an approach equivalent to evaluating the association between response and the residuals of consensus signatures after detrending them for their relationship with the TcellinfGEP (previously identified as a determinant of pembrolizumab response) and tumor type. Testing of the 10 prespecified non-TcellinfGEP consensus signatures for negative association [except proliferation (hypothesized positive association)] with response was adjusted for multiplicity.. Covariance patterns of the 11 signatures (including TcellinfGEP) identified in Merck-Moffitt and The Cancer Genome Atlas datasets showed highly concordant coexpression patterns in the RNA-sequencing data from pembrolizumab trials. TcellinfGEP was positively associated with response; signatures for angiogenesis, mMDSC, and stroma/EMT/TGFβ were negatively associated with response to pembrolizumab monotherapy.. These findings suggest that features beyond IFNγ-related T-cell inflammation may be relevant to anti-programmed death 1 monotherapy response and may define other axes of tumor biology as candidates for pembrolizumab combinations. See related commentary by Cho et al., p. 1479.

    Topics: Antibodies, Monoclonal, Humanized; Antineoplastic Agents, Immunological; Humans; Neoplasms; RNA; Transcriptome; Transforming Growth Factor beta

2022
Spontaneous formation and spatial self-organization of mechanically induced mesenchymal-like cells within geometrically confined cancer cell monolayers.
    Biomaterials, 2022, Volume: 281

    There is spatiotemporal heterogeneity in cell phenotypes and mechanical properties in tumor tissues, which is associated with cancer invasion and metastasis. It is well-known that exogenous growth factors like transforming growth factor (TGF)-β, can induce epithelial-mesenchymal transition (EMT)-based phenotypic transformation and the formation of EMT patterning on geometrically confined monolayers with mechanics heterogeneity. In the absence of exogenous TGF-β stimulation, however, whether geometric confinement-caused mechanics heterogeneity of cancer cell monolayers alone can trigger the EMT-based phenotypic heterogeneity still remains mysterious. Here, we develop a micropattern-based cell monolayer model to investigate the regulation of mechanics heterogeneity on the cell phenotypic switch. We reveal that mechanics heterogeneity itself is enough to spontaneously induce the emergence of mesenchymal-like phenotype and asymmetrical activation of TGF-β-SMAD signaling. Spatiotemporal dynamics of patterned cell monolayers with mesenchymal-like phenotypes is essentially regulated by tissue-scale cell behaviors like proliferation, migration as well as heterogeneous cytoskeletal contraction. The inhibition of cell contraction abrogates the asymmetrical TGF-β-SMAD signaling activation level and the emergence of mesenchymal-like phenotype. Our work not only sheds light on the key regulation of mechanics heterogeneity caused by spatially geometric confinement on regional mesenchymal-like phenotype of cancer cell monolayers, but highlights the key role of biophysical/mechanical cues in triggering phenotypic switch.

    Topics: Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factor beta1

2022
DNA-PK Inhibitor Peposertib Amplifies Radiation-Induced Inflammatory Micronucleation and Enhances TGFβ/PD-L1 Targeted Cancer Immunotherapy.
    Molecular cancer research : MCR, 2022, 04-01, Volume: 20, Issue:4

    Radiotherapy is the most widely used cancer treatment and improvements in its efficacy and safety are highly sought-after. Peposertib (also known as M3814), a potent and selective DNA-dependent protein kinase (DNA-PK) inhibitor, effectively suppresses the repair of radiation-induced DNA double-strand breaks (DSB) and regresses human xenograft tumors in preclinical models. Irradiated cancer cells devoid of p53 activity are especially sensitive to the DNA-PK inhibitor, as they lose a key cell-cycle checkpoint circuit and enter mitosis with unrepaired DSBs, leading to catastrophic consequences. Here, we show that inhibiting the repair of DSBs induced by ionizing radiation with peposertib offers a powerful new way for improving radiotherapy by simultaneously enhancing cancer cell killing and response to a bifunctional TGFβ "trap"/anti-PD-L1 cancer immunotherapy. By promoting chromosome misalignment and missegregation in p53-deficient cancer cells with unrepaired DSBs, DNA-PK inhibitor accelerated micronuclei formation, a key generator of cytosolic DNA and activator of cGAS/STING-dependent inflammatory signaling as it elevated PD-L1 expression in irradiated cancer cells. Triple combination of radiation, peposertib, and bintrafusp alfa, a fusion protein simultaneously inhibiting the profibrotic TGFβ and immunosuppressive PD-L1 pathways was superior to dual combinations and suggested a novel approach to more efficacious radioimmunotherapy of cancer.. Selective inhibition of DNA-PK in irradiated cancer cells enhances inflammatory signaling and activity of dual TGFβ/PD-L1 targeted therapy and may offer a more efficacious combination option for the treatment of locally advanced solid tumors.

    Topics: B7-H1 Antigen; DNA; Humans; Immunotherapy; Neoplasms; Protein Kinase Inhibitors; Pyridazines; Quinazolines; Transforming Growth Factor beta

2022
Loss of LRRC33-Dependent TGFβ1 Activation Enhances Antitumor Immunity and Checkpoint Blockade Therapy.
    Cancer immunology research, 2022, 04-01, Volume: 10, Issue:4

    TGFβ has multiple roles and gene products (TGFβ1, -β2, and -β3), which make global targeting of TGFβ undesirable. Expression of TGFβ requires association with milieu molecules, which localize TGFβ to the surface of specific cells or extracellular matrices. Here, we found that LRRC33 was specifically associated with TGFβ1, not TGFβ2 and TGFβ3, and was required for surface display and activation of TGFβ1 on tumor-infiltrating myeloid cells. Loss of LRRC33-dependent TGFβ1 activation slowed tumor growth and metastasis by enhancing innate and adaptive antitumor immunity in multiple mouse syngeneic tumor models. LRRC33 loss resulted in a more immunogenic microenvironment, with decreased myeloid-derived suppressor cells, more active CD8+ T and NK cells, and more skewing toward tumor-suppressive M1 macrophages. LRRC33 loss and PD-1 blockade synergized in controlling B16.F10 tumor growth. Our results demonstrate the importance of LRRC33 in tumor biology and highlight the therapeutic potential of dual blockade of the LRRC33/TGFβ1 axis and PD-1/PD-L1 in cancer immunotherapy.

    Topics: Animals; Cell Line, Tumor; Disease Models, Animal; Immunotherapy; Macrophages; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
The Effect of Geometry and TGF-β Signaling on Tumor Cell Migration from Free-Standing Microtissues.
    Advanced healthcare materials, 2022, Volume: 11, Issue:12

    Recapitulation of 3D multicellular tissues in vitro is of great interest to the field of tumor biology to study the integrated effect of local biochemical and biophysical signals on tumor cell migration and invasion. However, most microengineered tissues and spheroids are unable to recapitulate in vitro the complexities of 3D geometries found in vivo. Here, lithographically defined degradable alginate microniches are presented to produce free-standing tumor microtissues, with precisely controlled geometry, high viability, and allowing for high cell proliferation. The role of microtissue geometry and TGF-β signaling in tumor cell migration is further investigated. TGF-β is found to induce the expression of p-myosin II, vimentin, and YAP/TAZ nuclear localization at the periphery of the microtissue, where enhanced nuclear stiffness and orientation are also observed. Upon embedding in a collagen matrix, microtissues treated with TGF-β maintain their geometric integrity, possibly due to the higher cell tension observed around the periphery. In contrast, cells in microtissues not treated with TGF-β are highly mobile and invade the surrounding matrix rapidly, with the initial migration strongly dependent on the local geometry. The microtissues presented here are promising model systems for studying the influence of biophysical properties and soluble factors on tumor cell migration.

    Topics: Alginates; Cell Movement; Collagen; Humans; Neoplasms; Transforming Growth Factor beta

2022
Dominant negative TGF-β receptor type II in T lymphocytes promotes anti-tumor immunity by modulating T cell subsets and enhancing CTL responses.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2022, Volume: 148

    Transforming growth factor-β (TGF-β) is a multifunctional regulatory cytokine that maintains tolerance in the immune system by regulating the proliferation, differentiation and survival of lymphocytes. TGF-β blockade therapy for cancer has achieved some results but shows limited efficacy and side effects because these drugs are not selective and act on various types of cells throughout the body. We demonstrate here that dominant negative TGF-β receptor type II specifically targeting T cells decreases tumor load in tumor-bearing mice. In addition, the dominant negative TGF-β receptor type II promotes the proliferation and differentiation of T cells and increases the expression of T-bet, which in turn promotes the secretion of granzyme A, granzyme B, perforin and IFN-γ secreted by T cells, and enhances the cytotoxicity and anti-tumor effects of T cells. Moreover, we also found that dominant negative TGF-β receptor type II reduces the proportion of regulatory T cells (Tregs) in tumor tissue and spleen of tumor-bearing mice. Co-culture experiments with T cells and tumor cells revealed that dominant negative TGF-β receptor type II inhibited tumor cell proliferation and increased apoptosis. Our results indicate that specifically inhibiting TGF-β receptor type II in T cells increases anti-tumor immunity and has a strong therapeutic potential.

    Topics: Animals; Apoptosis; Cell Differentiation; Cell Proliferation; Cell Survival; Granzymes; Interferon-gamma; Mice; Neoplasms; Perforin; Receptor, Transforming Growth Factor-beta Type II; Spleen; T-Box Domain Proteins; T-Lymphocyte Subsets; T-Lymphocytes, Cytotoxic; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2022
Remodeling the tumor microenvironment via blockade of LAIR-1 and TGF-β signaling enables PD-L1-mediated tumor eradication.
    The Journal of clinical investigation, 2022, 04-15, Volume: 132, Issue:8

    Collagens in the extracellular matrix (ECM) provide a physical barrier to tumor immune infiltration, while also acting as a ligand for immune inhibitory receptors. Transforming growth factor-β (TGF-β) is a key contributor to shaping the ECM by stimulating the production and remodeling of collagens. TGF-β activation signatures and collagen-rich environments have both been associated with T cell exclusion and lack of responses to immunotherapy. Here, we describe the effect of targeting collagens that signal through the inhibitory leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) in combination with blockade of TGF-β and programmed cell death ligand 1 (PD-L1). This approach remodeled the tumor collagenous matrix, enhanced tumor infiltration and activation of CD8+ T cells, and repolarized suppressive macrophage populations, resulting in high cure rates and long-term tumor-specific protection across murine models of colon and mammary carcinoma. The results highlight the advantage of direct targeting of ECM components in combination with immune checkpoint blockade therapy.

    Topics: Animals; B7-H1 Antigen; Immunotherapy; Ligands; Mice; Neoplasms; Receptors, Immunologic; Transforming Growth Factor beta; Tumor Microenvironment

2022
Spatial CRISPR genomics identifies regulators of the tumor microenvironment.
    Cell, 2022, 03-31, Volume: 185, Issue:7

    While CRISPR screens are helping uncover genes regulating many cell-intrinsic processes, existing approaches are suboptimal for identifying extracellular gene functions, particularly in the tissue context. Here, we developed an approach for spatial functional genomics called Perturb-map. We applied Perturb-map to knock out dozens of genes in parallel in a mouse model of lung cancer and simultaneously assessed how each knockout influenced tumor growth, histopathology, and immune composition. Moreover, we paired Perturb-map and spatial transcriptomics for unbiased analysis of CRISPR-edited tumors. We found that in Tgfbr2 knockout tumors, the tumor microenvironment (TME) was converted to a fibro-mucinous state, and T cells excluded, concomitant with upregulated TGFβ and TGFβ-mediated fibroblast activation, indicating that TGFβ-receptor loss on cancer cells increased TGFβ bioavailability and its immunosuppressive effects on the TME. These studies establish Perturb-map for functional genomics within the tissue at single-cell resolution with spatial architecture preserved and provide insight into how TGFβ responsiveness of cancer cells can affect the TME.

    Topics: Animals; Clustered Regularly Interspaced Short Palindromic Repeats; Genomics; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
Fast Quantitation of TGF-β Signaling Using Adenoviral Reporter.
    Methods in molecular biology (Clifton, N.J.), 2022, Volume: 2488

    The transforming growth factor-β (TGF-β) is a multifunctional cytokine critical for embryogenesis and tissue homeostasis. Alterations in TGF-β signaling pathway are observed in several types of malignant tumors and often related with cancer progression and metastasis. TGF-β signaling is transduced across the plasma membrane after ligand-receptor binding and consequent phosphorylation of the intracellular effectors SMAD2/3 by TGF-β receptors. Phosphorylated SMAD2/3 accumulates in the nucleus after complex formation with SMAD4 to act as transcription factors and regulate the expression of genes critically associated with cell proliferation and differentiation. Traditional methodologies used to assess TGF-β signaling pathway lack accuracy and/or show poor scalability, limiting in vitro experiments and almost excluding their use in vivo. Here, we describe a fast method to quantitate TGF-β signaling pathway activity in vitro and in vivo by using adenoviral reporters. Its implementation in vitro allows quantitating cell response to TGF-β at concentrations as low as pictograms/mL. Additionally, the use of an in vivo imaging system (IVIS) enables quantitating and monitoring TGF-β signaling pathway activity over time during cancer progression, eliminating the requirement of animal euthanasia at multiple time points for this purpose. Importantly, this protocol has been consistently used in different models and effectively led to the visualization and measurement of TGF-β activity levels. Improving the sensitivity, specificity, and scalability of methods focused on characterizing this and other molecular pathways will result in a better understanding of their biology in physiological and pathological processes.

    Topics: Animals; Neoplasms; Phosphorylation; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2022
NLGP regulates RGS5-TGFβ axis to promote pericyte-dependent vascular normalization during restricted tumor growth.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2022, Volume: 36, Issue:5

    Altered RGS5-associated intracellular pericyte signaling and its abnormal crosstalk with endothelial cells (ECs) result chaotic tumor-vasculature, prevent effective drug delivery, promote immune-evasion and many more to ensure ultimate tumor progression. Moreover, the frequency of lethal-RGS5

    Topics: Animals; CD8-Positive T-Lymphocytes; Endothelial Cells; Glycoproteins; Mice; Neoplasms; Pericytes; Phosphatidylinositol 3-Kinases; RGS Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2022
Vestigial-like family member 3 stimulates cell motility by inducing high-mobility group AT-hook 2 expression in cancer cells.
    Journal of cellular and molecular medicine, 2022, Volume: 26, Issue:9

    Vestigial-like family member 3 (VGLL3) is a cofactor for TEA domain transcription factors (TEADs). Although VGLL3 is known to be highly expressed and stimulate cell proliferation in mesenchymal cancer cells, its involvement in mesenchymal phenotypes is largely unknown. In this study, we found that VGLL3 promotes epithelial-to-mesenchymal transition (EMT)-like phenotypic changes. We found that A549 human lung cancer cells stably expressing VGLL3 exhibit spindle-like morphological changes, reduction in the epithelial marker E-cadherin and induction of the mesenchymal marker Snail. Notably, VGLL3-expressing cells exhibited enhanced motility. The DNA-binding protein high-mobility group AT-hook 2 (HMGA2) was found to be a target of the VGLL3-TEAD4 complex, and HMGA2 knockdown repressed EMT-like phenotypic changes in VGLL3-expressing cells. VGLL3-dependent phenotypic changes are involved in transforming growth factor-β (TGF-β)-induced EMT progression. VGLL3 or HMGA2 knockdown repressed the motility of the mesenchymal breast cancer MDA-MB-231 cells. Importantly, high levels of VGLL3 expression were shown to have a positive correlation with poor prognosis in various human cancers, such as breast, colon, ovarian, head and neck, pancreatic, renal, gastric and cervical cancers. These results suggest that VGLL3 promotes EMT-like cell motility by inducing HMGA2 expression and accelerates cancer progression.

    Topics: Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Family; Neoplasms; Signal Transduction; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2022
TGF-β signaling in myeloproliferative neoplasms contributes to myelofibrosis without disrupting the hematopoietic niche.
    The Journal of clinical investigation, 2022, 06-01, Volume: 132, Issue:11

    Myeloproliferative neoplasms (MPNs) are associated with significant alterations in the bone marrow microenvironment that include decreased expression of key niche factors and myelofibrosis. Here, we explored the contribution of TGF-β to these alterations by abrogating TGF-β signaling in bone marrow mesenchymal stromal cells. Loss of TGF-β signaling in Osx-Cre-targeted MSCs prevented the development of myelofibrosis in both MPLW515L and Jak2V617F models of MPNs. In contrast, despite the absence of myelofibrosis, loss of TGF-β signaling in mesenchymal stromal cells did not rescue the defective hematopoietic niche induced by MPLW515L, as evidenced by decreased bone marrow cellularity, hematopoietic stem/progenitor cell number, and Cxcl12 and Kitlg expression, and the presence of splenic extramedullary hematopoiesis. Induction of myelofibrosis by MPLW515L was intact in Osx-Cre Smad4fl/fl recipients, demonstrating that SMAD4-independent TGF-β signaling mediates the myelofibrosis phenotype. Indeed, treatment with a c-Jun N-terminal kinase (JNK) inhibitor prevented the development of myelofibrosis induced by MPLW515L. Together, these data show that JNK-dependent TGF-β signaling in mesenchymal stromal cells is responsible for the development of myelofibrosis but not hematopoietic niche disruption in MPNs, suggesting that the signals that regulate niche gene expression in bone marrow mesenchymal stromal cells are distinct from those that induce a fibrogenic program.

    Topics: Bone Marrow; Humans; Myeloproliferative Disorders; Neoplasms; Primary Myelofibrosis; Transforming Growth Factor beta; Tumor Microenvironment

2022
Ets family proteins regulate the EMT transcription factors Snail and ZEB in cancer cells.
    FEBS open bio, 2022, Volume: 12, Issue:7

    The epithelial-mesenchymal transition (EMT) is a crucial morphological event that occurs during epithelial tumor progression. Snail and ZEB1/2 (ZEB1 and ZEB2), known as EMT transcription factors, are key regulators of this transition. ZEB1/2 are positively correlated with EMT phenotypes and the aggressiveness of cancers. On the contrary, Snail is also correlated with the aggressiveness of cancers, but is not correlated with the expression of EMT marker proteins. Snail is induced by transforming growth factor-β (TGF-β), a well-known inducer of EMT, in various cancer cells. Interestingly, Snail induction by TGF-β is markedly enhanced by active Ras signals. Thus, cancer cells harboring an active Ras mutation exhibit a drastic induction of Snail by TGF-β alone. Here, we found that members of the E26 transformation-specific (Ets) transcription factor family, Ets1 and Ets2, contribute to the upregulation of both Snail and ZEB1/2. Snail induction by TGF-β and active Ras is dramatically inhibited using siRNAs against both Ets1 and Ets2 together, but not on their own; in addition, siRNAs against both Ets1 and Ets2 also downregulate the constitutive expression of Snail and ZEB1/2 in cancer cells. Examination of several alternatively spliced variants of Ets1 revealed that p54-Ets1, which includes exon VII, but not p42-Ets1, which excludes exon VII, regulates the expression of the EMT transcription factors, suggesting that Ets1 is a crucial molecule for regulating Snail and ZEB1/2, and thus cancer progression is mediated through post-translational modification of the exon VII domain.

    Topics: Epithelial-Mesenchymal Transition; Neoplasms; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta; Zinc Finger E-box-Binding Homeobox 1

2022
Transforming Growth Factor Beta Promotes the Expansion of Cancer Stem Cells via S1PR3 by Ligand-Independent Notch Activation.
    Biological & pharmaceutical bulletin, 2022, Volume: 45, Issue:5

    Growing evidence suggests that cancer originates from cancer stem cells (CSCs), which can be identified by aldehyde dehydrogenase (ALDH) activity-based flow cytometry. However, the regulation of CSC growth is not fully understood. In the present study, we investigated the effects of Transforming Growth Factor-β (TGFβ) in breast CSC expansion. Stimulation with TGFβ increased the ALDH-positive breast CSC population via the phosphorylation of sphingosine kinase 1 (SphK1), a sphingosine-1-phosphate (S1P)-producing enzyme, and subsequent S1P-mediated S1P receptor 3 (S1PR3) activation. These data suggest that TGFβ promotes breast CSC expansion via the ALK5/SphK1/S1P/S1PR3 signaling pathway. Our findings provide new insights into the role of TGFβ in the regulation of CSCs.

    Topics: Ligands; Neoplasms; Neoplastic Stem Cells; Phosphorylation; Signal Transduction; Transforming Growth Factor beta

2022
Roles of DKK3 in cellular adhesion, motility, and invasion through extracellular interaction with TGFBI.
    The FEBS journal, 2022, Volume: 289, Issue:20

    Dickkopf-related protein (DKK) 3, a member of the DKK family, is a secreted glycoprotein that acts as a modulator of Wnt signaling in organogenesis and carcinogenesis. Recent studies have demonstrated that DKK3 has a variety of functions, suggesting that it plays roles not only in tumorigenesis, but also tumor neovascularization, prostatic acinus formation, cardiac vascular remodeling, renal and cardiac fibrosis, and immunological activity. The function of DKK3 is therefore of great interest, but details of the receptors and mechanisms involved have remained unclear. Here, we focused on the extracellular function of DKK3 as a secreted protein, and identified transforming growth factor beta induced protein ig-h3 (TGFBI) as a secreted protein interacting with DKK3. To investigate the function of these secreted proteins, we employed an in vitro cell model involving human hepatocellular carcinoma cells, human embryonic kidney cells, or non-neoplastic hepatocyte cells. We showed that DKK3 functions as an extracellular matrix-like molecule supporting adhesion, motility, and invasion, and that its interaction with TGFBI inhibits the functions of secreted DKK3 in cells expressing both proteins. These results suggest that this extracellular interaction between DKK3 and TGFBI modulates cell adhesion and motility through focal adhesion kinase signaling, and that this might serve as a potential therapeutic target in the context of cancer invasion and metastasis.

    Topics: Adaptor Proteins, Signal Transducing; Carcinogenesis; Cell Adhesion; Focal Adhesion Protein-Tyrosine Kinases; Glycoproteins; Humans; Neoplasms; Transforming Growth Factor beta; Wnt Signaling Pathway

2022
PHF13 epigenetically activates TGFβ driven epithelial to mesenchymal transition.
    Cell death & disease, 2022, 05-21, Volume: 13, Issue:5

    Epigenetic alteration is a pivotal factor in tumor metastasis. PHD finger protein 13 (PHF13) is a recently identified epigenetic reader of H3K4me2/3 that functions as a transcriptional co-regulator. In this study, we demonstrate that PHF13 is required for pancreatic-cancer-cell growth and metastasis. Integrative analysis of transcriptome and epigenetic profiles provide further mechanistic insights into the epigenetic regulation of genes associated with cell metastasis during the epithelial-to-mesenchymal transition (EMT) induced by transforming growth factor β (TGFβ). Our data suggest PHF13 depletion impairs activation of TGFβ stimulated genes and correlates with a loss of active epigenetic marks (H3K4me3 and H3K27ac) at these genomic regions. These observations argue for a dependency of TGFβ target activation on PHF13. Furthermore, PHF13-dependent chromatin regions are enriched in broad H3K4me3 domains and super-enhancers, which control genes critical to cancer-cell migration and invasion, such as SNAI1 and SOX9. Overall, our data indicate a functional and mechanistic correlation between PHF13 and EMT.

    Topics: Cell Line, Tumor; Chromatin; DNA-Binding Proteins; Epigenesis, Genetic; Epithelial Cells; Epithelial-Mesenchymal Transition; Humans; Neoplasms; Transcription Factors; Transforming Growth Factor beta

2022
CIS and TGF-β regulatory pathways influence immunity to bacterial infection.
    Immunology, 2022, Volume: 167, Issue:1

    Immunotherapy has revolutionized cancer therapy by reactivating tumour-resident cytotoxic lymphocytes. More recently, immunotherapy has emerged to restore immunity against infectious agents, including bacterial infections. Immunotherapy primarily targets inhibitory pathways in T cells, however interest in other effector populations, such as natural killer (NK) cells, is growing. We have previously discovered that NK cell metabolism, proliferation and activation can be neutralized through the immunosuppressive transforming growth factor (TGF)-β pathway by inducing plasticity of NK cells and differentiation into innate lymphoid cell (ILC)1-like subsets. NK cells are also regulated through cytokine-inducible SH2-containing protein (CIS), which is induced by interleukin (IL)-15 and is a potent intracellular checkpoint suppressing NK cell survival and function. Targeting these two distinct pathways to restore NK cell function has shown promise in cancer models, but their application in bacterial infection remains unknown. Here, we investigate whether enhancement of NK cell function can improve anti-bacterial immunity, using Salmonella Typhimurium as a model. We identified conversion of NK cells to ILC1-like for the first time in the context of bacterial infection, where TGF-β signalling contributed to this plasticity. Future study should focus on identifying further drivers of ILC1 plasticity and its functional implication in bacterial infection model. We further describe that CIS-deficient mice displayed enhanced pro-inflammatory function and dramatically enhanced anti-bacterial immunity. Inhibition of CIS may present as a viable therapeutic option to enhance immunity towards bacterial infection.

    Topics: Animals; Bacterial Infections; Immunity, Innate; Killer Cells, Natural; Mice; Neoplasms; Transforming Growth Factor beta

2022
Effect of Eribulin on Angiogenesis and the Expression of Endothelial Adhesion Molecules.
    Anticancer research, 2022, Volume: 42, Issue:6

    Tumor vasculature is an important component of the tumor microenvironment and deeply affects anticancer immune response. Eribulin is a non-taxane inhibitor of the mitotic spindle. However, off-target effects interfering with the tumor vasculature have been reported. The mechanisms responsible of this effect are still unclear.. We designed an in vitro study to investigate the effect of eribulin, with or without TGF-β, on neo-angiogenesis, and on the expression of the adhesion molecules ICAM-1 and VCAM-1. We also investigated the effects of paclitaxel and vinorelbine under the same experimental conditions.. Eribulin up-regulated the epithelial markers VE-cadherin and CD-31 in HUVEC and inhibited tube formation in HUVEC cells cultured in Matrigel. The drug effectively arrested tube formation even in the presence of TGF-β and counteracted the TGF-β-induced change in cell shape from the endothelial cobblestone-like morphology to an elongated spindle-shaped morphology. We also observed that eribulin was able to upregulate ICAM-1 and to counteract its down-regulation induced by TGF-β.. Eribulin exerts different off-label effects: increases vascular remodeling, counteracts the endothelial-to-mesenchymal transition (EndMT) mediated by TGF-β and promotes tumor infiltration by immune cells via increasing the expression of ICAM-1 and transcription of CD31 and VE-cadherin. Moreover, eribulin was able to inhibit vasculature remodeling and the induction of EndMT mediated by TGF-β better than vinorelbine and paclitaxel. The effects observed in this study might have important therapeutic consequence if the drug is combined with immunotherapy.

    Topics: Furans; Humans; Intercellular Adhesion Molecule-1; Ketones; Neoplasms; Paclitaxel; Transforming Growth Factor beta; Tumor Microenvironment; Vinorelbine

2022
ERK-Smurf1-RhoA signaling is critical for TGFβ-drived EMT and tumor metastasis.
    Life science alliance, 2022, Volume: 5, Issue:10

    Epithelial-mesenchymal transition (EMT) has fundamental roles in various biological processes. However, there are still questions pending in this fast-moving field. Here we report that in TGFβ-induced EMT, ERK-mediated Smurf1 phosphorylation is a prerequisite step for RhoA degradation and the consequent mesenchymal state achievement. Upon TGFβ treatment, activated ERK phosphorylates Thr223 of Smurf1, a member of HECT family E3 ligase, to promote Smurf1-mediated polyubiquitination and degradation of RhoA, thereby leading to cell skeleton rearrangement and EMT. Blockade of phosphorylation of Smurf1 inhibits TGFβ-induced EMT, and accordingly, dramatically blocks lung metastasis of murine breast cancer in mice. Hence, our study reveals an unknown role of ERK in TGFβ-induced EMT and points out a potential strategy in therapeutic intervention.

    Topics: Animals; Biological Phenomena; Epithelial-Mesenchymal Transition; Mice; Neoplasms; Transforming Growth Factor beta; Ubiquitin-Protein Ligases

2022
SMAD4 Controls Cancer Cell Metabolism by Regulating Methylmalonic Aciduria Cobalamin Deficiency (cbl) B Type.
    Molecules and cells, 2022, Jun-30, Volume: 45, Issue:6

    Suppressor of mothers against decapentaplegic homolog (SMAD) 4 is a pluripotent signaling mediator that regulates myriad cellular functions, including cell growth, cell division, angiogenesis, apoptosis, cell invasion, and metastasis, through transforming growth factor β (TGF-β)-dependent and -independent pathways. SMAD4 is a critical modulator in signal transduction and functions primarily as a transcription factor or cofactor. Apart from being a DNA-binding factor, the additional SMAD4 mechanisms in tumor suppression remain elusive. We previously identified methyl malonyl aciduria cobalamin deficiency B type (MMAB) as a critical SMAD4 binding protein using a proto array analysis. This study confirmed the interaction between SMAD4 and MMAB using bimolecular fluorescence complementation (BiFC) assay, proximity ligation assay (PLA), and conventional immunoprecipitation. We found that transient SMAD4 overexpression down-regulates MMAB expression via a proteasome-dependent pathway. SMAD4-MMAB interaction was independent of TGF-β signaling. Finally, we determined the effect of MMAB downregulation on cancer cells. siRNA-mediated knockdown of MMAB affected cancer cell metabolism in HeLa cells by decreasing ATP production and glucose consumption as well as inducing apoptosis. These findings suggest that SMAD4 controls cancer cell metabolism by regulating MMAB.

    Topics: Amino Acid Metabolism, Inborn Errors; Cell Line, Tumor; HeLa Cells; Humans; Neoplasms; Smad4 Protein; Transforming Growth Factor beta; Vitamin B 12

2022
Ursodeoxycholic acid reduces antitumor immunosuppression by inducing CHIP-mediated TGF-β degradation.
    Nature communications, 2022, 06-14, Volume: 13, Issue:1

    TGF-β is essential for inducing systemic tumor immunosuppression; thus, blocking TGF-β can greatly enhance antitumor immunity. However, there are still no effective TGF-β inhibitors in clinical use. Here, we show that the clinically approved compound ursodeoxycholic acid (UDCA), by degrading TGF-β, enhances antitumor immunity through restraining Treg cell differentiation and activation in tumor-bearing mice. Furthermore, UDCA synergizes with anti-PD-1 to enhance antitumor immunity and tumor-specific immune memory in tumor-bearing mice. UDCA phosphorylates TGF-β at T282 site via TGR5-cAMP-PKA axis, causing increased binding of TGF-β to carboxyl terminus of Hsc70-interacting protein (CHIP). Then, CHIP ubiquitinates TGF-β at the K315 site, initiating p62-dependent autophagic sorting and subsequent degradation of TGF-β. Notably, results of retrospective analysis shows that combination therapy with anti-PD-1 or anti-PD-L1 and UDCA has better efficacy in tumor patients than anti-PD-1 or anti-PD-L1 alone. Thus, our results show a mechanism for TGF-β regulation and implicate UDCA as a potential TGF-β inhibitor to enhance antitumor immunity.

    Topics: Animals; Cell Line, Tumor; Humans; Immunosuppression Therapy; Mice; Neoplasms; Retrospective Studies; Transforming Growth Factor beta; Ursodeoxycholic Acid

2022
TGFβ and CIS Inhibition Overcomes NK-cell Suppression to Restore Antitumor Immunity.
    Cancer immunology research, 2022, 09-01, Volume: 10, Issue:9

    Antibodies targeting "immune checkpoints" have revolutionized cancer therapy by reactivating tumor-resident cytotoxic lymphocytes, primarily CD8+ T cells. Interest in targeting analogous pathways in other cytotoxic lymphocytes is growing. Natural killer (NK) cells are key to cancer immunosurveillance by eradicating metastases and driving solid tumor inflammation. NK-cell antitumor function is dependent on the cytokine IL15. Ablation of the IL15 signaling inhibitor CIS (Cish) enhances NK-cell antitumor immunity by increasing NK-cell metabolism and persistence within the tumor microenvironment (TME). The TME has also been shown to impair NK-cell fitness via the production of immunosuppressive transforming growth factor β (TGFβ), a suppression which occurs even in the presence of high IL15 signaling. Here, we identified an unexpected interaction between CIS and the TGFβ signaling pathway in NK cells. Independently, Cish- and Tgfbr2-deficient NK cells are both hyperresponsive to IL15 and hyporesponsive to TGFβ, with dramatically enhanced antitumor immunity. Remarkably, when both these immunosuppressive genes are simultaneously deleted in NK cells, mice are largely resistant to tumor development, suggesting that combining suppression of these two pathways might represent a novel therapeutic strategy to enhance innate anticancer immunity.

    Topics: Animals; Cell Line, Tumor; Interleukin-15; Killer Cells, Natural; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
Cimetidine and Ibuprofen Modulate T Cell Responses in a Mouse Model of Breast Cancer.
    Asian Pacific journal of cancer prevention : APJCP, 2022, Jun-01, Volume: 23, Issue:6

    Cimetidine and ibuprofen exhibit immunomodulatory effects as an antagonist of histamine H2 receptor, and a cyclooxygenase inhibitor, respectively. Here, the effects of cimetidine and ibuprofen on some effector T cell-related parameters were investigated using a breast cancer (BC) model. BC was established in Balb/c mice using the 4T1 cell line. On day 10 after tumor induction, the BC-bearing mice were classified into four groups and treated with PBS, cimetidine (20 mg/kg), ibuprofen (20 mg/kg) or a combination of "cimetidine + ibuprofen" via intraperitoneal injection (daily from days 11 to 30). The mice were sacrificed on day 31 and the frequency of splenic Th1 and Treg cells, plasma IFN-γ and TGF-β levels, and intra-tumoral T-bet, GATA3, FOXP3 and RORγt expressions were detected using flowcytometry, ELISA and real-time-PCR, respectively. In untreated cancerous mice, the percentage of splenic Th1 cells and plasma IFN-γ levels were lower (P<0.003 and P<0.01, respectively), whereas the percentage of splenic Treg cells and plasma TGF-β levels were higher than in healthy mice (P<0.04 and P<0.005, respectively). Treatment of BC-bearing mice with cimetidine, ibuprofen or both drugs promoted the frequency of Th1 cells (P<0.05, P<0.007 and P<0.005, respectively) as well as IFN-γ levels (P<0.004, P<0.0001 and P<0.03, respectively), while reduced the frequencies of Treg cells (P<0.02, P<0.03 and P<0.01, respectively), TGF-β levels (P<0.006, P<0.02 and P<0.002, respectively), intra-tumoral expression of FOXP3 (P<0.006, P<0.005 and P<0.005, respectively), and intra-tumoral expression of RORγt (P<0.04, P<0.03 and P<0.05, respectively) compared with untreated BC mice. The "cimetidine + ibuprofen"-treated mice displayed greater T-bet expression than the un-treated mice (P<0.006). Cimetidine and/or ibuprofen-treated BC-bearing mice exhibited reduced intra-tumoral expression of GATA3 compared with the untreated BC mice, but the differences were not significant. Cimetidine and ibuprofen correct some effector T cell-related parameters in cancerous mice. Immunotherapeutic potentials cimetidine and ibuprofen in cancers need investigations.

    Topics: Animals; Cimetidine; Disease Models, Animal; Forkhead Transcription Factors; Ibuprofen; Mice; Mice, Inbred BALB C; Neoplasms; Nuclear Receptor Subfamily 1, Group F, Member 3; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2022
Complex study of Cancer Morbidity and Inflammatory Markers, Presented in the Blood Serum of the Rural Population of Sachkhere District of Georgia.
    Asian Pacific journal of cancer prevention : APJCP, 2022, Jun-01, Volume: 23, Issue:6

    The purpose of the study was to compare the morbidity from cancer (expressed as incidence) to the average levels of blood serum inflammatory markers in the population of the Sachkhere region (Georgia).. healthy residents of the Sachkhere district were examined. In the blood serum samples of patients, the cytokines (IL-1α, IL-10, TGF-β, IL-12, IL-17, TNF-α, IL-6) and NOx content, as well as the total antioxidant activity of the non-enzymatic system (TAA) were determined; using light microscopy, buccal micronuclei (MnB) of epithelial cells of the oral mucosa, as indicators of chromosomal disorders, were studied.. Study results show, that cancer incidence in Sareki was statistically significantly higher as in Chorvila and Sairkhe (p=0.002; p=0.004); in Sareki inhabitant's blood serum levels of the IL-6,  NO are increased (p=0.004, p=0.05), and IL-17,  TGFβ, and IL-10 levels are decreased (p=0.010, p=0.001, p=0.033) in comparison to data in Chorvila; in Chorvila inhabitants' indicators of TAA  of blood serum and  MnB of epithelium cell levels were lower (p=0.001,p=0.045) then in Sairkhe and Sareki.. The existence of statistically reliable associations between the levels of cancer incidence in the populations of the surveyed villages and the indicators of immune and oxidative status in their virtually healthy subpopulations, with a high degree of persuasiveness, allows us to assume a close causal link between them. Clarifying the reasons for the identified patterns and their significance requires more detailed studies.

    Topics: Biomarkers; Georgia; Humans; Interleukin-10; Interleukin-17; Interleukin-6; Morbidity; Neoplasms; Rural Population; Serum; Transforming Growth Factor beta

2022
Long noncoding RNA Smyca coactivates TGF-β/Smad and Myc pathways to drive tumor progression.
    Journal of hematology & oncology, 2022, 07-06, Volume: 15, Issue:1

    Metastasis and chemoresistance are major culprits of cancer mortality, but factors contributing to these processes are incompletely understood.. Bioinformatics methods were used to identify the relations of Smyca expression to clinicopathological features of human cancers. RNA-sequencing analysis was used to reveal Smyca-regulated transcriptome. RNA pull-down and RNA immunoprecipitation were used to examine the binding of Smyca to Smad3/4 and c-Myc/Max. Chromatin immunoprecipitation and chromatin isolation by RNA purification were used to determine the binding of transcription factors and Smyca to various gene loci, respectively. Real-time RT-PCR and luciferase assay were used to examine gene expression levels and promoter activities, respectively. Xenograft mouse models were performed to evaluate the effects of Smyca on metastasis and chemoresistance. Nanoparticle-assisted gapmer antisense oligonucleotides delivery was used to target Smyca in vivo.. We identify lncRNA Smyca for its association with poor prognosis of many cancer types. Smyca potentiates metabolic reprogramming, migration, invasion, cancer stemness, metastasis and chemoresistance. Mechanistically, Smyca enhances TGF-β/Smad signaling by acting as a scaffold for promoting Smad3/Smad4 association and further serves as a Smad target to amplify/prolong TGF-β signaling. Additionally, Smyca potentiates c-Myc-mediated transcription by enhancing the recruitment of c-Myc/Max complex to a set of target promoters and c-Myc binding to TRRAP. Through potentiating TGF-β and c-Myc pathways, Smyca synergizes the Warburg effect elicited by both pathways but evades the anti-proliferative effect of TGF-β. Targeting Smyca prevents metastasis and overcomes chemoresistance.. This study uncovers a lncRNA that coordinates tumor-relevant pathways to orchestra a pro-tumor program and establishes the clinical values of Smyca in cancer prognosis and therapy.

    Topics: Animals; Humans; Mice; Neoplasms; Promoter Regions, Genetic; RNA, Long Noncoding; Transforming Growth Factor beta

2022
USP8 promotes cancer progression and extracellular vesicle-mediated CD8+ T cell exhaustion by deubiquitinating the TGF-β receptor TβRII.
    The EMBO journal, 2022, 08-16, Volume: 41, Issue:16

    TGF-β signaling is a key player in tumor progression and immune evasion, and is associated with poor response to cancer immunotherapies. Here, we identified ubiquitin-specific peptidase 8 (USP8) as a metastasis enhancer and a highly active deubiquitinase in aggressive breast tumors. USP8 acts both as a cancer stemness-promoting factor and an activator of the TGF-β/SMAD signaling pathway. USP8 directly deubiquitinates and stabilizes the type II TGF-β receptor TβRII, leading to its increased expression in the plasma membrane and in tumor-derived extracellular vesicles (TEVs). Increased USP8 activity was observed in patients resistant to neoadjuvant chemotherapies. USP8 promotes TGF-β/SMAD-induced epithelial-mesenchymal transition (EMT), invasion, and metastasis in tumor cells. USP8 expression also enables TβRII+ circulating extracellular vesicles (crEVs) to induce T cell exhaustion and chemoimmunotherapy resistance. Pharmacological inhibition of USP8 antagonizes TGF-β/SMAD signaling, and reduces TβRII stability and the number of TβRII+ crEVs to prevent CD8+ T cell exhaustion and to reactivate anti-tumor immunity. Our findings not only reveal a novel mechanism whereby USP8 regulates the cancer microenvironment but also demonstrate the therapeutic advantages of engineering USP8 inhibitors to simultaneously suppress metastasis and improve the efficacy of cancer immunotherapy.

    Topics: CD8-Positive T-Lymphocytes; Endopeptidases; Endosomal Sorting Complexes Required for Transport; Extracellular Vesicles; Humans; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment; Ubiquitin Thiolesterase

2022
WNT signaling and cancer stemness.
    Essays in biochemistry, 2022, 09-16, Volume: 66, Issue:4

    Cancer stemness, defined as the self-renewal and tumor-initiation potential of cancer stem cells (CSCs), is a cancer biology property featuring activation of CSC signaling networks. Canonical WNT signaling through Frizzled and LRP5/6 receptors is transmitted to the β-catenin-TCF/LEF-dependent transcription machinery to up-regulate MYC, CCND1, LGR5, SNAI1, IFNG, CCL28, CD274 (PD-L1) and other target genes. Canonical WNT signaling causes expansion of rapidly cycling CSCs and modulates both immune surveillance and immune tolerance. In contrast, noncanonical WNT signaling through Frizzled or the ROR1/2 receptors is transmitted to phospholipase C, Rac1 and RhoA to control transcriptional outputs mediated by NFAT, AP-1 and YAP-TEAD, respectively. Noncanonical WNT signaling supports maintenance of slowly cycling, quiescent or dormant CSCs and promotes epithelial-mesenchymal transition via crosstalk with TGFβ (transforming growth factor-β) signaling cascades, while the TGFβ signaling network induces immune evasion. The WNT signaling network orchestrates the functions of cancer-associated fibroblasts, endothelial cells and immune cells in the tumor microenvironment and fine-tunes stemness in human cancers, such as breast, colorectal, gastric and lung cancers. Here, WNT-related cancer stemness features, including proliferation/dormancy plasticity, epithelial-mesenchymal plasticity and immune-landscape plasticity, will be discussed. Porcupine inhibitors, β-catenin protein-protein interaction inhibitors, β-catenin proteolysis targeting chimeras, ROR1 inhibitors and ROR1-targeted biologics are investigational drugs targeting WNT signaling cascades. Mechanisms of cancer plasticity regulated by the WNT signaling network are promising targets for therapeutic intervention; however, further understanding of context-dependent reprogramming trajectories might be necessary to optimize the clinical benefits of WNT-targeted monotherapy and applied combination therapy for patients with cancer.

    Topics: B7-H1 Antigen; beta Catenin; Biological Products; Drugs, Investigational; Endothelial Cells; Humans; Neoplasms; Transcription Factor AP-1; Transforming Growth Factor beta; Transforming Growth Factors; Tumor Microenvironment; Type C Phospholipases; Wnt Signaling Pathway

2022
Reciprocal SOX2 regulation by SMAD1-SMAD3 is critical for anoikis resistance and metastasis in cancer.
    Cell reports, 2022, 07-26, Volume: 40, Issue:4

    Growth factors in tumor environments are regulators of cell survival and metastasis. Here, we reveal the dichotomy between TGF-β superfamily growth factors BMP and TGF-β/activin and their downstream SMAD effectors. Gene expression profiling uncovers SOX2 as a key contextual signaling node regulated in an opposing manner by BMP2, -4, and -9 and TGF-β and activin A to impact anchorage-independent cell survival. We find that SOX2 is repressed by BMPs, leading to a reduction in intraperitoneal tumor burden and improved survival of tumor-bearing mice. Repression of SOX2 is driven by SMAD1-dependent histone H3K27me3 recruitment and DNA methylation at SOX2's promoter. Conversely, TGF-β, which is elevated in patient ascites, and activin A can promote SOX2 expression and anchorage-independent survival by SMAD3-dependent histone H3K4me3 recruitment. Our findings identify SOX2 as a contextual and contrastingly regulated node downstream of TGF-β members controlling anchorage-independent survival and metastasis in ovarian cancers.

    Topics: Animals; Anoikis; Bone Morphogenetic Proteins; Histones; Mice; Neoplasms; Smad1 Protein; Smad3 Protein; SOXB1 Transcription Factors; Transforming Growth Factor beta

2022
HNF4A Defines Molecular Subtypes and Vulnerability to Transforming Growth Factor β-Pathway Targeted Therapies in Cancers of the Distal Esophagus.
    Gastroenterology, 2022, Volume: 163, Issue:5

    Topics: Esophagus; Hepatocyte Nuclear Factor 4; Humans; Neoplasms; Transforming Growth Factor beta

2022
TGF-β regulates the stem-like state of PD-1+ TCF-1+ virus-specific CD8 T cells during chronic infection.
    The Journal of experimental medicine, 2022, 10-03, Volume: 219, Issue:10

    Recent studies have defined a novel population of PD-1+ TCF-1+ stem-like CD8 T cells in chronic infections and cancer. These quiescent cells reside in lymphoid tissues, are critical for maintaining the CD8 T cell response under conditions of persistent antigen, and provide the proliferative burst after PD-1 blockade. Here we examined the role of TGF-β in regulating the differentiation of virus-specific CD8 T cells during chronic LCMV infection of mice. We found that TGF-β signaling was not essential for the generation of the stem-like CD8 T cells but was critical for maintaining the stem-like state and quiescence of these cells. TGF-β regulated the unique transcriptional program of the stem-like subset, including upregulation of inhibitory receptors specifically expressed on these cells. TGF-β also promoted the terminal differentiation of exhausted CD8 T cells by suppressing the effector-associated program. Together, the absence of TGF-β signaling resulted in significantly increased accumulation of effector-like CD8 T cells. These findings have implications for immunotherapies in general and especially for T cell therapy against chronic infections and cancer.

    Topics: Animals; CD8-Positive T-Lymphocytes; Lymphocytic Choriomeningitis; Lymphocytic choriomeningitis virus; Mice; Neoplasms; Persistent Infection; Programmed Cell Death 1 Receptor; Transforming Growth Factor beta

2022
Local anesthetic lidocaine-inducible gene, growth differentiation factor-15 suppresses the growth of cancer cell lines.
    Scientific reports, 2022, 08-25, Volume: 12, Issue:1

    Administration of local anesthetics, such as lidocaine, in the perioperative period improves outcomes of cancer patients. However, its precise mechanism is still unresolved. The growth of human cancer cell lines, including HeLa cells, are suppressed by lidocaine treatment. We identified that growth differentiation factor-15 (GDF-15) was commonly upregulated in lidocaine-treated cancer cell lines. GDF-15 is a divergent member of the transforming growth factor-β (TGF-β) superfamily and it is produced as an unprocessed pro-protein form and then cleaved to generate a mature form. In lidocaine-treated HeLa cells, increased production of GDF-15 in the endoplasmic reticulum (ER) was observed and unprocessed pro-protein form of GDF-15 was secreted extracellularly. Further, lidocaine induced apoptosis and apoptosis-inducible Tribbles homologue 3 (TRIB3) was also commonly upregulated in lidocaine-treated cancer cell lines. In addition, transcription factor C/EBP homologous protein (CHOP), which is a positive regulator of not only GDF-15 but TRIB3 was also induced by lidocaine. Lidocaine-induced growth suppression and apoptosis was suppressed by knockdown of GDF-15 or TRIB3 expression by small interference RNA (siRNA). These observations suggest that lidocaine suppresses the growth of cancer cells through increasing GDF-15 and TRIB3 expression, suggesting its potential application as cancer therapy.

    Topics: Anesthetics, Local; Growth Differentiation Factor 15; Growth Differentiation Factor 9; HeLa Cells; Humans; Lidocaine; Neoplasms; Transforming Growth Factor beta

2022
Model-informed approach for risk management of bleeding toxicities for bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1.
    Cancer chemotherapy and pharmacology, 2022, Volume: 90, Issue:4

    Bintrafusp alfa (BA) is a bifunctional fusion protein composed of the extracellular domain of the transforming growth factor-β (TGF-β) receptor II fused to a human immunoglobulin G1 antibody blocking programmed death ligand 1 (PD-L1). The recommended phase 2 dose (RP2D) was selected based on phase 1 efficacy, safety, and pharmacokinetic (PK)-pharmacodynamic data, assuming continuous inhibition of PD-L1 and TGF-β is required. Here, we describe a model-informed dose modification approach for risk management of BA-associated bleeding adverse events (AEs).. The PK and AE data from studies NCT02517398, NCT02699515, NCT03840915, and NCT04246489 (n = 936) were used. Logistic regression analyses were conducted to evaluate potential relationships between bleeding AEs and BA time-averaged concentration (C. The probability of bleeding AEs increased with increasing C. A pragmatic model-informed approach for management of bleeding AEs was implemented in ongoing clinical trials of BA. This approach is expected to improve benefit-risk profile; however, its effectiveness will need to be evaluated based on safety data generated after implementation.

    Topics: B7-H1 Antigen; Clinical Studies as Topic; Hemorrhage; Humans; Immunologic Factors; Neoplasms; Risk Management; Transforming Growth Factor beta

2022
Redistribution of the SWI/SNF Complex Dictates Coordinated Transcriptional Control over Epithelial-Mesenchymal Transition of Normal Breast Cells through TGF-β Signaling.
    Cells, 2022, 08-24, Volume: 11, Issue:17

    Therapeutic targets in cancer cells defective for the tumor suppressor ARID1A are fundamentals of synthetic lethal strategies. However, whether modulating ARID1A function in premalignant breast epithelial cells could be exploited to reduce carcinogenic potential remains to be elucidated. In search of chromatin-modulating mechanisms activated by anti-proliferative agents in normal breast epithelial (HME-hTert) cells, we identified a distinct pattern of genome-wide H3K27 histone acetylation marks characteristic for the combined treatment by the cancer preventive rexinoid bexarotene (Bex) and carvedilol (Carv). Among these marks, several enhancers functionally linked to TGF-β signaling were enriched for ARID1A and Brg1, subunits within the SWI/SNF chromatin-remodeling complex. The recruitment of ARID1A and Brg1 was associated with the suppression of TGFBR2, KLF4, and FoxQ1, and the induction of BMP6, while the inverse pattern ensued upon the knock-down of ARID1A. Bex+Carv treatment resulted in fewer cells expressing N-cadherin and dictated a more epithelial phenotype. However, the silencing of ARID1A expression reversed the ability of Bex and Carv to limit epithelial-mesenchymal transition. The nuclear levels of SMAD4, a canonical mediator of TGF-β action, were more effectively suppressed by the combination than by TGF-β. In contrast, TGF-β treatment exceeded the ability of Bex+Carv to lower nuclear FoxQ1 levels and induced markedly higher E-cadherin positivity, indicating a target-selective antagonism of Bex+Carv to TGF-β action. In summary, the chromatin-wide redistribution of ARID1A by Bex and Carv treatment is instrumental in the suppression of genes mediating TGF-β signaling, and, thus, the morphologic reprogramming of normal breast epithelial cells. The concerted engagement of functionally linked targets using low toxicity clinical agents represents an attractive new approach for cancer interception.

    Topics: Cadherins; Chromatin; Chromatin Assembly and Disassembly; Epithelial-Mesenchymal Transition; Forkhead Transcription Factors; Humans; Neoplasms; Transforming Growth Factor beta

2022
Selective targeting of GARP-LTGFβ axis in the tumor microenvironment augments PD-1 blockade via enhancing CD8
    Journal for immunotherapy of cancer, 2022, Volume: 10, Issue:9

    Immune checkpoint blockade (ICB) has revolutionized cancer immunotherapy. However, most patients with cancer fail to respond clinically. One potential reason is the accumulation of immunosuppressive transforming growth factor β (TGFβ) in the tumor microenvironment (TME). TGFβ drives cancer immune evasion in part by inducing regulatory T cells (Tregs) and limiting CD8. We investigated the role of GARP in human patients with cancer by analyzing existing large databases. In addition, we generated and humanized an anti-GARP monoclonal antibody and evaluated its antitumor efficacy and underlying mechanisms of action in murine models of cancer.. We demonstrate that GARP overexpression in human cancers correlates with a tolerogenic TME and poor clinical response to ICB, suggesting GARP blockade may improve cancer immunotherapy. We report on a unique anti-human GARP antibody (named PIIO-1) that specifically binds the ligand-interacting domain of all latent TGFβ isoforms. PIIO-1 lacks recognition of GARP-TGFβ complex on platelets. Using human. GARP contributes to multiple aspects of immune resistance in cancer. Anti-human GARP antibody PIIO-1 is an efficacious and safe strategy to block GARP-mediated LTGFβ activation, enhance CD8

    Topics: Animals; CD8-Positive T-Lymphocytes; Glycoproteins; Humans; Immune Checkpoint Inhibitors; Membrane Proteins; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
Multiscale modeling of collective cell migration elucidates the mechanism underlying tumor-stromal interactions in different spatiotemporal scales.
    Scientific reports, 2022, 09-28, Volume: 12, Issue:1

    Metastasis is the pathogenic spread of cancer cells from a primary tumor to a secondary site which happens at the late stages of cancer. It is caused by a variety of biological, chemical, and physical processes, such as molecular interactions, intercellular communications, and tissue-level activities. Complex interactions of cancer cells with their microenvironment components such as cancer associated fibroblasts (CAFs) and extracellular matrix (ECM) cause them to adopt an invasive phenotype that promotes tumor growth and migration. This paper presents a multiscale model for integrating a wide range of time and space interactions at the molecular, cellular, and tissue levels in a three-dimensional domain. The modeling procedure starts with presenting nonlinear dynamics of cancer cells and CAFs using ordinary differential equations based on TGFβ, CXCL12, and LIF signaling pathways. Unknown kinetic parameters in these models are estimated using hybrid unscented Kalman filter and the models are validated using experimental data. Then, the principal role of CAFs on metastasis is revealed by spatial-temporal modeling of circulating signals throughout the TME. At this stage, the model has evolved into a coupled ODE-PDE system that is capable of determining cancer cells' status in one of the quiescent, proliferating or migratory conditions due to certain metastasis factors and ECM characteristics. At the tissue level, we consider a force-based framework to model the cancer cell proliferation and migration as the final step towards cancer cell metastasis. The ability of the multiscale model to depict cancer cells' behavior in different levels of modeling is confirmed by comparing its outputs with the results of RT PCR and wound scratch assay techniques. Performance evaluation of the model indicates that the proposed multiscale model can pave the way for improving the efficiency of therapeutic methods in metastasis prevention.

    Topics: Cancer-Associated Fibroblasts; Cell Communication; Cell Movement; Humans; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
Combination of oral STING agonist MSA-2 and anti-TGF-β/PD-L1 bispecific antibody YM101: a novel immune cocktail therapy for non-inflamed tumors.
    Journal of hematology & oncology, 2022, 10-08, Volume: 15, Issue:1

    Non-inflamed tumors, including immune-excluded and immune-desert tumors, are commonly resistant to anti-PD-1/PD-L1 (α-PD-1/PD-L1) therapy. Our previous study reported the potent antitumor activity of anti-TGF-β/PD-L1 bispecific antibody YM101 in immune-excluded tumors. However, YM101 had limited antitumor activity in immune-desert models. MSA-2 is a novel oral stimulator of interferon genes (STING) agonist, which activates the innate immune system and may synergize with YM101 in overcoming immunotherapy resistance.. The dose-dependent effect of MSA-2 on STING signaling was determined by interferon-β level. The maturation and function of dendritic cell (DC) were measured by flow cytometry, RNA-seq, one-way mixed lymphocyte reaction (MLR), OVA peptide pulse, and cytokine/chemokine detection. The synergistic effect between MSA-2 and YM101 was assessed by one-way MLR. The macrophage activation was measured by flow cytometry and cytokine/chemokine detection. The in vivo antitumor activity of MSA-2 combined with YM101 was explored in syngeneic murine tumor models. After treatments, the alterations in the tumor microenvironment (TME) were detected by flow cytometry, immunohistochemistry staining, immunofluorescence staining, RNA-seq, and single-cell RNA-seq (scRNA-seq).. MSA-2 could promote the maturation and antigen presentation capability of murine DC. In the one-way MLR assay, MSA-2 synergized with YM101 in enhancing naive T cell activation. Moreover, MSA-2 stimulated the classical activation of macrophage, without significant influence on alternative activation. Further in vivo explorations showed that MSA-2 increased multiple proinflammatory cytokines and chemokines in the TME. MSA-2 combined with YM101 remarkedly retarded tumor growth in immune-excluded and immune-desert models, with superior antitumor activity to monotherapies. Flow cytometry, bulk RNA-seq, and scRNA-seq assays indicated that the combination therapy simultaneously boosted the innate and adaptive immunity, promoted antigen presentation, improved T cell migration and chemotaxis, and upregulated the numbers and activities of tumor-infiltrating lymphocytes.. Our results demonstrate that MSA-2 synergizes with YM101 in boosting antitumor immunity. This immune cocktail therapy effectively overcomes immunotherapy resistance in immune-excluded and immune-desert models.

    Topics: Animals; Antibodies, Bispecific; B7-H1 Antigen; Cell Line, Tumor; Cytokines; Humans; Immunotherapy; Interferon-beta; Interferons; Membrane Proteins; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2022
Computational analysis of prodomain cysteines in human TGF-β proteins reveals frequent loss of disulfide-dependent regulation in tumors.
    G3 (Bethesda, Md.), 2022, 12-01, Volume: 12, Issue:12

    The functionally diverse members of the human Transforming Growth Factor-β (TGF-β) family are tightly regulated. TGF-β regulation includes 2 disulfide-dependent mechanisms-dimerization and partner protein binding. The specific cysteines participating in these regulatory mechanisms are known in just 3 of the 33 human TGF-β proteins. Human prodomain alignments revealed that 24 TGF-β prodomains contain conserved cysteines in 2 highly exposed locations. There are 3 in the region of the β8 helix that mediates dimerization near the prodomain carboxy terminus. There are 2 in the Association region that mediates partner protein binding near the prodomain amino terminus. The alignments predict the specific cysteines contributing to disulfide-dependent regulation of 72% of human TGF-β proteins. Database mining then identified 9 conserved prodomain cysteine mutations and their disease phenotypes in 7 TGF-β proteins. Three common adenoma phenotypes for prodomain cysteine mutations suggested 7 new regulatory heterodimer pairs. Two common adenoma phenotypes for prodomain and binding partner cysteine mutations revealed 17 new regulatory interactions. Overall, the analysis of human TGF-β prodomains suggests a significantly expanded scope of disulfide-dependent regulation by heterodimerization and partner protein binding; regulation that is often lost in tumors.

    Topics: Cysteine; Disulfides; Humans; Neoplasms; Protein Binding; Transforming Growth Factor beta

2022
Tumor-produced and aging-associated oncometabolite methylmalonic acid promotes cancer-associated fibroblast activation to drive metastatic progression.
    Nature communications, 2022, 10-20, Volume: 13, Issue:1

    The systemic metabolic shifts that occur during aging and the local metabolic alterations of a tumor, its stroma and their communication cooperate to establish a unique tumor microenvironment (TME) fostering cancer progression. Here, we show that methylmalonic acid (MMA), an aging-increased oncometabolite also produced by aggressive cancer cells, activates fibroblasts in the TME, which reciprocally secrete IL-6 loaded extracellular vesicles (EVs) that drive cancer progression, drug resistance and metastasis. The cancer-associated fibroblast (CAF)-released EV cargo is modified as a result of reactive oxygen species (ROS) generation and activation of the canonical and noncanonical TGFβ signaling pathways. EV-associated IL-6 functions as a stroma-tumor messenger, activating the JAK/STAT3 and TGFβ signaling pathways in tumor cells and promoting pro-aggressive behaviors. Our findings define the role of MMA in CAF activation to drive metastatic reprogramming, unveiling potential therapeutic avenues to target MMA at the nexus of aging, the tumor microenvironment and metastasis.

    Topics: Cancer-Associated Fibroblasts; Extracellular Vesicles; Humans; Interleukin-6; Methylmalonic Acid; Neoplasms; Reactive Oxygen Species; Transforming Growth Factor beta; Tumor Microenvironment

2022
Potential Anti-Tumor Activity of Nardoguaianone L Isolated from
    Molecules (Basel, Switzerland), 2022, Nov-03, Volume: 27, Issue:21

    Topics: Apoptosis; Humans; Nardostachys; Neoplasms; Transforming Growth Factor beta

2022
Tumor-Induced T Cell Polarization by Schwann Cells.
    Cells, 2022, 11-09, Volume: 11, Issue:22

    Nerve-cancer crosstalk resulting in either tumor neurogenesis or intratumoral neurodegeneration is critically controlled by Schwann cells, the principal glial cells of the peripheral nervous system. Though the direct stimulating effect of Schwann cells on malignant cell proliferation, motility, epithelial-mesenchymal transition, and the formation of metastases have been intensively investigated, the ability of Schwann cells to affect the effector and regulatory immune cells in the tumor environment is significantly less studied. Here, we demonstrated that tumor cells could stimulate Schwann cells to produce high levels of prostaglandin E, which could be blocked by COX-2 inhibitors. This effect was mediated by tumor-derived TGF-β as neutralization of this cytokine in the tumor-conditioned medium completely blocked the inducible prostaglandin E production by Schwann cells. Similar protective effects were also induced by the Schwann cell pretreatment with TGF-βR1/ALK4/5/7 and MAPK/ERK kinase inhibitors of the canonical and non-canonical TGF-β signaling pathways, respectively. Furthermore, prostaglandin E derived from tumor-activated Schwann cells blocked the proliferation of CD3/CD28-activated T cells and upregulated the expression of CD73 and PD-1 on both CD4+ and CD8+ T cells, suggesting T cell polarization to the exhausted phenotype. This new pathway of tumor-induced T cell inhibition via the activation of neuroglial cells represents new evidence of the importance of nerve-cancer crosstalk in controlling tumor development and progression. A better understanding of the tumor-neuro-immune axis supports the development of efficient targets for harnessing this axis and improving the efficacy of cancer therapy.

    Topics: Humans; Lymphocyte Activation; Neoplasms; Prostaglandins; Schwann Cells; Transforming Growth Factor beta

2022
Next-generation immunotherapy for solid tumors: combination immunotherapy with crosstalk blockade of TGFβ and PD-1/PD-L1.
    Expert opinion on investigational drugs, 2022, Volume: 31, Issue:11

    In solid tumor immunotherapy, less than 20% of patients respond to anti-programmed cell death 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) agents. The role of transforming growth factor β (TGFβ) in diverse immunity is well-established; however, systemic blockade of TGFβ is associated with toxicity. Accumulating evidence suggests the role of crosstalk between TGFβ and PD-1/PD-L1 pathways.. We focus on TGFβ and PD-1/PD-L1 signaling pathway crosstalk and the determinant role of TGFβ in the resistance of immune checkpoint blockade. We provide the rationale for combination anti-TGFβ and anti-PD-1/PD-L1 therapies for solid tumors and discuss the current status of dual blockade therapy in preclinical and clinical studies.. The heterogeneity of tumor microenvironment across solid tumors complicates patient selection, treatment regimens, and response and toxicity assessment for investigation of dual blockade agents. However, clinical knowledge from single-agent studies provides infrastructure to translate dual blockade therapies. Dual TGFβ and PD-1/PD-L1 blockade results in enhanced T-cell infiltration into tumors, a primary requisite for successful immunotherapy. A bifunctional fusion protein specifically targets TGFβ in the tumor microenvironment, avoiding systemic toxicity, and prevents interaction of PD-1+ cytotoxic cells with PD-L1+ tumor cells.

    Topics: B7-H1 Antigen; Humans; Immunotherapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2022
The role of YAP1 in survival prediction, immune modulation, and drug response: A pan-cancer perspective.
    Frontiers in immunology, 2022, Volume: 13

    Dysregulation of the Hippo signaling pathway has been implicated in multiple pathologies, including cancer, and. We compared. We found that. In conclusion,

    Topics: Cancer-Associated Fibroblasts; Hedgehog Proteins; Humans; Myeloid-Derived Suppressor Cells; Neoplasms; Transforming Growth Factor beta

2022
Preventing Surgery-Induced NK Cell Dysfunction Using Anti-TGF-β Immunotherapeutics.
    International journal of molecular sciences, 2022, Nov-23, Volume: 23, Issue:23

    Natural Killer (NK) cell cytotoxicity and interferon-gamma (IFNγ) production are profoundly suppressed postoperatively. This dysfunction is associated with increased morbidity and cancer recurrence. NK activity depends on the integration of activating and inhibitory signals, which may be modulated by transforming growth factor-beta (TGF-β). We hypothesized that impaired postoperative NK cell IFNγ production is due to altered signaling pathways caused by postoperative TGF-β. NK cell receptor expression, downstream phosphorylated targets, and IFNγ production were assessed using peripheral blood mononuclear cells (PBMCs) from patients undergoing cancer surgery. Healthy NK cells were incubated in the presence of healthy/baseline/postoperative day (POD) 1 plasma and in the presence/absence of a TGF-β-blocking monoclonal antibody (mAb) or the small molecule inhibitor (smi) SB525334. Single-cell RNA sequencing (scRNA-seq) was performed on PBMCs from six patients with colorectal cancer having surgery at baseline/on POD1. Intracellular IFNγ, activating receptors (CD132, CD212, NKG2D, DNAM-1), and downstream target (STAT5, STAT4, p38 MAPK, S6) phosphorylation were significantly reduced on POD1. Furthermore, this dysfunction was phenocopied in healthy NK cells through incubation with rTGF-β1 or POD1 plasma and was prevented by the addition of anti-TGF-β immunotherapeutics (anti-TGF-β mAb or TGF-βR smi). Targeted gene analysis revealed significant decreases in S6 and FKBP12, an increase in Shp-2, and a reduction in NK metabolism-associated transcripts on POD1. pSmad2/3 was increased and pS6 was reduced in response to rTGF-β1 on POD1, changes that were prevented by anti-TGF-β immunotherapeutics. Together, these results suggest that both canonical and mTOR pathways downstream of TGF-β mediate phenotypic changes that result in postoperative NK cell dysfunction.

    Topics: Antibodies, Monoclonal; Humans; Killer Cells, Natural; Leukocytes, Mononuclear; Neoplasms; Receptors, Natural Killer Cell; Transforming Growth Factor beta

2022
Two to tango! IL-13 and TGF-β drive myelofibrosis.
    Blood, 2022, 12-29, Volume: 140, Issue:26

    Topics: Humans; Interleukin-13; Interleukin-4; Myeloproliferative Disorders; Neoplasms; Primary Myelofibrosis; Transforming Growth Factor beta

2022
The antitumor activity of human Vγ9Vδ2 T cells is impaired by TGF-β through significant phenotype, transcriptomic and metabolic changes.
    Frontiers in immunology, 2022, Volume: 13

    Despite significant advances, the eradication of cancer remains a clinical challenge which justifies the urgent exploration of additional therapeutic strategies such as immunotherapies. Human peripheral Vγ9Vδ2 T cells represent an attractive candidate subset for designing safe, feasible and effective adoptive T cell transfer-based therapies. However, following their infiltration within tumors, γδ T cells are exposed to various regulating constituents and signals from the tumor microenvironment (TME), which severely alter their antitumor functions. Here, we show that TGF-β, whose elevated production in some solid tumors is linked to a poor prognosis, interferes with the antigenic activation of human Vγ9Vδ2 T cells

    Topics: Humans; Neoplasms; Phenotype; Receptors, Antigen, T-Cell, gamma-delta; T-Lymphocytes; Transcriptome; Transforming Growth Factor beta; Tumor Microenvironment

2022
Simultaneous targeting of TGF-β/PD-L1 synergizes with radiotherapy by reprogramming the tumor microenvironment to overcome immune evasion.
    Cancer cell, 2021, 10-11, Volume: 39, Issue:10

    Topics: Animals; Humans; Immune Checkpoint Inhibitors; Immune Evasion; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2021
Combine and conquer: manganese synergizing anti-TGF-β/PD-L1 bispecific antibody YM101 to overcome immunotherapy resistance in non-inflamed cancers.
    Journal of hematology & oncology, 2021, 09-15, Volume: 14, Issue:1

    Our previous work showed that the anti-TGF-β/PD-L1 bispecific antibody YM101 effectively overcame anti-PD-L1 resistance in immune-excluded tumor models. However, in immune-desert models, the efficacy of YM101 was limited. Bivalent manganese (Mn. The effect of Mn. Mn. Combining Mn

    Topics: Animals; Antibodies, Bispecific; Antineoplastic Agents, Immunological; Cell Line, Tumor; Drug Synergism; Humans; Immune Checkpoint Inhibitors; Immunotherapy; Manganese; Mice, Inbred BALB C; Neoplasms; Transforming Growth Factor beta

2021
CD137 Costimulation Counteracts TGFβ Inhibition of NK-cell Antitumor Function.
    Cancer immunology research, 2021, Volume: 9, Issue:12

    Enhancing natural killer (NK) cell-based cancer immunotherapy by overcoming immunosuppression is an area of intensive research. Here, we have demonstrated that the anti-CD137 agonist urelumab can overcome TGFβ-mediated inhibition of human NK-cell proliferation and antitumor function. Transcriptomic, immunophenotypic, and functional analyses showed that CD137 costimulation modified the transcriptional program induced by TGFβ on human NK cells by rescuing their proliferation in response to IL2, preserving their expression of activating receptors (NKG2D) and effector molecules (granzyme B, IFNγ) while allowing the acquisition of tumor-homing/retention features (CXCR3, CD103). Activated NK cells cultured in the presence of TGFβ1 and CD137 agonist recovered CCL5 and IFNγ secretion and showed enhanced direct and antibody-dependent cytotoxicity upon restimulation with cancer cells. Trastuzumab treatment of fresh breast carcinoma-derived multicellular cultures induced CD137 expression on tumor-infiltrating CD16

    Topics: Animals; Case-Control Studies; Cell Line, Tumor; Female; Gene Expression; Humans; Immunotherapy; Killer Cells, Natural; Microarray Analysis; Neoplasms; Transforming Growth Factor beta; Tumor Necrosis Factor Receptor Superfamily, Member 9

2021
Co-targeting TGF-β and PD-L1 with radiation therapy: The Goldilocks principle.
    Cell reports. Medicine, 2021, 09-21, Volume: 2, Issue:9

    In the current issue of

    Topics: B7-H1 Antigen; Humans; Neoplasms; Transforming Growth Factor beta

2021
Regulatory T cells promote cancer immune-escape through integrin αvβ8-mediated TGF-β activation.
    Nature communications, 2021, 10-28, Volume: 12, Issue:1

    Presence of TGFβ in the tumor microenvironment is one of the most relevant cancer immune-escape mechanisms. TGFβ is secreted in an inactive form, and its activation within the tumor may depend on different cell types and mechanisms than its production. Here we show in mouse melanoma and breast cancer models that regulatory T (Treg) cells expressing the β8 chain of αvβ8 integrin (Itgβ8) are the main cell type in the tumors that activates TGFβ, produced by the cancer cells and stored in the tumor micro-environment. Itgβ8 ablation in Treg cells impairs TGFβ signalling in intra-tumoral T lymphocytes but not in the tumor draining lymph nodes. Successively, the effector function of tumor infiltrating CD8

    Topics: Animals; Female; Humans; Integrins; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Microenvironment

2021
Genome-Editing-Mediated Restructuring of Tumor Immune Microenvironment for Prevention of Metastasis.
    ACS nano, 2021, Nov-23, Volume: 15, Issue:11

    Modulating the tumor immune microenvironment to activate immune cells has been investigated to convert cold to hot tumors. Here, we report that metal-lipid hybrid nanoparticle (MLN)-mediated gene editing of transforming growth factor-β (TGF-β) can restructure the tumor microenvironment to an "immune activated" state for subsequent immunotherapy. MLNs with cationic lipids and elemental metallic Au inside were designed to deliver plasmid DNA encoding TGF-β single guide RNA and Cas9 protein (pC9sTgf) and to convert near-infrared light (NIR) to heat. Upon NIR irradiation, MLNs induced photothermal anticancer effects and calreticulin exposure on B16F10 cancer cells. Lipoplexes of pC9sTgf and MLN (pC9sTgf@MLN) provided gene editing of B16F10 cells and

    Topics: Animals; Cell Line, Tumor; Gene Editing; Immunotherapy; Metal Nanoparticles; Mice; Neoplasms; Phototherapy; Transforming Growth Factor beta; Tumor Microenvironment

2021
Fibroblast pyruvate carboxylase is required for collagen production in the tumour microenvironment.
    Nature metabolism, 2021, Volume: 3, Issue:11

    The aberrant production of collagen by fibroblasts is a hallmark of many solid tumours and can influence cancer progression. How the mesenchymal cells in the tumour microenvironment maintain their production of extracellular matrix proteins as the vascular delivery of glutamine and glucose becomes compromised remains unclear. Here we show that pyruvate carboxylase (PC)-mediated anaplerosis in tumour-associated fibroblasts contributes to tumour fibrosis and growth. Using cultured mesenchymal and cancer cells, as well as mouse allograft models, we provide evidence that extracellular lactate can be utilized by fibroblasts to maintain tricarboxylic acid (TCA) cycle anaplerosis and non-essential amino acid biosynthesis through PC activity. Furthermore, we show that fibroblast PC is required for collagen production in the tumour microenvironment. These results establish TCA cycle anaplerosis as a determinant of extracellular matrix collagen production, and identify PC as a potential target to inhibit tumour desmoplasia.

    Topics: Animals; Cancer-Associated Fibroblasts; Cell Line; Citric Acid Cycle; Collagen; Disease Susceptibility; Enzyme Activation; Fibrosis; Gene Expression Regulation, Enzymologic; Glutamine; Humans; Lactic Acid; Mice; Neoplasms; Protein Biosynthesis; Pyruvate Carboxylase; Transforming Growth Factor beta; Tumor Microenvironment

2021
[Effects of Signaling Activation of Bone Morphogenetic Protein 2 on the Differentiation and Infiltration of Regulatory T Lymphocytes in Tumors].
    Zhongguo yi xue ke xue yuan xue bao. Acta Academiae Medicinae Sinicae, 2021, Dec-30, Volume: 43, Issue:6

    Objective To determine whether the signaling activation of bone morphogenetic protein 2(BMP2)can induce myeloid-derived suppressor cells(MDSC)to secret transforming growth factor β(TGF-β),further enhancing the differentiation and infiltration of regulatory T lymphocytes(Treg)into tumor tissue. Methods The BMP2-induced mRNA and protein expression of TGF-β in MDSC was detected by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay(ELISA),respectively.The effect of BMP2-induced TGF-β secretion by MDSC on Treg differentiation was then determined by flow cytometry.Finally,we implanted the recombined human bone morphogenetic protein 2(rhBMP2)collagen gels into tumor-burdened mice to examine the role of BMP2 in Treg differentiation via MDSC-secreted TGF-β

    Topics: Animals; Bone Morphogenetic Protein 2; Cell Differentiation; Mice; Myeloid-Derived Suppressor Cells; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2021
Identification of Potential Inhibitors Against the TGF-β/BMPs-Activin Receptor- like Kinase 1 Signal Pathway.
    Current computer-aided drug design, 2021, Volume: 17, Issue:4

    In many diseased states, especially fibrosis and cancer, TGF-β family members are overexpressed and the outcome of signaling is diverted toward disease progression. As the result of activin receptor-like kinase 1 (ALK1) plays a key role in TGF-β signaling, discovering inhibitors of ALK1 to block TGF-β signaling for a therapeutic benefit has become an effective strategy.. In this work, ZINC15894217 and ZINC12404282 were identified as potential ALK1 inhibitors using molecular docking, molecular dynamics simulation and MM/PBSA calculations studies. The analysis of energy decomposition found that Val208, Val216, Lys229, Gly283, Arg334 and Leu337 acted as crucial residues for ligand binding and system stabilizing.. In addition, these compounds displayed excellent pharmacological and structural properties, which can be further evaluated through in vitro and in vivo experiments for the inhibition of ALK1 to be developed as drugs against fibrosis and tumor.. Overall, our study illustrated a time- and cost-effective computer aided drug design procedure to identify potential ALK1 inhibitors. It would provide useful information for further development of ALK1 inhibitors to improve disease related to TGF-β signal pathway.

    Topics: Humans; Molecular Docking Simulation; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2021
Cytotoxic T cells isolated from healthy donors and cancer patients kill TGFβ-expressing cancer cells in a TGFβ-dependent manner.
    Cellular & molecular immunology, 2021, Volume: 18, Issue:2

    Transforming growth factor-beta (TGFβ) is a highly potent immunosuppressive cytokine. Although TGFβ is a tumor suppressor in early/premalignant cancer lesions, the cytokine has several tumor-promoting effects in advanced cancer; abrogation of the antitumor immune response is one of the most important tumor-promoting effects. As several immunoregulatory mechanisms have recently been shown to be targets of specific T cells, we hypothesized that TGFβ is targeted by naturally occurring specific T cells and thus could be a potential target for immunomodulatory cancer vaccination. Hence, we tested healthy donor and cancer patient T cells for spontaneous T-cell responses specifically targeting 38 20-mer epitopes derived from TGFβ1. We identified numerous CD4

    Topics: Case-Control Studies; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Epitopes, T-Lymphocyte; HLA-A2 Antigen; Humans; Neoplasms; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta

2021
Hierarchically Releasing Bio-Responsive Nanoparticles for Complete Tumor Microenvironment Modulation via TGF-β Pathway Inhibition and TAF Reduction.
    ACS applied materials & interfaces, 2021, Jan-20, Volume: 13, Issue:2

    Topics: 3T3 Cells; Animals; Antibiotics, Antineoplastic; Benzamides; Cancer-Associated Fibroblasts; Cell Line, Tumor; Delayed-Action Preparations; Dioxoles; Doxorubicin; Female; Hydrogen-Ion Concentration; Mice; Mice, Inbred BALB C; Nanoparticles; Neoplasms; Rotaxanes; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2021
Inhibition of TGFβ improves hematopoietic stem cell niche and ameliorates cancer-related anemia.
    Stem cell research & therapy, 2021, 01-18, Volume: 12, Issue:1

    Cancer cachexia is a wasting syndrome that is quite common in terminal-stage cancer patients. Cancer-related anemia is one of the main features of cancer cachexia and mostly results in a poor prognosis. The disadvantages of the current therapies are obvious, but few new treatments have been developed because the pathological mechanism remains unclear.. C57BL/6 mice were subcutaneously injected with Lewis lung carcinoma cells to generate a cancer-related anemia model. The treated group received daily intraperitoneal injections of SB505124. Blood parameters were determined with a routine blood counting analyzer. Erythroid cells and hematopoietic stem/progenitor cells were analyzed by flow cytometry. The microarchitecture changes of the femurs were determined by micro-computed tomography scans. Smad2/3 phosphorylation was analyzed by immunofluorescence and Western blotting. The changes in the hematopoietic stem cell niche were revealed by qPCR analysis of both fibrosis-related genes and hematopoietic genes, fibroblastic colony-forming unit assays, and lineage differentiation of mesenchymal stromal cells.. The mouse model exhibited hematopoietic suppression, marked by a decrease of erythrocytes in the peripheral blood, as well as an increase of immature erythroblasts and reduced differentiation of multipotent progenitors in the bone marrow. The ratio of bone volume/total volume, trabecular number, and cortical wall thickness all appeared to decrease, and the increased osteoclast number has led to the release of latent TGFβ and TGFβ signaling over-activation. Excessive TGFβ deteriorated the hematopoietic stem cell niche, inducing fibrosis of the bone marrow as well as the transition of mesenchymal stromal cells. Treatment with SB505124, a small-molecule inhibitor of TGFβ signaling, significantly attenuated the symptoms of cancer-related anemia in this model, as evidenced by the increase of erythrocytes in the peripheral blood and the normalized proportion of erythroblast cell clusters. Meanwhile, hindered hematopoiesis and deteriorated hematopoietic stem cell niche were also shown to be restored with SB505124 treatment.. This study investigated the role of TGFβ released by bone remodeling in the progression of cancer-related anemia and revealed a potential therapeutic approach for relieving defects in hematopoiesis.

    Topics: Anemia; Animals; Cell Differentiation; Hematopoiesis; Hematopoietic Stem Cells; Humans; Mice; Mice, Inbred C57BL; Neoplasms; Stem Cell Niche; Transforming Growth Factor beta; X-Ray Microtomography

2021
Cancer Environment Immunotherapy: targeting TGF-β finds its way towards tissue healing and vasculature remodeling.
    Signal transduction and targeted therapy, 2021, 01-29, Volume: 6, Issue:1

    Topics: Humans; Immunotherapy; Neoplasms; Transforming Growth Factor beta; Wound Healing

2021
Malignant subclone drives metastasis of genetically and phenotypically heterogenous cell clusters through fibrotic niche generation.
    Nature communications, 2021, 02-08, Volume: 12, Issue:1

    A concept of polyclonal metastasis has recently been proposed, wherein tumor cell clusters break off from the primary site and are disseminated. However, the involvement of driver mutations in such polyclonal mechanism is not fully understood. Here, we show that non-metastatic AP cells metastasize to the liver with metastatic AKTP cells after co-transplantation to the spleen. Furthermore, AKTP cell depletion after the development of metastases results in the continuous proliferation of the remaining AP cells, indicating a role of AKTP cells in the early step of polyclonal metastasis. Importantly, AKTP cells, but not AP cells, induce fibrotic niche generation when arrested in the sinusoid, and such fibrotic microenvironment promotes the colonization of AP cells. These results indicate that non-metastatic cells can metastasize via the polyclonal metastasis mechanism using the fibrotic niche induced by malignant cells. Thus, targeting the fibrotic niche is an effective strategy for halting polyclonal metastasis.

    Topics: Animals; Cell Aggregation; Cell Proliferation; Clone Cells; Fibrosis; Hepatic Stellate Cells; Liver; Mice, Inbred NOD; Neoplasm Metastasis; Neoplasms; Organoids; Phenotype; Spleen; Transforming Growth Factor beta

2021
Loss of TGFβ signaling increases alternative end-joining DNA repair that sensitizes to genotoxic therapies across cancer types.
    Science translational medicine, 2021, 02-10, Volume: 13, Issue:580

    Among the pleotropic roles of transforming growth factor-β (TGFβ) signaling in cancer, its impact on genomic stability is least understood. Inhibition of TGFβ signaling increases use of alternative end joining (alt-EJ), an error-prone DNA repair process that typically functions as a "backup" pathway if double-strand break repair by homologous recombination or nonhomologous end joining is compromised. However, the consequences of this functional relationship on therapeutic vulnerability in human cancer remain unknown. Here, we show that TGFβ broadly controls the DNA damage response and suppresses alt-EJ genes that are associated with genomic instability. Mechanistically based TGFβ and alt-EJ gene expression signatures were anticorrelated in glioblastoma, squamous cell lung cancer, and serous ovarian cancer. Consistent with error-prone repair, more of the genome was altered in tumors classified as low TGFβ and high alt-EJ, and the corresponding patients had better outcomes. Pan-cancer analysis of solid neoplasms revealed that alt-EJ genes were coordinately expressed and anticorrelated with TGFβ competency in 16 of 17 cancer types tested. Moreover, regardless of cancer type, tumors classified as low TGFβ and high alt-EJ were characterized by an insertion-deletion mutation signature containing short microhomologies and were more sensitive to genotoxic therapy. Collectively, experimental studies revealed that loss or inhibition of TGFβ signaling compromises the DNA damage response, resulting in ineffective repair by alt-EJ. Translation of this mechanistic relationship into gene expression signatures identified a robust anticorrelation that predicts response to genotoxic therapies, thereby expanding the potential therapeutic scope of TGFβ biology.

    Topics: DNA Breaks, Double-Stranded; DNA Damage; DNA End-Joining Repair; DNA Repair; Humans; Neoplasms; Transforming Growth Factor beta

2021
Geoffroea decorticans fruit extracts inhibit the wnt/β-catenin pathway, a therapeutic target in cancer.
    Biochemical and biophysical research communications, 2021, 03-26, Volume: 546

    Geoffroea decorticans (chañar) is commonly used for culinary and medicinal purposes in rural communities. The aim of this work was to chemically characterize three Geoffroea decorticans extracts and determine their capacity to modulate the wnt/β-catenin pathway. This signaling pathway plays a key role in embryonic development but its overactivation leads to cancer cell growth. Phytochemical analysis of extracts showed presence of major classes of phytochemicals. Gas chromatography-mass spectrometry results revealed the presence of acids, esters and furanic compounds. Using Xenopus embryos as in vivo model organisms, we found that the extracts modulated dorso-ventral axis formation and rescued hyperdorsalized phenotypes produced by LiCl treatment. In agreement with these findings, Geoffroea decorticans extracts decreased β-catenin levels and suppressed the expression of wnt target genes such as xnr3 and chordin, thus demonstrating an inhibitory regulation of the wnt/β-catenin signaling pathway. All these results support a new role for Geoffroea decorticans fruit derivatives with possible anti-carcinogenic actions.

    Topics: Animals; beta Catenin; Fabaceae; Female; Fruit; Gene Expression Regulation; Glycoproteins; Intercellular Signaling Peptides and Proteins; Lithium Chloride; Male; Molecular Targeted Therapy; Neoplasms; Plant Extracts; Transforming Growth Factor beta; Wnt Signaling Pathway; Xenopus laevis; Xenopus Proteins

2021
MicroRNA-362 negatively and positively regulates SMAD4 expression in TGF-β/SMAD signaling to suppress cell migration and invasion.
    International journal of medical sciences, 2021, Volume: 18, Issue:8

    Topics: Animals; Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Humans; Male; Mice; MicroRNAs; Neoplasm Invasiveness; Neoplasms; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2021
Comment on "Tumor-initiating cells establish an IL-33-TGF-β niche signaling loop to promote cancer progression".
    Science (New York, N.Y.), 2021, 04-09, Volume: 372, Issue:6538

    Taniguchi

    Topics: Humans; Interleukin-33; Neoplasms; Neoplastic Stem Cells; Receptors, IgE; Transforming Growth Factor beta

2021
Response to Comment on "Tumor-initiating cells establish an IL-33-TGF-β niche signaling loop to promote cancer progression".
    Science (New York, N.Y.), 2021, 04-09, Volume: 372, Issue:6538

    Kamphuis

    Topics: Humans; Interleukin-33; Neoplasms; Neoplastic Stem Cells; Receptors, IgE; Transforming Growth Factor beta

2021
BFAR coordinates TGFβ signaling to modulate Th9-mediated cancer immunotherapy.
    The Journal of experimental medicine, 2021, 07-05, Volume: 218, Issue:7

    TGFβ is essential for the generation of anti-tumor Th9 cells; on the other hand, it causes resistance against anti-tumor immunity. Despite recent progress, the underlying mechanism reconciling the double-edged effect of TGFβ signaling in Th9-mediated cancer immunotherapy remains elusive. Here, we find that TGFβ-induced down-regulation of bifunctional apoptosis regulator (BFAR) represents the key mechanism preventing the sustained activation of TGFβ signaling and thus impairing Th9 inducibility. Mechanistically, BFAR mediates K63-linked ubiquitination of TGFβR1 at K268, which is critical to activate TGFβ signaling. Thus, BFAR deficiency or K268R knock-in mutation suppresses TGFβR1 ubiquitination and Th9 differentiation, thereby inhibiting Th9-mediated cancer immunotherapy. More interestingly, BFAR-overexpressed Th9 cells exhibit promising therapeutic efficacy to curtail tumor growth and metastasis and promote the sensitivity of anti-PD-1-mediated checkpoint immunotherapy. Thus, our findings establish BFAR as a key TGFβ-regulated gene to fine-tune TGFβ signaling that causes Th9 induction insensitivity, and they highlight the translational potential of BFAR in promoting Th9-mediated cancer immunotherapy.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Apoptosis Regulatory Proteins; Cell Differentiation; Down-Regulation; Humans; Immunotherapy; Membrane Proteins; Mice; Mice, Inbred C57BL; Mice, Knockout; Neoplasms; Signal Transduction; T-Lymphocytes, Helper-Inducer; Transforming Growth Factor beta

2021
Single-cell transcriptomics reveals the effect of PD-L1/TGF-β blockade on the tumor microenvironment.
    BMC biology, 2021, 05-25, Volume: 19, Issue:1

    The anti-tumor activity of anti-PD-1/PD-L1 therapies correlates with T cell infiltration in tumors. Thus, a major goal in oncology is to find strategies that enhance T cell infiltration and efficacy of anti-PD-1/PD-L1 therapy. TGF-β has been shown to contribute to T cell exclusion, and anti-TGF-β improves anti-PD-L1 efficacy in vivo. However, TGF-β inhibition has frequently been shown to induce toxicity in the clinic, and the clinical efficacy of combination PD-L1 and TGF-β blockade has not yet been proven. To identify strategies to overcome resistance to PD-L1 blockade, the transcriptional programs associated with PD-L1 and/or TGF-β blockade in the tumor microenvironment should be further elucidated.. We used single-cell RNA sequencing in a mouse model to characterize the transcriptomic effects of PD-L1 and/or TGF-β blockade on nearly 30,000 single cells in the tumor and surrounding microenvironment. Combination treatment led to upregulation of immune response genes, including multiple chemokine genes such as CCL5, in macrophages, and downregulation of extracellular matrix genes in fibroblasts. Analysis of publicly available tumor transcriptome profiles showed that the chemokine CCL5 was strongly associated with immune cell infiltration in various human cancers. Further investigation with in vivo models showed that intratumorally administered CCL5 enhanced cytotoxic lymphocytes and the anti-tumor activity of anti-PD-L1.. Taken together, our data could be leveraged translationally to complement or find alternatives to anti-PD-L1 plus anti-TGF-β combination therapy, for example through companion biomarkers, and/or to identify novel targets that could be modulated to overcome resistance.

    Topics: Animals; B7-H1 Antigen; Mice; Neoplasms; Transcriptome; Transforming Growth Factor beta; Tumor Microenvironment

2021
The TGF-β-Th2 axis: A new target for cancer therapy?
    Allergy, 2021, Volume: 76, Issue:11

    Topics: Humans; Interleukin-4; Neoplasms; Th2 Cells; Transforming Growth Factor beta

2021
TGF-β inhibition can overcome cancer primary resistance to PD-1 blockade: A mathematical model.
    PloS one, 2021, Volume: 16, Issue:6

    Immune checkpoint inhibitors have demonstrated, over the recent years, impressive clinical response in cancer patients, but some patients do not respond at all to checkpoint blockade, exhibiting primary resistance. Primary resistance to PD-1 blockade is reported to occur under conditions of immunosuppressive tumor environment, a condition caused by myeloid derived suppressor cells (MDSCs), and by T cells exclusion, due to increased level of T regulatory cells (Tregs). Since TGF-β activates Tregs, TGF-β inhibitor may overcome primary resistance to anti-PD-1. Indeed, recent mice experiments show that combining anti-PD-1 with anti-TGF-β yields significant therapeutic improvements compared to anti-TGF-β alone. The present paper introduces two cancer-specific parameters and, correspondingly, develops a mathematical model which explains how primary resistance to PD-1 blockade occurs, in terms of the two cancer-specific parameters, and how, in combination with anti-TGF-β, anti-PD-1 provides significant benefits. The model is represented by a system of partial differential equations and the simulations are in agreement with the recent mice experiments. In some cancer patients, treatment with anti-PD-1 results in rapid progression of the disease, known as hyperprogression disease (HPD). The mathematical model can also explain how this situation arises, and it predicts that HPD may be reversed by combining anti-TGF-β to anti-PD-1. The model is used to demonstrate how the two cancer-specific parameters may serve as biomarkers in predicting the efficacy of combination therapy with PD-1 and TGF-β inhibitors.

    Topics: Animals; Antineoplastic Agents, Immunological; CD8-Positive T-Lymphocytes; Drug Resistance, Neoplasm; Immune Checkpoint Inhibitors; Mice; Models, Theoretical; Neoplasms; Programmed Cell Death 1 Receptor; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Microenvironment

2021
Transforming growth factor-β blockade modulates tumor mechanical microenvironments for enhanced antitumor efficacy of photodynamic therapy.
    Nanoscale, 2021, Jun-14, Volume: 13, Issue:22

    Photodynamic therapy (PDT) is frequently used in cancer treatment in clinical settings. However, its applications in stroma-rich solid tumors, e.g., triple negative breast cancer, are limited by abnormal mechanical microenvironments. Solid stress accumulated in stroma-rich solid tumors compresses tumor blood vessels, hampers the delivery of photosensitizers (PSs) in tumor tissues, and poses a major challenge for potent PDT. Here, we report a novel combination strategy to augment PDT based cancer therapy by combining hydroxyethyl starch-chlorin e6 conjugate self-assembled nanoparticles (HES-Ce6 NPs) with the transforming growth factor-β (TGFβ) inhibitor LY2157299 (LY). HES-Ce6 conjugates, as synthesized by one step esterification reaction, could self-assemble into uniform HES-Ce6 NPs, which exhibited enhanced photostability and generated more reactive oxygen species (ROS) under 660 nm laser irradiation than free Ce6. Prior to PDT, intragastric administration of LY decreased collagen deposition, alleviated solid stress, and decompressed tumor blood vessels. As a result, the reconstructed tumor mechanical microenvironment promoted accumulation and penetration of HES-Ce6 NPs into tumor tissues, contributing to augmented antitumor efficacy of HES-Ce6 NP mediated PDT. Modulating tumor mechanical microenvironments using TGFβ blockade to enhance the delivery of PSs in tumors with excessive extracellular matrix represents an efficient strategy for treating stroma-rich solid tumors.

    Topics: Cell Line, Tumor; Humans; Nanoparticles; Neoplasms; Photochemotherapy; Photosensitizing Agents; Porphyrins; Pyrazoles; Quinolines; Transforming Growth Factor beta; Tumor Microenvironment

2021
Combined tumor-directed recruitment and protection from immune suppression enable CAR T cell efficacy in solid tumors.
    Science advances, 2021, Volume: 7, Issue:24

    CAR T cell therapy remains ineffective in solid tumors, due largely to poor infiltration and T cell suppression at the tumor site. T regulatory (T

    Topics: Humans; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2021
Concomitant attenuation of HMGCR expression and activity enhances the growth inhibitory effect of atorvastatin on TGF-β-treated epithelial cancer cells.
    Scientific reports, 2021, 06-17, Volume: 11, Issue:1

    Epithelial-mesenchymal transition (EMT) in primary tumor cells is a key prerequisite for metastasis initiation. Statins, cholesterol-lowering drugs, can delay metastasis formation in vivo and attenuate the growth and proliferation of tumor cells in vitro. The latter effect is stronger in tumor cells with a mesenchymal-like phenotype than in those with an epithelial one. However, the effect of statins on epithelial cancer cells treated with EMT-inducing growth factors such as transforming growth factor-β (TGF-β) remains unclear. Here, we examined the effect of atorvastatin on two epithelial cancer cell lines following TGF-β treatment. Atorvastatin-induced growth inhibition was stronger in TGF-β-treated cells than in cells not thusly treated. Moreover, treatment of cells with atorvastatin prior to TGF-β treatment enhanced this effect, which was further potentiated by the simultaneous reduction in the expression of the statin target enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). Dual pharmacological targeting of HMGCR can thus strongly inhibit the growth and proliferation of epithelial cancer cells treated with TGF-β and may also improve statin therapy-mediated attenuation of metastasis formation in vivo.

    Topics: Atorvastatin; Biomarkers, Tumor; Cell Count; Cell Line, Tumor; Cell Proliferation; Cell Size; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Hydroxymethylglutaryl CoA Reductases; Models, Biological; Neoplasms; RNA, Messenger; Transforming Growth Factor beta

2021
Bakuchiol, main component of root bark of Ulmus davidiana var. japonica, inhibits TGF-β-induced in vitro EMT and in vivo metastasis.
    Archives of biochemistry and biophysics, 2021, 09-30, Volume: 709

    Cancer is a second leading cause of death worldwide, and metastasis is the major cause of cancer-related mortality. The epithelial-mesenchymal transition (EMT), known as phenotypic change from epithelial cells to mesenchymal cells, is a crucial biological process during development. However, inappropriate activation of EMT contributes to tumor progression and promoting metastasis; therefore, inhibiting EMT is considered a promising strategy for developing drugs that can treat or prevent cancer. In the present study, we investigated the anti-cancer effect of bakuchiol (BC), a main component of Ulmus davidiana var. japonica, in human cancer cells using A549, HT29 and MCF7 cells. In MTT and colony forming assay, BC exerted cytotoxicity activity against cancer cells and inhibited proliferation of these cells. Anti-metastatic effects by BC were further confirmed by observing decreased migration and invasion in TGF-β-induced cancer cells after BC treatment. Furthermore, BC treatment resulted in increase of E-cadherin expression and decrease of Snail level in Western blotting and immunofluorescence analysis, supporting its anti-metastatic activity. In addition, BC inhibited lung metastasis of tail vein injected human cancer cells in animal model. These findings suggest that BC inhibits migration and invasion of cancers by suppressing EMT and in vivo metastasis, thereby may be a potential therapeutic agent for treating cancers.

    Topics: Animals; Antineoplastic Agents; Cadherins; Cell Line, Tumor; Cell Movement; Cell Proliferation; Epithelial-Mesenchymal Transition; Humans; Mice, SCID; Neoplasm Metastasis; Neoplasms; Phenols; Plant Bark; Plant Extracts; Plant Roots; Snail Family Transcription Factors; Transforming Growth Factor beta; Ulmus; Xenograft Model Antitumor Assays

2021
Integrin αvβ8 on T cells suppresses anti-tumor immunity in multiple models and is a promising target for tumor immunotherapy.
    Cell reports, 2021, 07-06, Volume: 36, Issue:1

    αvβ8 integrin, a key activator of transforming growth factor β (TGF-β), inhibits anti-tumor immunity. We show that a potent blocking monoclonal antibody against αvβ8 (ADWA-11) causes growth suppression or complete regression in syngeneic models of squamous cell carcinoma, mammary cancer, colon cancer, and prostate cancer, especially when combined with other immunomodulators or radiotherapy. αvβ8 is expressed at the highest levels in CD4+CD25+ T cells in tumors, and specific deletion of β8 from T cells is as effective as ADWA-11 in suppressing tumor growth. ADWA-11 increases expression of a suite of genes in tumor-infiltrating CD8+ T cells normally inhibited by TGF-β and involved in tumor cell killing, including granzyme B and interferon-γ. The in vitro cytotoxic effect of tumor CD8 T cells is inhibited by CD4+CD25+ cells, and this suppressive effect is blocked by ADWA-11. These findings solidify αvβ8 integrin as a promising target for cancer immunotherapy.

    Topics: Animals; Antibodies, Neoplasm; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cell Line, Tumor; Cell Proliferation; CTLA-4 Antigen; Gene Expression Regulation, Neoplastic; Granzymes; Immunity; Immunotherapy; Integrins; Interferon-gamma; Lymphocyte Depletion; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; Models, Biological; Mutation; Neoplasms; Signal Transduction; Smad3 Protein; Survival Analysis; T-Lymphocytes; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor Receptor Superfamily, Member 9

2021
Inflammation and tumor progression: signaling pathways and targeted intervention.
    Signal transduction and targeted therapy, 2021, 07-12, Volume: 6, Issue:1

    Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.

    Topics: Antigen Presentation; Dendritic Cells; Humans; Immunity, Cellular; Inflammasomes; Inflammation; Interferons; Interleukins; Janus Kinases; Mitogen-Activated Protein Kinase Kinases; Molecular Targeted Therapy; Neoplasms; NF-kappa B; Signal Transduction; STAT Transcription Factors; Toll-Like Receptors; Transforming Growth Factor beta

2021
Hormonally Regulated Myogenic miR-486 Influences Sex-specific Differences in Cancer-induced Skeletal Muscle Defects.
    Endocrinology, 2021, 10-01, Volume: 162, Issue:10

    Cancer-induced skeletal muscle defects show sex-specific differences in severity with men performing poorly compared to women. Hormones and sex chromosomal differences are suggested to mediate these differences, but the functional skeletal muscle markers to document these differences are unknown. We show that the myogenic microRNA miR-486 is a marker of sex-specific differences in cancer-induced skeletal muscle defects. Cancer-induced loss of circulating miR-486 was more severe in men with bladder, lung, and pancreatic cancers compared to women with the same cancer types. In a syngeneic model of pancreatic cancer, circulating and skeletal muscle loss of miR-486 was more severe in male mice compared to female mice. Estradiol (E2) and the clinically used selective estrogen receptor modulator toremifene increased miR-486 in undifferentiated and differentiated myoblast cell line C2C12 and E2-inducible expression correlated with direct binding of estrogen receptor alpha (ERα) to the regulatory region of the miR-486 gene. E2 and toremifene reduced the actions of cytokines such as myostatin, transforming growth factor β, and tumor necrosis factor α, which mediate cancer-induced skeletal muscle wasting. E2- and toremifene-treated C2C12 myoblast/myotube cells contained elevated levels of active protein kinase B (AKT) with a corresponding decrease in the levels of its negative regulator PTEN, which is a target of miR-486. We propose an ERα:E2-miR-486-AKT signaling axis, which reduces the deleterious effects of cancer-induced cytokines/chemokines on skeletal muscle mass and/or function.

    Topics: Animals; Cell Differentiation; Cell Line, Tumor; Estradiol; Female; Gene Expression Regulation, Neoplastic; Humans; Male; Mice; Mice, Inbred C57BL; MicroRNAs; Muscle, Skeletal; Muscular Diseases; Myostatin; Neoplasms; Sex Factors; Signal Transduction; Toremifene; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2021
AMBRA1 Promotes TGFβ Signaling via Nonproteolytic Polyubiquitylation of Smad4.
    Cancer research, 2021, 10-01, Volume: 81, Issue:19

    Transforming growth factor β (TGFβ) is prometastatic in advanced cancers and its biological activities are mainly mediated by the Smad family of proteins. Smad4 is the central signal transducer and transcription factor in the TGFβ pathway, yet the underlying mechanisms that govern transcriptional activities of Smad4 are not fully understood. Here, we show that AMBRA1, a member of the DDB1 and CUL4-associated factor (DCAF) family of proteins, serves as the substrate receptor for Smad4 in the CUL4-RING (CRL4) ubiquitin ligase complex. The CRL4-AMBRA1 ubiquitin ligase mediates nonproteolytic polyubiquitylation of Smad4 to enhance its transcriptional functions. Consequently, AMBRA1 potentiated TGFβ signaling and critically promoted TGFβ-induced epithelial-to-mesenchymal transition, migration, and invasion of breast cancer cells. Mouse models of breast cancer demonstrated that AMBRA1 promotes metastasis. Collectively, these results show that CRL4-AMBRA1 facilitates TGFβ-driven metastasis by increasing Smad4 polyubiquitylation, suggesting AMBRA1 may serve as a new therapeutic target in metastatic breast cancer. SIGNIFICANCE: This study identifies AMBRA1 as a novel regulator of TGFβ signaling and breast cancer metastasis, supporting further exploration of AMBRA1 as a target for cancer therapy.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Carrier Proteins; Cell Line, Tumor; Cells, Cultured; Disease Models, Animal; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Heterografts; Humans; Mice; Mutation; Neoplasms; Protein Binding; Signal Transduction; Smad4 Protein; Substrate Specificity; Transforming Growth Factor beta; Ubiquitination

2021
A TGF-β-MTA1-SOX4-EZH2 signaling axis drives epithelial-mesenchymal transition in tumor metastasis.
    Oncogene, 2020, Volume: 39, Issue:10

    MTA1, SOX4, EZH2, and TGF-β are all potent inducers of epithelial-mesenchymal transition (EMT) in cancer; however, the signaling relationship among these molecules in EMT is poorly understood. Here, we investigated the function of MTA1 in cancer cells and demonstrated that MTA1 overexpression efficiently activates EMT. This activation resulted in a significant increase in the migratory and invasive properties of three different cancer cell lines through a common mechanism involving SOX4 activation, screened from a gene expression profiling analysis. We showed that both SOX4 and MTA1 are induced by TGF-β and both are indispensable for TGF-β-mediated EMT. Further investigation identified that MTA1 acts upstream of SOX4 in the TGF-β pathway, emphasizing a TGF-β-MTA1-SOX4 signaling axis in EMT induction. The histone methyltransferase EZH2, a component of the polycomb (PcG) repressive complex 2 (PRC2), was identified as a critical responsive gene of the TGF-β-MTA1-SOX4 signaling in three different epithelial cancer cell lines, suggesting that this signaling acts broadly in cancer cells in vitro. The MTA1-SOX4-EZH2 signaling cascade was further verified in TCGA pan-cancer patient samples and in a colon cancer cDNA microarray, and activation of genes in this signaling pathway predicted an unfavorable prognosis in colon cancer patients. Collectively, our data uncover a SOX4-dependent EMT-inducing mechanism underlying MTA1-driven cancer metastasis and suggest a widespread TGF-β-MTA1-SOX4-EZH2 signaling axis that drives EMT in various cancers. We propose that this signaling may be used as a common therapeutic target to control epithelial cancer metastasis.

    Topics: Cell Line, Tumor; Colonic Neoplasms; Enhancer of Zeste Homolog 2 Protein; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Neoplasm Metastasis; Neoplasms; Prognosis; Repressor Proteins; Signal Transduction; SOXC Transcription Factors; Trans-Activators; Transforming Growth Factor beta

2020
Gain-of-function pathogenic variants in SMAD4 are associated with neoplasia in Myhre syndrome.
    American journal of medical genetics. Part A, 2020, Volume: 182, Issue:2

    Myhre syndrome is an increasingly diagnosed rare syndrome that is caused by one of two specific heterozygous gain-of-function pathogenic variants in SMAD4. The phenotype includes short stature, characteristic facial appearance, hearing loss, laryngotracheal stenosis, arthritis, skeletal abnormalities, learning and social challenges, distinctive cardiovascular defects, and a striking fibroproliferative response in the ear canals, airways, and serosal cavities (peritoneum, pleura, pericardium). Confirmation of the clinical diagnosis is usually prompted by the characteristic appearance with developmental delay and autistic-like behavior using targeted gene sequencing or by whole exome sequencing. We describe six patients (two not previously reported) with molecularly confirmed Myhre syndrome and neoplasia. Loss-of-function pathogenic variants in SMAD4 cause juvenile polyposis syndrome and we hypothesize that the gain-of-function pathogenic variants observed in Myhre syndrome may contribute to neoplasia in the patients reported herein. The frequency of neoplasia (9.8%, 6/61) in this series (two new, four reported patients) and endometrial cancer (8.8%, 3/34, mean age 40 years) in patients with Myhre syndrome, raises the possibility of cancer susceptibility in these patients. We alert clinicians to the possibility of detecting this syndrome when cancer screening panels are used. We propose that patients with Myhre syndrome are more susceptible to neoplasia, encourage increased awareness, and suggest enhanced clinical monitoring.

    Topics: Adult; Cryptorchidism; Endometrial Neoplasms; Exome Sequencing; Facies; Female; Gain of Function Mutation; Growth Disorders; Hand Deformities, Congenital; Heterozygote; Humans; Intellectual Disability; Male; Middle Aged; Mutation; Neoplasms; Phenotype; Smad4 Protein; Transforming Growth Factor beta

2020
TGF-β orchestrates fibrogenic and developmental EMTs via the RAS effector RREB1.
    Nature, 2020, Volume: 577, Issue:7791

    Epithelial-to-mesenchymal transitions (EMTs) are phenotypic plasticity processes that confer migratory and invasive properties to epithelial cells during development, wound-healing, fibrosis and cancer

    Topics: Animals; Cell Line, Tumor; DNA-Binding Proteins; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Fibrosis; Gastrulation; Humans; Mice; Mitogen-Activated Protein Kinases; Neoplasms; Organoids; ras Proteins; Smad Proteins; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2020
Histone deacetylase inhibitors dysregulate DNA repair proteins and antagonize metastasis-associated processes.
    Journal of cancer research and clinical oncology, 2020, Volume: 146, Issue:2

    We set out to determine whether clinically tested epigenetic drugs against class I histone deacetylases (HDACs) affect hallmarks of the metastatic process.. We treated permanent and primary renal, lung, and breast cancer cells with the class I histone deacetylase inhibitors (HDACi) entinostat (MS-275) and valproic acid (VPA), the replicative stress inducer hydroxyurea (HU), the DNA-damaging agent cis-platinum (L-OHP), and the cytokine transforming growth factor-β (TGFβ). We used proteomics, quantitative PCR, immunoblot, single cell DNA damage assays, and flow cytometry to analyze cell fate after drug exposure.. We show that HDACi interfere with DNA repair protein expression and trigger DNA damage and apoptosis alone and in combination with established chemotherapeutics. Furthermore, HDACi disrupt the balance of cell adhesion protein expression and abrogate TGFβ-induced cellular plasticity of transformed cells.. HDACi suppress the epithelial-mesenchymal transition (EMT) and compromise the DNA integrity of cancer cells. These data encourage further testing of HDACi against tumor cells.

    Topics: Animals; Benzamides; Cell Plasticity; Cisplatin; DNA Repair; DNA Repair Enzymes; DNA-Binding Proteins; Drug Resistance, Neoplasm; Histone Deacetylase Inhibitors; Humans; Hydroxyurea; Male; Mice; Mice, Inbred BALB C; Neoplasm Metastasis; Neoplasms; Pyridines; Transforming Growth Factor beta; Valproic Acid

2020
Selection of the Recommended Phase 2 Dose for Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1.
    Clinical pharmacology and therapeutics, 2020, Volume: 108, Issue:3

    Bintrafusp alfa, a first-in-class bifunctional fusion protein composed of the extracellular domain of the TGF-βRII receptor (TGF-β "trap") fused to a human IgG1-blocking PD-L1, showed a manageable safety profile and clinical activity in phase I studies in patients with heavily pretreated advanced solid tumors. The recommended phase 2 dose (RP2D) was selected based on integration of modeling, simulations, and all available data. A 1,200-mg every 2 weeks (q2w) dose was predicted to maintain serum trough concentration (C

    Topics: Administration, Intravenous; Animals; Antineoplastic Agents, Immunological; B7-H1 Antigen; Clinical Trials, Phase I as Topic; Clinical Trials, Phase II as Topic; Computer Simulation; Disease Models, Animal; Drug Administration Schedule; Drug Dosage Calculations; Humans; Immune Checkpoint Inhibitors; Mice; Models, Theoretical; Molecular Targeted Therapy; Neoplasms; Research Design; Transforming Growth Factor beta

2020
TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors.
    JCI insight, 2020, 02-27, Volume: 5, Issue:4

    In recent years, chimeric antigen receptor-modified T cell (CAR T cell) therapy has proven to be a promising approach against cancer. Nonetheless, this approach still faces multiple challenges in eliminating solid tumors, one of which being the immunosuppressive tumor microenvironment (TME). Here, we demonstrated that knocking out the endogenous TGF-β receptor II (TGFBR2) in CAR T cells with CRISPR/Cas9 technology could reduce the induced Treg conversion and prevent the exhaustion of CAR T ce lls. Meanwhile, TGFBR2-edited CAR T cells had better in vivo tumor elimination efficacy, both in cell line-derived xenograft and patient-derived xenograft solid tumor models, whether administered locally or systemically. In addition, the TGFBR2-edited CAR T cells could eliminate contralaterally reinoculated xenografts in mice effectively, with an increased proportion of memory subsets within circulating CAR T cells of central memory and effector memory subsets. In conclusion, we greatly improved the in vitro and in vivo function of CAR T cells in TGF-β-rich tumor environments by knocking out endogenous TGFBR2 and propose a potentially new method to improve the efficacy of CAR T cell therapy for treating solid tumors.

    Topics: Animals; Clustered Regularly Interspaced Short Palindromic Repeats; Forkhead Transcription Factors; Humans; Mice; Mice, Knockout; Neoplasms; Receptor, Transforming Growth Factor-beta Type II; Receptors, Chimeric Antigen; T-Lymphocytes; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2020
Monoubiquitination of p120-catenin is essential for TGFβ-induced epithelial-mesenchymal transition and tumor metastasis.
    Science advances, 2020, Volume: 6, Issue:4

    Disassembly of intercellular junctions is a hallmark of epithelial-mesenchymal transition (EMT). However, how the junctions disassemble remains largely unknown. Here, we report that E3 ubiquitin ligase Smurf1 targets p120-catenin, a core component of adherens junction (AJ) complex, for monoubiquitination during transforming growth factor β (TGFβ)-induced EMT, thereby leading to AJ dissociation. Upon TGFβ treatment, activated extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylates T900 of p120-catenin to promote its interaction with Smurf1 and subsequent monoubiquitination. Inhibition of T900 phosphorylation or ubiquitination of p120-catenin abrogates TGFβ-induced AJ dissociation and consequent tight junction (TJ) dissociation and cytoskeleton rearrangement, hence markedly blocking lung metastasis of murine breast cancer. Moreover, the T900 phosphorylation level of p120-catenin is positively correlated with malignancy of human breast cancer. Hence, our study reveals the underlying mechanism by which TGFβ induces dissociation of AJs during EMT and provides a potential strategy to block tumor metastasis.

    Topics: Adherens Junctions; Animals; Breast Neoplasms; Catenins; Cell Line, Tumor; Cells, Cultured; Delta Catenin; Epithelial-Mesenchymal Transition; Female; Humans; Mice; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Neoplasms; Phosphorylation; Transforming Growth Factor beta; Ubiquitin-Protein Ligases; Ubiquitination

2020
[TGF-β-targeted immunotherapy aiming for new and efficient treatments of cancer].
    Medecine sciences : M/S, 2020, Volume: 36, Issue:1

    Topics: Animals; Antineoplastic Agents, Immunological; Humans; Immunotherapy; Molecular Targeted Therapy; Neoplasms; Therapies, Investigational; Transforming Growth Factor beta; Treatment Outcome

2020
Annexin A5 as an immune checkpoint inhibitor and tumor-homing molecule for cancer treatment.
    Nature communications, 2020, 02-28, Volume: 11, Issue:1

    The interaction between immune cells and phosphatidylserine (PS) molecules exposed on the surface of apoptotic-tumor bodies, such as those induced by chemotherapies, contributes to the formation of an immunosuppressive tumor microenvironment (TME). Annexin A5 (AnxA5) binds with high affinity to PS externalized by apoptotic cells, thereby hindering their interaction with immune cells. Here, we show that AnxA5 administration rescue the immunosuppressive state of the TME induced by chemotherapy. Due to the preferential homing of AnxA5 to the TME enriched with PS+ tumor cells, we demonstrate in vivo that fusing tumor-antigen peptide to AnxA5 significantly enhances its immunogenicity and antitumor efficacy when administered after chemotherapy. Also, the therapeutic antitumor effect of an AnxA5-peptide fusion can be further enhanced by administration of other immune checkpoint inhibitors. Our findings support the administration of AnxA5 following chemotherapy as a promising immune checkpoint inhibitor for cancer treatment.

    Topics: Animals; Annexin A5; Antibodies, Blocking; Antigens, Neoplasm; Cancer Vaccines; Cell Line, Tumor; Cisplatin; Disease Models, Animal; Female; Humans; Immunologic Factors; Mice, Inbred BALB C; Mice, Inbred C57BL; Neoplasms; Papillomavirus E7 Proteins; Phosphatidylserines; Recombinant Fusion Proteins; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor-alpha

2020
Transforming growth factor beta (TGF-β) activity in immuno-oncology studies.
    Methods in enzymology, 2020, Volume: 636

    Transforming growth factor beta (TGF-β) is a potent pleiotropic polypeptide cytokine, with a complex and context dependent control of its activation, signaling and effector functions. This cytokine is pivotal in the regulation of immunological responses, tumor initiation and development, stromal homeostasis and all their intricate related interactions. Last decade advances in cancer immunotherapy have reactivated the clinical interest on potential drug with TGF-β inhibition effect, combined with immunomodulating enhancer drugs. The correct quantification of the in vitro and in vivo biological activity of this cytokine is essential to understand the intrinsic underlying biological mechanisms and TGF-β role in the immune system, tumor and stromal codevelopment, modulation and interactions. There is a wide variety of available procedures to quantify TGF-β activity, which includes different methodological approximations like ELISA, Bioassays including reporter gene assays, Flow cytometry (FC), Western blotting (WB), immunochemical/fluorescence microscopy, among others. Here, we detail available methods for TGF-β biological activity analysis, together with their applicability and suitability for each experimental setting, in order to get a complete analytical perspective and more comprehensive information along the development and design of combined antitumor immunotherapies, which include the inhibition of TGF-β biological activity.

    Topics: Cytokines; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2020
Selective inhibition of TGFβ1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape.
    Science translational medicine, 2020, 03-25, Volume: 12, Issue:536

    Despite breakthroughs achieved with cancer checkpoint blockade therapy (CBT), many patients do not respond to anti-programmed cell death-1 (PD-1) due to primary or acquired resistance. Human tumor profiling and preclinical studies in tumor models have recently uncovered transforming growth factor-β (TGFβ) signaling activity as a potential point of intervention to overcome primary resistance to CBT. However, the development of therapies targeting TGFβ signaling has been hindered by dose-limiting cardiotoxicities, possibly due to nonselective inhibition of multiple TGFβ isoforms. Analysis of mRNA expression data from The Cancer Genome Atlas revealed that

    Topics: Animals; Cardiotoxicity; CD8-Positive T-Lymphocytes; Cell Line, Tumor; Humans; Mice; Neoplasms; Rats; Signal Transduction; Transforming Growth Factor beta

2020
Multi-Omics Analysis Reveals the Pan-Cancer Landscape of Bone Morphogenetic Proteins.
    Medical science monitor : international medical journal of experimental and clinical research, 2020, Apr-05, Volume: 26

    BACKGROUND Bone morphogenetic proteins (BMPs) are widely involved in cancer development. However, a wealth of conflicting data raises the question of whether BMPs serve as oncogenes or as cancer suppressors. MATERIAL AND METHODS By integrating multi-omics data across cancers, we comprehensively analyzed the genomic and pharmacogenomic landscape of BMP genes across cancers. RESULTS Surprisingly, our data indicate that BMPs are globally downregulated in cancers. Further genetics and epigenetics analyses show that this abnormal expression is driven by copy number variations, especially heterozygous amplification. We next assessed the BMP-associated pathways and demonstrated that they suppress cell cycle and estrogen hormone pathways. Bone morphogenetic protein interacts with 58 compounds, and their dysfunction can induce drug sensitivity. CONCLUSIONS Our results define the landscape of the BMP family at a systems level and open potential therapeutic opportunities for cancer patients.

    Topics: Animals; Bone Morphogenetic Proteins; DNA Copy Number Variations; Humans; Neoplasms; Transforming Growth Factor beta

2020
TGFβ suppresses CD8
    Nature communications, 2020, 04-09, Volume: 11, Issue:1

    Transforming growth factor beta (TGFβ) is a multipotent immunosuppressive cytokine. TGFβ excludes immune cells from tumors, and TGFβ inhibition improves the efficacy of cytotoxic and immune therapies. Using preclinical colorectal cancer models in cell type-conditional TGFβ receptor I (ALK5) knockout mice, we interrogate this mechanism. Tumor growth delay and radiation response are unchanged in animals with Treg or macrophage-specific ALK5 deletion. However, CD8αCre-ALK5

    Topics: Animals; CD8-Positive T-Lymphocytes; Cell Line, Tumor; Cell Movement; Cell Survival; Chemokine CXCL10; Gene Expression Regulation; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Transgenic; Neoplasms; Promoter Regions, Genetic; Protein Binding; Receptors, CXCR3; Smad2 Protein; Transforming Growth Factor beta

2020
Targeting TGFβ Signalling in Cancer: Toward Context-Specific Strategies.
    Trends in cancer, 2020, Volume: 6, Issue:7

    The transforming growth factor beta (TGFβ) signalling pathway regulates a range of important cellular processes in a context-dependent manner. Recent discoveries have provided important insights into the regulation of the TGFβ pathway and its change from an antitumorigenic to a protumorigenic pathway. These findings may have important implications for cancer stem cell (CSC) functions and therapeutic strategies.

    Topics: Humans; Neoplasms; Neoplastic Stem Cells; Signal Transduction; Transforming Growth Factor beta

2020
Dynamic extracellular matrix stiffening induces a phenotypic transformation and a migratory shift in epithelial cells.
    Integrative biology : quantitative biosciences from nano to macro, 2020, 06-19, Volume: 12, Issue:6

    Soft tissue tumors, including breast cancer, become stiffer throughout disease progression. This increase in stiffness has been shown to correlate to malignant phenotype and epithelial-to-mesenchymal transition (EMT) in vitro. Unlike current models, utilizing static increases in matrix stiffness, our group has previously created a system that allows for dynamic stiffening of an alginate-matrigel composite hydrogel to mirror the native dynamic process. Here, we utilize this system to evaluate the role of matrix stiffness on EMT and metastasis both in vitro and in vivo. Epithelial cells were seen to lose normal morphology and become protrusive and migratory after stiffening. This shift corresponded to a loss of epithelial markers and gain of mesenchymal markers in both the cell clusters and migrated cells. Furthermore, stiffening in a murine model reduced tumor burden and increased migratory behavior prior to tumor formation. Inhibition of FAK and PI3K in vitro abrogated the morphologic and migratory transformation of epithelial cell clusters. This work demonstrates the key role extracellular matrix stiffening has in tumor progression through integrin signaling and, in particular, its ability to drive EMT-related changes and metastasis.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Movement; Disease Progression; Epithelial Cells; Epithelial-Mesenchymal Transition; Extracellular Matrix; Female; Humans; Hydrogels; In Vitro Techniques; Integrins; Mammary Neoplasms, Animal; Mice; Microscopy, Confocal; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Phenotype; Signal Transduction; Transforming Growth Factor beta

2020
Architecture of Cancer-Associated Fibroblasts in Tumor Microenvironment: Mapping Their Origins, Heterogeneity, and Role in Cancer Therapy Resistance.
    Omics : a journal of integrative biology, 2020, Volume: 24, Issue:6

    The tumor stroma, a key component of the tumor microenvironment (TME), is a key determinant of response and resistance to cancer treatment. The stromal cells, extracellular matrix (ECM), and blood vessels influence cancer cell response to therapy and play key roles in tumor relapse and therapeutic outcomes. Of the stromal cells present in the TME, much attention has been given to cancer-associated fibroblasts (CAFs) as they are the most abundant and important in cancer initiation, progression, and therapy resistance. Besides releasing several factors, CAFs also synthesize the ECM, a key component of the tumor stroma. In this expert review, we examine the role of CAFs in the regulation of tumor cell behavior and reveal how CAF-derived factors and signaling influence tumor cell heterogeneity and development of novel strategies to combat cancer. Importantly, CAFs display both phenotypic and functional heterogeneity, with significant ramifications on CAF-directed therapies. Principal anti-cancer therapies targeting CAFs take the form of: (1) CAFs' ablation through use of immunotherapies, (2) re-education of CAFs to normalize the cells, (3) cellular therapies involving CAFs delivering drugs such as oncolytic adenoviruses, and (4) stromal depletion via targeting the ECM and its related signaling. The CAFs' heterogeneity could be a result of different cellular origins and the cancer-specific tumor microenvironmental effects, underscoring the need for further multiomics and biochemical studies on CAFs and the subsets. Lastly, we present recent advances in therapeutic targeting of CAFs and the success of such endeavors or their lack thereof. We recommend that to advance global public health and personalized medicine, treatments in the oncology clinic should be combinatorial in nature, strategically targeting both cancer cells and stromal cells, and their interactions.

    Topics: Animals; Biomarkers; Cancer-Associated Fibroblasts; Cell Communication; Cell Transformation, Neoplastic; Disease Management; Disease Susceptibility; Drug Resistance, Neoplasm; Extracellular Matrix; Humans; Neoplasms; Neovascularization, Pathologic; Radiation Tolerance; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2020
Boosting Natural Killer Cell-Based Cancer Immunotherapy with Selenocystine/Transforming Growth Factor-Beta Inhibitor-Encapsulated Nanoemulsion.
    ACS nano, 2020, 09-22, Volume: 14, Issue:9

    Natural killer (NK) cell-based immunotherapy represents a promising strategy to overcome the bottlenecks of cancer treatment. However, the therapeutic efficacy is greatly limited by downregulation of recognition ligands on the tumor cell surface, and the immunosuppressive effects can be thwarted by the tumor microenvironment such as secretion of transforming growth factor-beta (TGF-β), which could stunt the NK cell-mediated immune response. To overcome these limitations, herein we developed a nanoemulsion system (SSB NMs) to co-deliver TGF-β inhibitor and selenocysteine (SeC) to achieve amplified anticancer efficacy. SSB NMs significantly enhanced the lytic potency of NK92 cells by 2.1-fold. Moreover, a subtoxic dose of SSB NMs effectively sensitized MDA-MB-231 triple-negative breast cancer (TNBC) cells to NK cells derived from seven clinical patients, resulting in an up to 13.8-fold increase in cancer lysis. Mechanistic studies reveal that the sensitizing effects relied on natural killer group 2, member D (NKG2D)/NKG2D ligands (NKG2DLs) signaling with the involvement of DNA damage response. SSB NMs also effectively restrained TGF-β/TGF-β RI/Smad2/3 signaling, which thus enhanced NKG2DL expression on tumor cells and stimulated NKG2D surface expression on NK92 cells, ultimately contributing to the enhanced immune response. Furthermore, SSB NMs sustained release of SeC and TGF-β inhibitor and synergized with NK92 cells to induce significant anticancer effects

    Topics: Cell Line, Tumor; Cystine; Humans; Immunotherapy; Killer Cells, Natural; Neoplasms; Organoselenium Compounds; Transforming Growth Factor beta; Transforming Growth Factors

2020
Age-induced accumulation of methylmalonic acid promotes tumour progression.
    Nature, 2020, Volume: 585, Issue:7824

    The risk of cancer and associated mortality increases substantially in humans from the age of 65 years onwards

    Topics: Adult; Aged; Aging; Animals; Cell Line, Tumor; Disease Progression; Female; Gene Expression Regulation, Neoplastic; Humans; Male; Methylmalonic Acid; Mice; Middle Aged; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Signal Transduction; SOXC Transcription Factors; Transcriptome; Transforming Growth Factor beta

2020
Large-scale pan-cancer analysis reveals broad prognostic association between TGF-β ligands, not Hedgehog, and GLI1/2 expression in tumors.
    Scientific reports, 2020, 09-02, Volume: 10, Issue:1

    GLI1 expression is broadly accepted as a marker of Hedgehog pathway activation in tumors. Efficacy of Hedgehog inhibitors is essentially limited to tumors bearing activating mutations of the pathway. GLI2, a critical Hedgehog effector, is necessary for GLI1 expression and is a direct transcriptional target of TGF-β/SMAD signaling. We examined the expression correlations of GLI1/2 with TGFB and HH genes in 152 distinct transcriptome datasets totaling over 23,500 patients and representing 37 types of neoplasms. Their prognostic value was measured in over 15,000 clinically annotated tumor samples from 26 tumor types. In most tumor types, GLI1 and GLI2 follow a similar pattern of expression and are equally correlated with HH and TGFB genes. However, GLI1/2 broadly share prognostic value with TGFB genes and a mesenchymal/EMT signature, not with HH genes. Our results provide a likely explanation for the frequent failure of anti-Hedgehog therapies in tumors, as they suggest a key role for TGF-β, not Hedgehog, ligands, in tumors with elevated GLI1/2-expression.

    Topics: Computational Biology; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Hedgehog Proteins; Humans; Ligands; Multivariate Analysis; Neoplasms; Nuclear Proteins; Oligonucleotide Array Sequence Analysis; Prognosis; Proportional Hazards Models; Risk Factors; Signal Transduction; Transcriptome; Transforming Growth Factor beta; Zinc Finger Protein GLI1; Zinc Finger Protein Gli2

2020
TGF-β suppresses type 2 immunity to cancer.
    Nature, 2020, Volume: 587, Issue:7832

    The immune system uses two distinct defence strategies against infections: microbe-directed pathogen destruction characterized by type 1 immunity

    Topics: Animals; CD8-Positive T-Lymphocytes; Cell Death; Cell Hypoxia; Cell Line; Disease Progression; Female; Gene Expression Regulation; Humans; Interferon-gamma; Interleukin-4; Male; Mice; Mice, Inbred C57BL; Neoplasms; Receptor, Transforming Growth Factor-beta Type II; Signal Transduction; Stromal Cells; Th2 Cells; Transforming Growth Factor beta

2020
Dual genome-wide CRISPR knockout and CRISPR activation screens identify mechanisms that regulate the resistance to multiple ATR inhibitors.
    PLoS genetics, 2020, Volume: 16, Issue:11

    The ataxia telangiectasia and Rad3-related (ATR) protein kinase is a key regulator of the cellular response to DNA damage. Due to increased amount of replication stress, cancer cells heavily rely on ATR to complete DNA replication and cell cycle progression. Thus, ATR inhibition is an emerging target in cancer therapy, with multiple ATR inhibitors currently undergoing clinical trials. Here, we describe dual genome-wide CRISPR knockout and CRISPR activation screens employed to comprehensively identify genes that regulate the cellular resistance to ATR inhibitors. Specifically, we investigated two different ATR inhibitors, namely VE822 and AZD6738, in both HeLa and MCF10A cells. We identified and validated multiple genes that alter the resistance to ATR inhibitors. Importantly, we show that the mechanisms of resistance employed by these genes are varied, and include restoring DNA replication fork progression, and prevention of ATR inhibitor-induced apoptosis. In particular, we describe a role for MED12-mediated inhibition of the TGFβ signaling pathway in regulating replication fork stability and cellular survival upon ATR inhibition. Our dual genome-wide screen findings pave the way for personalized medicine by identifying potential biomarkers for ATR inhibitor resistance.

    Topics: Apoptosis; Ataxia Telangiectasia Mutated Proteins; Biomarkers, Tumor; CRISPR-Cas Systems; DNA Replication; Drug Resistance, Neoplasm; Drug Screening Assays, Antitumor; Gene Knockdown Techniques; HeLa Cells; Humans; Indoles; Mediator Complex; Morpholines; Neoplasms; Protein Kinase Inhibitors; Pyrimidines; Signal Transduction; Sulfonamides; Sulfoxides; Transforming Growth Factor beta

2020
Identification of novel TGF-β regulated genes with pro-migratory roles.
    Biochimica et biophysica acta. Molecular basis of disease, 2019, 12-01, Volume: 1865, Issue:12

    Transforming growth factor-β (TGF-β) signaling plays fundamental roles in the development and homeostasis of somatic cells. Dysregulated TGF-β signaling contributes to cancer progression and relapse to therapies by inducing epithelial-to-mesenchymal transition (EMT), enriching cancer stem cells, and promoting immunosuppression. Although many TGF-β-regulated genes have been identified, only a few datasets were obtained by next-generation sequencing. In this study, we performed RNA-sequencing analysis of MCF10A cells and identified 1166 genes that were upregulated and 861 genes that were downregulated by TGF-β. Gene set enrichment analysis revealed that focal adhesion and metabolic pathways were the top enriched pathways of the up- and downregulated genes, respectively. Genes in these pathways also possess significant predictive value for renal cancers. Moreover, we confirmed that TGF-β induced expression of MICAL1 and 2, and the histone demethylase, KDM7A, and revealed their regulatory roles on TGF-β-induced cell migration. We also show a critical effect of KDM7A in regulating the acetylation of H3K27 on TGF-β-induced genes. In sum, this study identified novel effectors that mediate the pro-migratory role of TGF-β signaling, paving the way for future studies that investigate the function of MICAL family members in cancer and the novel epigenetic mechanisms downstream TGF-β signaling.

    Topics: A549 Cells; Cell Line, Tumor; Cell Movement; Epigenesis, Genetic; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Jumonji Domain-Containing Histone Demethylases; Microfilament Proteins; Mixed Function Oxygenases; Neoplasms; Signal Transduction; Transcriptome; Transforming Growth Factor beta

2019
Tumor-derived TGF-β inhibits mitochondrial respiration to suppress IFN-γ production by human CD4
    Science signaling, 2019, 09-17, Volume: 12, Issue:599

    Transforming growth factor-β (TGF-β) is produced by tumors, and increased amounts of this cytokine in the tumor microenvironment and serum are associated with poor patient survival. TGF-β-mediated suppression of antitumor T cell responses contributes to tumor growth and survival. However, TGF-β also has tumor-suppressive activity; thus, dissecting cell type-specific molecular effects may inform therapeutic strategies targeting this cytokine. Here, using human peripheral and tumor-associated lymphocytes, we investigated how tumor-derived TGF-β suppresses a key antitumor function of CD4

    Topics: Adenosine Triphosphate; Animals; CD4-Positive T-Lymphocytes; Humans; Interferon-gamma; Mitochondria; Mitochondrial Proton-Translocating ATPases; Neoplasms; Oxygen Consumption; Phosphorylation; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2019
Modeling the stem cell hypothesis: Investigating the effects of cancer stem cells and TGF-β on tumor growth.
    Mathematical biosciences and engineering : MBE, 2019, 08-07, Volume: 16, Issue:6

    We propose a mathematical model to describe the interaction of cancer stem cells, tumor cells, and the immune system in order to better understand tumor growth in the presence of cancer stem cells. We consider the system in two scenarios: with no-treatment and with a chemotherapy treatment regimen. We develop a system of differential equations, fit the parameters to experimental data, and perform sensitivity and stability analysis. The model simulations show that the tumor cells grow as predicted with no-treatment and that with chemotherapy, which targets only the tumor cells, the cancer will eventually relapse. As chemotherapy does not target the cancer stem cells, we conclude that the tumor cells recover due to the presence of cancer stem cells.

    Topics: Animals; Antineoplastic Agents; Calibration; CD8-Positive T-Lymphocytes; Drug Administration Schedule; Fluorouracil; Gene Expression Regulation, Neoplastic; Humans; Mice; Models, Biological; Neoplasm Recurrence, Local; Neoplasms; Neoplastic Stem Cells; Reproducibility of Results; Transforming Growth Factor beta; Treatment Outcome

2019
Synthesis and biological evaluation of quercetin and resveratrol peptidyl derivatives as potential anticancer and antioxidant agents.
    Amino acids, 2019, Volume: 51, Issue:2

    Quercetin and resveratrol are polyphenolic compounds, members of the flavonoid and the stilbene family, respectively, both medicinally important as dietary anticancer and antioxidant agents. They are present in a variety of foods-including fruits, vegetables, tea, wine, as well as other dietary supplements-and are responsible for various health benefits. Different quercetin and resveratrol esters of Leu/Met-enkephalin and tetrapeptide Leu-Ser-Lys-Leu (LSKL) were synthesized as model systems for monitoring the influence of the peptides on biological activity of resveratrol and quercetin. General formula of the main peptidyl-quercetin derivatives is 2-[3-(aa)

    Topics: Antineoplastic Agents; Antioxidants; Cell Proliferation; Chromatography, High Pressure Liquid; Dietary Supplements; Enkephalin, Leucine; Enkephalin, Methionine; Esters; HCT116 Cells; Humans; MCF-7 Cells; Neoplasms; Peptides; Phytochemicals; Quercetin; Resveratrol; Solubility; Transforming Growth Factor beta

2019
Towards the contribution of the p38MAPK pathway to the dual role of TGFβ in cancer: A boolean model approach.
    Computers in biology and medicine, 2019, Volume: 104

    The transforming growth factor-beta (TGF-β) pathway is involved in the regulation of cell growth and differentiation. In normal cells or in the early stages of cancer, this pathway can control proliferation stimuli by inducing cell cycle arrest or apoptosis (through the MAP-kinase protein p38MAPK), while in late stages it seems to act as a tumor promoter. This feature is known as the TGF-β dual role in cancer and it is not completely explained. This seems to arise through the accumulation of mutations in cancer development that affect the normal function of these pathways. In this work we propose a Boolean model of the crosstalk between the TGF-β, p38 MAPK and cell cycle checkpoint pathways which qualitatively describes this dual behavior. The model shows that for the wild type case, TGF-β acts as tumor supressor by inducing cell cycle arrest or apoptosis, as expected. However, the loss of function (LoF) of its two signaling proteins: SMAD2 and SMAD3 has immortalization effects due to the activation of the PI3K/AKT pathway that contributes to inhibit apoptosis. In silico mutations of the model elements were compared with cell phenotypes in experiments presenting agreement. In addition, we performed a series of double gene perturbations (that simulate random deleterious mutations) to determine the main regulators of the network. The results suggest that SMAD2/3 and p38MAPK are key players in processing the network input. In addition, when the LoF of SMAD2/3 is combined with the LoF of p38MAPK and p53, cell cycle arrest is completely abrogated. In conclusion, the model allows to visualize, through in silico mutations, the dual role of TGF-β: for the wild-type case TGF-β is able to block proliferation, however deleterious mutations can impair cell cycle arrest promoting cellular proliferation.

    Topics: Apoptosis; Cell Cycle; Humans; MAP Kinase Signaling System; Models, Biological; Neoplasm Proteins; Neoplasms; p38 Mitogen-Activated Protein Kinases; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta

2019
In vitro analysis of immunomodulatory effects of mesenchymal stem cell- and tumor cell -derived exosomes on recall antigen-specific responses.
    International immunopharmacology, 2019, Volume: 67

    The aim of the present study was to evaluate in vitro effects of exosomes derived from mesenchymal stem cells (MSCs) or tumor cells on recall-antigen-specific immune responses.. The exosomes were isolated from the supernatant of the cultures of the adipose-derived MSCs, and 4T1 cell line. The splenocytes isolated from experimental autoimmune encephalomyelitis (EAE) mice were utilized to evaluate the effects of exosomes on recall-antigen-specific responses. The expression of master regulators for T cell sub-types and the levels of their corresponding cytokines were evaluated.. Treatment by disease-inducing peptide (MOG. It was ultimately attained that TEX and MSC-EXO utilized various mechanisms to modulate the recall immune responses. TEX was more potent than MSC-EXO to induce regulatory responses by upregulating the production of Foxp3, IL-10, and TGF-β.

    Topics: Animals; Carcinoma; Cell Line, Tumor; Epithelial Cells; Exosomes; Female; Interleukin-10; Mammary Neoplasms, Animal; Mesenchymal Stem Cells; Mice; Mice, Inbred C57BL; Neoplasms; Spleen; Transforming Growth Factor beta

2019
ALK phosphorylates SMAD4 on tyrosine to disable TGF-β tumour suppressor functions.
    Nature cell biology, 2019, Volume: 21, Issue:2

    Loss of TGF-β tumour suppressive response is a hallmark of human cancers. As a central player in TGF-β signal transduction, SMAD4 (also known as DPC4) is frequently mutated or deleted in gastrointestinal and pancreatic cancer. However, such genetic alterations are rare in most cancer types and the underlying mechanism for TGF-β resistance is not understood. Here we describe a mechanism of TGF-β resistance in ALK-positive tumours, including lymphoma, lung cancer and neuroblastoma. We demonstrate that, in ALK-positive tumours, ALK directly phosphorylates SMAD4 at Tyr 95. Phosphorylated SMAD4 is unable to bind to DNA and fails to elicit TGF-β gene responses and tumour suppressing responses. Chemical or genetic interference of the oncogenic ALK restores TGF-β responses in ALK-positive tumour cells. These findings reveal that SMAD4 is tyrosine-phosphorylated by an oncogenic tyrosine kinase during tumorigenesis. This suggests a mechanism by which SMAD4 is inactivated in cancers and provides guidance for targeted therapies in ALK-positive cancers.

    Topics: Anaplastic Lymphoma Kinase; Animals; Cell Line; Cell Line, Tumor; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; HEK293 Cells; Humans; Mice, Inbred BALB C; Mice, Nude; Neoplasms; Phosphorylation; Smad4 Protein; Transforming Growth Factor beta; Transplantation, Heterologous; Tyrosine

2019
Effect of pembrolizumab on CD4
    Clinical and experimental immunology, 2019, Volume: 196, Issue:3

    Tumor immune evasion involves the expansion of avidly proliferating immunosuppressive cells and inhibition of effector T cell proliferation. Immune checkpoints (IC) block the activation pathways of tumor-reactive T cells. IC pathways are often exploited by tumor cells to evade immune destruction, and blocking these pathways through IC inhibitors (ICI) has shown promising results in multiple malignancies. In this study, we investigated the effects of an ICI, pembrolizumab, on various T cell subsets in vitro. We compared the suppressive activity of CD4

    Topics: Antibodies, Monoclonal, Humanized; Antineoplastic Agents; CD4 Antigens; Cells, Cultured; Hepatitis A Virus Cellular Receptor 2; Humans; Immunophenotyping; Immunotherapy; Interleukin-2 Receptor alpha Subunit; Neoplasms; Peptides; Programmed Cell Death 1 Receptor; Protein Precursors; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Escape

2019
PDZK1-interacting protein 1 (PDZK1IP1) traps Smad4 protein and suppresses transforming growth factor-β (TGF-β) signaling.
    The Journal of biological chemistry, 2019, 03-29, Volume: 294, Issue:13

    Transforming growth factor (TGF)-β signaling in humans is stringently regulated to prevent excessive TGF-β signaling. In tumors, TGF-β signaling can both negatively and positively regulate tumorigenesis dependent on tumor type, but the reason for these opposite effects is unclear. TGF-β signaling is mainly mediated via the Smad-dependent pathway, and herein we found that PDZK1-interacting protein 1 (PDZK1IP1) interacts with Smad4. PDZK1IP1 inhibited both the TGF-β and the bone morphogenetic protein (BMP) pathways without affecting receptor-regulated Smad (R-Smad) phosphorylation. Rather than targeting R-Smad phosphorylation, PDZK1IP1 could interfere with TGF-β- and BMP-induced R-Smad/Smad4 complex formation. Of note, PDZK1IP1 retained Smad4 in the cytoplasm of TGF-β-stimulated cells. To pinpoint PDZK1IP1's functional domain, we created several PDZK1IP1 variants and found that its middle region, from Phe

    Topics: Animals; Cell Line, Tumor; Cell Movement; Cell Proliferation; Humans; Male; Membrane Proteins; Mice, Inbred BALB C; Neoplasms; Phosphorylation; Protein Interaction Maps; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta

2019
Enhanced cancer cell invasion caused by fibroblasts when fluid flow is present.
    Biomechanics and modeling in mechanobiology, 2019, Volume: 18, Issue:4

    It has been demonstrated that interstitial fluid (IF) flow can play a crucial role in tumor cell progression. Swartz and collaborators (Cancer Cell 11: 526-538, Shields et al. 2007) demonstrated that cells that secrete the lymphoid homing chemokines CCL21/CCL19 and express their receptor CCR7 could use flow to bias the secreted chemokine, causing pericellular gradients that stimulate cells to migrate in the direction of the flow. In a further work by Shieh et al. (Cancer Res 71: 790-800, 2011), a synergetic enhancement of tumor cell invasion caused by interaction between tumor cells and fibroblasts in the presence of fluid flow was reported. In the present work, we extend a previous proposed cell-fluid mathematical model for autologous chemotaxis (Chem Eng Sci 191: 268-287, Waldeland and Evje 2018) to also include fibroblasts. This results in a cell-fibroblast-fluid model. Motivated by the experimental findings by Shieh et al, the momentum balance equation for the fibroblasts involves (1) a stress term that accounts for chemotaxis in the direction of positive gradients in secreted growth factor TGF-[Formula: see text]; (2) a fibroblast-ECM interaction term; (3) a cancer cell-fibroblast interaction term. Imposing reasonable simplifying assumptions, we derive an explicit expression for the cancer cell velocity [Formula: see text] that reveals a balance between a fluid-generated stress term, a chemotactic-driven migration term (autologous chemotaxis), and a new term that accounts for the possible mechanical interaction between fibroblasts and cancer cells. Similarly, the model provides an expression for the fibroblast velocity [Formula: see text] as well as the IF velocity [Formula: see text]. The three-phase model is then used for comparison of the simulated output with experimental results to elucidate some of the possible mechanism(s) behind the reported fibroblast-enhanced tumor cell invasion.

    Topics: Cell Movement; Extracellular Fluid; Extracellular Matrix; Fibroblasts; Humans; Models, Biological; Neoplasm Invasiveness; Neoplasms; Rheology; Transforming Growth Factor beta

2019
ALK-mediated Tyr95 phosphorylation of Smad4 impairs its transcription activity and the tumor suppressive activity of TGF-β.
    Science China. Life sciences, 2019, Volume: 62, Issue:3

    Topics: Anaplastic Lymphoma Kinase; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Phosphorylation; Protein Binding; Smad4 Protein; Transcriptional Activation; Transforming Growth Factor beta; Tumor Suppressor Proteins; Tyrosine

2019
Combined Inhibition of TGF-β Signaling and the PD-L1 Immune Checkpoint Is Differentially Effective in Tumor Models.
    Cells, 2019, 04-05, Volume: 8, Issue:4

    Antibodies blocking the programmed death-ligand 1 (PD-L1) have shown impressive and durable responses in clinical studies. However, this type of immunotherapy is only effective in a subset of patients and not sufficient for rejection of all tumor types. In this study, we explored in two mouse tumor models whether the antitumor effect could be enhanced by the combined blockade of PD-L1 and transforming growth factor-β (TGF-β), a potent immunosuppressive cytokine. The effect of anti-PD-L1 mouse monoclonal (mAb) and a TGF-β type I receptor small molecule kinase inhibitor (LY364947) was evaluated in the highly immunogenic mouse MC38 colon adenocarcinoma and the poorly immunogenic mouse KPC1 pancreatic tumor model. In the MC38 tumor model, LY364947 monotherapy did not show any antitumor effect, whereas treatment with anti-PD-L1 mAb significantly delayed tumor outgrowth. However, combination therapy showed the strongest therapeutic efficacy, resulting in improved long-term survival compared with anti-PD-L1 mAb monotherapy. This improved survival was associated with an increased influx of CD8⁺ T cells in the tumor microenvironment. In the KPC1 tumor model, LY364947 did not enhance the antitumor effect of anti-PD-L1 mAb. Despite this, delayed KPC1 tumor outgrowth was observed in the LY364947-treated group and this treatment led to a significant reduction of CD4⁺ T cells in the tumor microenvironment. Together, our data indicate that an additive anti-tumor response of dual targeting PD-L1 and TGF-β is dependent on the tumor model used, highlighting the importance of selecting appropriate cancer types, using in-depth analysis of the tumor microenvironment, which can benefit from combinatorial immunotherapy regimens.

    Topics: Animals; Antibodies, Monoclonal; Cell Line, Tumor; Cell Proliferation; Disease Models, Animal; Female; Immunotherapy; Mice; Mice, Inbred C57BL; Neoplasms; Programmed Cell Death 1 Receptor; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2019
Competitive endogenous RNA is an intrinsic component of EMT regulatory circuits and modulates EMT.
    Nature communications, 2019, 04-09, Volume: 10, Issue:1

    The competitive endogenous RNA (ceRNA) hypothesis suggests an intrinsic mechanism to regulate biological processes. However, whether the dynamic changes of ceRNAs can modulate miRNA activities remains controversial. Here, we examine the dynamics of ceRNAs during TGF-β-induced epithelial-to-mesenchymal transition (EMT). We observe that TGFBI, a transcript highly induced during EMT in A549 cells, acts as the ceRNA for miR-21 to modulate EMT. We further identify FN1 as the ceRNA for miR-200c in the canonical SNAIL-ZEB-miR200 circuit in MCF10A cells. Experimental assays and computational simulations demonstrate that the dynamically induced ceRNAs are directly coupled with the canonical double negative feedback loops and are critical to the induction of EMT. These results help to establish the relevance of ceRNA in cancer EMT and suggest that ceRNA is an intrinsic component of the EMT regulatory circuit and may represent a potential target to disrupt EMT during tumorigenesis.

    Topics: A549 Cells; Carcinogenesis; Computational Biology; Epithelial-Mesenchymal Transition; Extracellular Matrix Proteins; Fibronectins; Gene Expression Regulation, Neoplastic; Gene Regulatory Networks; Humans; MicroRNAs; Models, Biological; Neoplasms; RNA, Messenger; Transforming Growth Factor beta

2019
ALK7 Signaling Manifests a Homeostatic Tissue Barrier That Is Abrogated during Tumorigenesis and Metastasis.
    Developmental cell, 2019, 05-06, Volume: 49, Issue:3

    Herein, we report that the TGFß superfamily receptor ALK7 is a suppressor of tumorigenesis and metastasis, as revealed by functional studies in mouse models of pancreatic neuroendocrine and luminal breast cancer, complemented by experimental metastasis assays. Activation in neoplastic cells of the ALK7 signaling pathway by its principal ligand activin B induces apoptosis. During tumorigenesis, cancer cells use two different approaches to evade this barrier, either downregulating activin B and/or downregulating ALK7. Suppressing ALK7 expression additionally contributes to the capability for metastatic seeding. ALK7 is associated with shorter relapse-free survival of various human cancers and distant-metastasis-free survival of breast cancer patients. This study introduces mechanistic insights into primary and metastatic tumor development, in the form of a protective barrier that triggers apoptosis in cells that are not "authorized" to proliferate within a particular tissue, by virtue of those cells expressing ALK7 in a tissue microenvironment bathed in its ligand.

    Topics: Activin Receptors, Type I; Activins; Animals; Apoptosis; Breast Neoplasms; Carcinogenesis; Cell Line, Tumor; Cell Transformation, Neoplastic; Female; Heterografts; Homeostasis; Humans; Male; Mice; Mice, Inbred A; Mice, Inbred C57BL; Mice, SCID; Neoplasm Metastasis; Neoplasms; Pancreatic Neoplasms; Signal Transduction; Smad2 Protein; Transforming Growth Factor beta; Tumor Microenvironment

2019
An Oncolytic Adenovirus Targeting Transforming Growth Factor β Inhibits Protumorigenic Signals and Produces Immune Activation: A Novel Approach to Enhance Anti-PD-1 and Anti-CTLA-4 Therapy.
    Human gene therapy, 2019, Volume: 30, Issue:9

    In an effort to develop a new therapy for cancer and to improve antiprogrammed death inhibitor-1 (anti-PD-1) and anticytotoxic T lymphocyte-associated protein (anti-CTLA-4) responses, we have created a telomerase reverse transcriptase promoter-regulated oncolytic adenovirus rAd.sT containing a soluble transforming growth factor receptor II fused with human IgG Fc fragment (sTGFβRIIFc) gene. Infection of breast and renal tumor cells with rAd.sT produced sTGFβRIIFc protein with dose-dependent cytotoxicity. In immunocompetent mouse 4T1 breast tumor model, intratumoral delivery of rAd.sT inhibited both tumor growth and lung metastases. rAd.sT downregulated the expression of several transforming growth factor β (TGFβ) target genes involved in tumor growth and metastases, inhibited Th2 cytokine expression, and induced Th1 cytokines and chemokines, and granzyme B and perforin expression. rAd.sT treatment also increased the percentage of CD8

    Topics: Adenoviridae; Animals; Antineoplastic Agents, Immunological; Cell Line, Tumor; Combined Modality Therapy; CTLA-4 Antigen; Cytokines; Disease Models, Animal; Gene Transfer Techniques; Genetic Vectors; Humans; Immunity; Immunomodulation; Mice; Neoplasms; Oncolytic Virotherapy; Oncolytic Viruses; Programmed Cell Death 1 Receptor; Signal Transduction; T-Lymphocyte Subsets; Transduction, Genetic; Transforming Growth Factor beta; Virus Replication; Xenograft Model Antitumor Assays

2019
Tumour-derived transforming growth factor-β signalling contributes to fibrosis in patients with cancer cachexia.
    Journal of cachexia, sarcopenia and muscle, 2019, Volume: 10, Issue:5

    Cachexia is a paraneoplastic syndrome related with poor prognosis. The tumour micro-environment contributes to systemic inflammation and increased oxidative stress as well as to fibrosis. The aim of the present study was to characterise the inflammatory circulating factors and tumour micro-environment profile, as potentially contributing to tumour fibrosis in cachectic cancer patients.. 74 patients (weight stable cancer n = 31; cachectic cancer n = 43) diagnosed with colorectal cancer were recruited, and tumour biopsies were collected during surgery. Multiplex assay was performed to study inflammatory cytokines and growth factors. Immunohistochemistry analysis was carried out to study extracellular matrix components.. Higher protein expression of inflammatory cytokines and growth factors such as epidermal growth factor, granulocyte-macrophage colony-stimulating factor, interferon-α, and interleukin (IL)-8 was observed in the tumour and serum of cachectic cancer patients in comparison with weight-stable counterparts. Also, IL-8 was positively correlated with weight loss in cachectic patients (P = 0.04; r = 0.627). Immunohistochemistry staining showed intense collagen deposition (P = 0.0006) and increased presence of α-smooth muscle actin (P < 0.0001) in tumours of cachectic cancer patients, characterizing fibrosis. In addition, higher transforming growth factor (TGF)-β1, TGF-β2, and TGF-β3 expression (P = 0.003, P = 0.05, and P = 0.047, respectively) was found in the tumour of cachectic patients, parallel to p38 mitogen-activated protein kinase alteration. Hypoxia-inducible factor-1α mRNA content was significantly increased in the tumour of cachectic patients, when compared with weight-stable group (P = 0.005).. Our results demonstrate TGF-β pathway activation in the tumour in cachexia, through the (non-canonical) mitogen-activated protein kinase pathway. The results show that during cachexia, intratumoural inflammatory response contributes to the onset of fibrosis. Tumour remodelling, probably by TGF-β-induced transdifferentiation of fibroblasts to myofibroblasts, induces unbalanced inflammatory cytokine profile, angiogenesis, and elevation of extracellular matrix components (EMC). We speculate that these changes may affect tumour aggressiveness and present consequences in peripheral organs.

    Topics: Aged; Biomarkers; Biopsy; Body Composition; Body Mass Index; Cachexia; Cells, Cultured; Cytokines; Female; Fibroblasts; Fibrosis; Gene Expression; Humans; Hypoxia; Immunohistochemistry; Male; Middle Aged; Neoplasms; Oxidative Stress; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2019
Blockade of TGF-β signaling to enhance the antitumor response is accompanied by dysregulation of the functional activity of CD4
    Journal of translational medicine, 2019, 07-09, Volume: 17, Issue:1

    The pleiotropic cytokine, transforming growth factor (TGF)-β, and CD4. Using BALB/c, FoxP3eGFP and Rag. SM16 abrogates TGF-β-induced Treg generation in vitro but does not prevent global homeostatic expansion of CD4. These findings suggest that blockade of TGF-β signaling is a potentially useful strategy for blunting Treg function to enhance the anti-tumor response. Our data further suggest that the overall dampening of Treg function may involve the expansion of a quiescent Treg precursor population, which is CD4

    Topics: Animals; Antineoplastic Agents; CD4 Antigens; CD4-Positive T-Lymphocytes; Cell Count; Cell Line, Tumor; Cell Proliferation; Female; Forkhead Transcription Factors; Homeostasis; Interferon-gamma; Interleukin-2 Receptor alpha Subunit; Lymph Nodes; Mice, Knockout; Neoplasms; Signal Transduction; Spleen; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Burden

2019
Runx2-I is an Early Regulator of Epithelial-Mesenchymal Cell Transition in the Chick Embryo.
    Developmental dynamics : an official publication of the American Association of Anatomists, 2018, Volume: 247, Issue:3

    Although normally linked to bone and cartilage development, the Runt-related transcription factor, RUNX2, was reported in the mouse heart during development of the valves. We examined RUNX2 expression and function in the developing avian heart as it related to the epithelial-mesenchymal transition (EMT) in the atrioventricular canal. EMT can be separated into an activation stage involving hypertrophy and cell separation and an invasion stage where cells invade the extracellular matrix. The localization and activity of RUNX2 was explored in relation to these steps in the heart. As RUNX2 was also reported in cancer tissues, we examined its expression in the progression of esophageal cancer in staged tissues.. A specific isoform, RUNX2-I, is present and required for EMT by endothelia of the atrioventricular canal. Knockdown of RUNX2-I inhibits the cell-cell separation that is characteristic of initial activation of EMT. Loss of RUNX2-I altered expression of EMT markers to a greater extent during activation than during subsequent cell invasion. Transforming growth factor beta 2 (TGFβ2) mediates activation during cardiac endothelial EMT. Consistent with a role in activation, RUNX2-I is regulated by TGFβ2 and its activity is independent of similarly expressed Snai2 in regulation of EMT. Examination of RUNX2 expression in esophageal cancer showed its upregulation concomitant with the development of dysplasia and continued expression in adenocarcinoma.. These data introduce the RUNX2-I isoform as a critical early transcription factor mediating EMT in the developing heart after induction by TGFβ2. Its expression in tumor tissue suggests a similar role for RUNX2 in the EMT of metastasis. Developmental Dynamics 247:542-554, 2018. © 2017 Wiley Periodicals, Inc.

    Topics: Animals; Chick Embryo; Chickens; Core Binding Factor Alpha 1 Subunit; Epithelial-Mesenchymal Transition; Neoplasms; Protein Isoforms; Transcriptional Activation; Transforming Growth Factor beta

2018
Immune checkpoint blockade combined with IL-6 and TGF-β inhibition improves the therapeutic outcome of mRNA-based immunotherapy.
    International journal of cancer, 2018, 08-01, Volume: 143, Issue:3

    Improved understanding of cancer immunology has provided insight into the phenomenon of frequent tumor recurrence after initially successful immunotherapy. A delicate balance exists between the capacity of the immune system to control tumor growth and various resistance mechanisms that arise to avoid or even counteract the host's immune system. These resistance mechanisms include but are not limited to (i) adaptive expression of inhibitory checkpoint molecules in response to the proinflammatory environment and (ii) amplification of cancer stem cells, a small fraction of tumor cells possessing the capacity for self-renewal and mediating treatment resistance and formation of metastases after long periods of clinical remission. Several individual therapeutic agents have so far been developed to revert T-cell exhaustion or disrupt the cross-talk between cancer stem cells and the tumor-promoting microenvironment. Here, we demonstrate that a three-arm combination therapy-consisting of an mRNA-based vaccine to induce antigen-specific T-cell responses, monoclonal antibodies blocking inhibitory checkpoint molecules (PD-1, TIM-3, LAG-3), and antibodies targeting IL-6 and TGF-β-improves the therapeutic outcome in subcutaneous TC-1 tumors and significantly prolongs survival of treated mice. Our findings point to a need for a rational development of multidimensional anticancer therapies, aiming at the induction of tumor-specific immunity and simultaneously targeting multiple resistance mechanisms.

    Topics: Animals; Antineoplastic Agents, Immunological; Cell Line, Tumor; Disease Models, Animal; Gene Expression; Humans; Immunotherapy; Interleukin-6; Melanoma, Experimental; Mice; Neoplasms; Recurrence; RNA, Messenger; Signal Transduction; SOXB1 Transcription Factors; Transforming Growth Factor beta

2018
TGFβ Promotes Immune Evasion to Limit the Efficacy of Anti-PD-1/PD-L1.
    Cancer discovery, 2018, Volume: 8, Issue:4

    The TGFβ-activated stroma induces T-cell exclusion to suppress antitumor immunity.

    Topics: B7-H1 Antigen; Humans; Immune Evasion; Neoplasms; Programmed Cell Death 1 Receptor; T-Lymphocytes; Transforming Growth Factor beta

2018
Immunotherapy: Tear down this wall.
    Nature reviews. Immunology, 2018, Volume: 18, Issue:4

    Topics: B7-H1 Antigen; Humans; Immunotherapy; Neoplasms; T-Lymphocytes; Transforming Growth Factor beta

2018
Oncogenic Signaling Pathways in The Cancer Genome Atlas.
    Cell, 2018, 04-05, Volume: 173, Issue:2

    Genetic alterations in signaling pathways that control cell-cycle progression, apoptosis, and cell growth are common hallmarks of cancer, but the extent, mechanisms, and co-occurrence of alterations in these pathways differ between individual tumors and tumor types. Using mutations, copy-number changes, mRNA expression, gene fusions and DNA methylation in 9,125 tumors profiled by The Cancer Genome Atlas (TCGA), we analyzed the mechanisms and patterns of somatic alterations in ten canonical pathways: cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGFβ signaling, p53 and β-catenin/Wnt. We charted the detailed landscape of pathway alterations in 33 cancer types, stratified into 64 subtypes, and identified patterns of co-occurrence and mutual exclusivity. Eighty-nine percent of tumors had at least one driver alteration in these pathways, and 57% percent of tumors had at least one alteration potentially targetable by currently available drugs. Thirty percent of tumors had multiple targetable alterations, indicating opportunities for combination therapy.

    Topics: Databases, Genetic; Genes, Neoplasm; Humans; Neoplasms; Phosphatidylinositol 3-Kinases; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Wnt Proteins

2018
The Immune Landscape of Cancer.
    Immunity, 2018, 04-17, Volume: 48, Issue:4

    We performed an extensive immunogenomic analysis of more than 10,000 tumors comprising 33 diverse cancer types by utilizing data compiled by TCGA. Across cancer types, we identified six immune subtypes-wound healing, IFN-γ dominant, inflammatory, lymphocyte depleted, immunologically quiet, and TGF-β dominant-characterized by differences in macrophage or lymphocyte signatures, Th1:Th2 cell ratio, extent of intratumoral heterogeneity, aneuploidy, extent of neoantigen load, overall cell proliferation, expression of immunomodulatory genes, and prognosis. Specific driver mutations correlated with lower (CTNNB1, NRAS, or IDH1) or higher (BRAF, TP53, or CASP8) leukocyte levels across all cancers. Multiple control modalities of the intracellular and extracellular networks (transcription, microRNAs, copy number, and epigenetic processes) were involved in tumor-immune cell interactions, both across and within immune subtypes. Our immunogenomics pipeline to characterize these heterogeneous tumors and the resulting data are intended to serve as a resource for future targeted studies to further advance the field.

    Topics: Adolescent; Adult; Aged; Aged, 80 and over; Child; Female; Genomics; Humans; Interferon-gamma; Macrophages; Male; Middle Aged; Neoplasms; Prognosis; Th1-Th2 Balance; Transforming Growth Factor beta; Wound Healing; Young Adult

2018
TGF-β Inhibition and Immunotherapy: Checkmate.
    Immunity, 2018, 04-17, Volume: 48, Issue:4

    Immune checkpoint therapy can induce durable remissions, but many tumors demonstrate resistance. In a recent issue of Nature, Mariathasan et al. (2018) and Tauriello et al. (2018) identify stromal TGF-β signaling as a determinant of immune exclusion. Combination TGF-β inhibition and immunotherapy induces complete responses in mouse models.

    Topics: Animals; Disease Models, Animal; Immunotherapy; Mice; Neoplasms; T-Lymphocytes; Transforming Growth Factor beta

2018
Suppression of TGFβ-mediated conversion of endothelial cells and fibroblasts into cancer associated (myo)fibroblasts via HDAC inhibition.
    British journal of cancer, 2018, Volume: 118, Issue:10

    Cancer-associated fibroblasts (CAFs) support tumour progression and invasion, and they secrete abundant extracellular matrix (ECM) that may shield tumour cells from immune checkpoint or kinase inhibitors. Targeting CAFs using drugs that revert their differentiation, or inhibit their tumour-supportive functions, has been considered as an anti-cancer strategy.. We have used human and murine cell culture models, atomic force microscopy (AFM), microarray analyses, CAF/tumour cell spheroid co-cultures and transgenic fibroblast reporter mice to study how targeting HDACs using small molecule inhibitors or siRNAs re-directs CAF differentiation and function in vitro and in vivo.. From a small molecule screen, we identified Scriptaid, a selective inhibitor of HDACs 1/3/8, as a repressor of TGFβ-mediated CAF differentiation. Scriptaid inhibits ECM secretion, reduces cellular contraction and stiffness, and impairs collective cell invasion in CAF/tumour cell spheroid co-cultures. Scriptaid also reduces CAF abundance and delays tumour growth in vivo.. Scriptaid is a well-tolerated and effective HDACi that reverses many of the functional and phenotypic properties of CAFs. Impeding or reversing CAF activation/function by altering the cellular epigenetic regulatory machinery could control tumour growth and invasion, and be beneficial in combination with additional therapies that target cancer cells or immune cells directly.

    Topics: Animals; Cancer-Associated Fibroblasts; Cell Differentiation; Cell Line, Tumor; Cell Movement; Coculture Techniques; Endothelial Cells; Extracellular Matrix; Fibroblasts; Histone Deacetylase Inhibitors; Humans; Hydroxylamines; Mice; Microarray Analysis; Microscopy, Atomic Force; Neoplasms; Quinolines; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2018
A PDMS Device Coupled with Culture Dish for In Vitro Cell Migration Assay.
    Applied biochemistry and biotechnology, 2018, Volume: 186, Issue:3

    Cell migration and invasion are important factors during tumor progression and metastasis. Wound-healing assay and the Boyden chamber assay are efficient tools to investigate tumor development because both of them could be applied to measure cell migration rate. Therefore, a simple and integrated polydimethylsiloxane (PDMS) device was developed for cell migration assay, which could perform quantitative evaluation of cell migration behaviors, especially for the wound-healing assay. The integrated device was composed of three units, which included cell culture dish, PDMS chamber, and wound generation mold. The PDMS chamber was integrated with cell culture chamber and could perform six experiments under different conditions of stimuli simultaneously. To verify the function of this device, it was utilized to explore the tumor cell migration behaviors under different concentrations of fetal bovine serum (FBS) and transforming growth factor (TGF-β) at different time points. This device has the unique capability to create the "wound" area in parallel during cell migration assay and provides a simple and efficient platform for investigating cell migration assay in biomedical application.

    Topics: Cell Culture Techniques; Cell Line, Tumor; Cell Migration Assays; Cell Movement; Culture Media; Dimethylpolysiloxanes; Equipment Design; Humans; Neoplasms; Transforming Growth Factor beta; Wound Healing

2018
Metastatic cancers promote cachexia through ZIP14 upregulation in skeletal muscle.
    Nature medicine, 2018, Volume: 24, Issue:6

    Patients with metastatic cancer experience a severe loss of skeletal muscle mass and function known as cachexia. Cachexia is associated with poor prognosis and accelerated death in patients with cancer, yet its underlying mechanisms remain poorly understood. Here, we identify the metal-ion transporter ZRT- and IRT-like protein 14 (ZIP14) as a critical mediator of cancer-induced cachexia. ZIP14 is upregulated in cachectic muscles of mice and in patients with metastatic cancer and can be induced by TNF-α and TGF-β cytokines. Strikingly, germline ablation or muscle-specific depletion of Zip14 markedly reduces muscle atrophy in metastatic cancer models. We find that ZIP14-mediated zinc uptake in muscle progenitor cells represses the expression of MyoD and Mef2c and blocks muscle-cell differentiation. Importantly, ZIP14-mediated zinc accumulation in differentiated muscle cells induces myosin heavy chain loss. These results highlight a previously unrecognized role for altered zinc homeostasis in metastatic cancer-induced muscle wasting and implicate ZIP14 as a therapeutic target for its treatment.

    Topics: Animals; Cachexia; Cation Transport Proteins; Cell Differentiation; Cell Line; Cytokines; Disease Models, Animal; Humans; Mice, Inbred C57BL; Muscle, Skeletal; Myosin Heavy Chains; Neoplasm Metastasis; Neoplasms; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Up-Regulation; Zinc

2018
AMP-activated protein kinase promotes epithelial-mesenchymal transition in cancer cells through Twist1 upregulation.
    Journal of cell science, 2018, 07-26, Volume: 131, Issue:14

    The developmental programme of epithelial-mesenchymal transition (EMT), involving loss of epithelial and acquisition of mesenchymal properties, plays an important role in the invasion-metastasis cascade of cancer cells. In the present study, we show that activation of AMP-activated protein kinase (AMPK) using A769662 led to a concomitant induction of EMT in multiple cancer cell types, as observed by enhanced expression of mesenchymal markers, decrease in epithelial markers, and increase in migration and invasion. In contrast, inhibition or depletion of AMPK led to a reversal of EMT. Importantly, AMPK activity was found to be necessary for the induction of EMT by physiological cues such as hypoxia and TGFβ treatment. Furthermore, AMPK activation increased the expression and nuclear localization of Twist1, an EMT transcription factor. Depletion of Twist1 impaired AMPK-induced EMT phenotypes, suggesting that AMPK might mediate its effects on EMT, at least in part, through Twist1 upregulation. Inhibition or depletion of AMPK also attenuated metastasis. Thus, our data underscore a central role for AMPK in the induction of EMT and in metastasis, suggesting that strategies targeting AMPK might provide novel approaches to curb cancer spread.

    Topics: AMP-Activated Protein Kinases; Cell Line, Tumor; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Nuclear Proteins; Protein Transport; Transforming Growth Factor beta; Twist-Related Protein 1; Up-Regulation

2018
Radiation-Induced Myofibroblasts Promote Tumor Growth via Mitochondrial ROS-Activated TGFβ Signaling.
    Molecular cancer research : MCR, 2018, Volume: 16, Issue:11

    Fibroblasts are a key stromal cell in the tumor microenvironment (TME) and promote tumor growth via release of various growth factors. Stromal fibroblasts in cancer, called cancer-associated fibroblasts (CAF), are related to myofibroblasts, an activated form of fibroblast. While investigating the role of stroma fibroblasts on radiation-related carcinogenesis, it was observed following long-term fractionated radiation (FR) that the morphology of human diploid fibroblasts changed from smaller spindle shapes to larger flat shapes. These cells expressed smooth muscle actin (α-SMA) and platelet-derived growth factor receptors, markers of myofibroblasts and CAFs, respectively. Long-term FR induces progressive damage to the fibroblast nucleus and mitochondria via increases in mitochondrial reactive oxygen species (ROS) levels. Here, it is demonstrated that long-term FR-induced α-SMA-positive cells have decreased mitochondrial membrane potential and activated oxidative stress responses. Antioxidant N-acetyl cysteine suppressed radiation-induced mitochondrial damage and generation of myofibroblasts. These results indicate that mitochondrial ROS are associated with the acquisition of myofibroblasts after long-term FR. Mechanistically, mitochondrial ROS activated TGFβ signaling which in turn mediated the expression of α-SMA in radiation-induced myofibroblasts. Finally,

    Topics: Animals; Cell Differentiation; HeLa Cells; Humans; Lung; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Mitochondria; Myofibroblasts; Neoplasms; Oxidative Stress; Reactive Oxygen Species; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2018
Hsa-miR-5582-3P regulatory effect on TGFβ signaling through targeting of TGFβ-R1, TGFβ-R2, SMAD3, and SMAD4 transcripts.
    Journal of cellular biochemistry, 2018, Volume: 119, Issue:12

    Transforming growth factor β (TGFβ) signaling pathway which is regulated by factors such as microRNAs (miRNAs) has pivotal roles in various cellular processes. Here, we intended to verify bioinformatics predicted regulatory effect of hsa-miR-5582-3P against TGFβ/SMAD signaling pathway components. Quantitative reverse-transcription polymerase chain reaction (RT-qPCR) analysis indicated a negative correlation of expression between hsa-miR-5582-3P against TGFβ-R1, TGFβ-R2, SMAD3, and SMAD4 putative target genes in all of tested cell lines. Also, hsa-miR-5582-3P was significantly downregulated in glioma, breast, and ovarian tumor tissues compared with their normal pairs, detected by RT-qPCR. Then dual luciferase assay supported direct interaction between this miRNA and TGFβ-R1, TGFβ-R2, SMAD3, and SMAD4, 3' untranslated region sequences. Western blot analysis confirmed negative effect of hsa-miR-5582-3P overexpression on at least TGFβ-R1 expression. Consistently, hsa-miR-5582-3P overexpression brought about downregulation of TGFβ-R1, TGFβ-R2, SMAD3, and SMAD4 expression in HCT-116 cell line, followed by cell cycle arrest in sub-G1 phase, detected by flow cytometry. Altogether, our data suggest that hsa-miR-5582-3P reduces the TGFβ/SMAD signaling pathway through downregulation of TGFβ-R1, TGFβ-R2, SMAD3, and SMAD4 transcripts. These data introduce hsa-miR-5582-3P as a potential tumor suppressors-miR and a therapy candidate to be tested in cancers in which TGFβ/SMAD is deregulated.

    Topics: Female; Gene Expression Regulation, Neoplastic; HCT116 Cells; HEK293 Cells; Humans; MicroRNAs; Neoplasms; Signal Transduction; Smad3 Protein; Smad4 Protein; Transforming Growth Factor beta

2018
From individual to collective 3D cancer dissemination: roles of collagen concentration and TGF-β.
    Scientific reports, 2018, 08-24, Volume: 8, Issue:1

    Cancer cells have the ability to migrate from the primary (original) site to other places in the body. The extracellular matrix affects cancer cell migratory capacity and has been correlated with tissue-specific spreading patterns. However, how the matrix orchestrates these behaviors remains unclear. Here, we investigated how both higher collagen concentrations and TGF-β regulate the formation of H1299 cell (a non-small cell lung cancer cell line) spheroids within 3D collagen-based matrices and promote cancer cell invasive capacity. We show that at low collagen concentrations, tumor cells move individually and have moderate invasive capacity, whereas when the collagen concentration is increased, the formation of cell clusters is promoted. In addition, when the concentration of TGF-β in the microenvironment is lower, most of the clusters are aggregates of cancer cells with a spheroid-like morphology and poor migratory capacity. In contrast, higher concentrations of TGF-β induced the formation of clusters with a notably higher invasive capacity, resulting in clear strand-like collective cell migration. Our results show that the concentration of the extracellular matrix is a key regulator of the formation of tumor clusters that affects their development and growth. In addition, chemical factors create a microenvironment that promotes the transformation of idle tumor clusters into very active, invasive tumor structures. These results collectively demonstrate the relevant regulatory role of the mechano-chemical microenvironment in leading the preferential metastasis of tumor cells to specific tissues with high collagen concentrations and TFG-β activity.

    Topics: Actins; Animals; Cattle; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Shape; Collagen; Humans; Hydrogel, Polyethylene Glycol Dimethacrylate; Imaging, Three-Dimensional; Microfluidics; Multivariate Analysis; Neoplasms; Porosity; Spheroids, Cellular; Transforming Growth Factor beta

2018
TGF-β signaling in cancer.
    Acta biochimica et biophysica Sinica, 2018, Oct-01, Volume: 50, Issue:10

    Signals from the transforming growth factor-β (TGF-β) superfamily mediate a broad spectrum of cellular processes and are deregulated in many diseases, including cancer. TGF-β signaling has dual roles in tumorigenesis. In the early phase of tumorigenesis, TGF-β has tumor suppressive functions, primarily through cell cycle arrest and apoptosis. However, in the late stage of cancer, TGF-β acts as a driver of tumor progression and metastasis by increasing tumor cell invasiveness and migration and promoting chemo-resistance. Here, we briefly review the mechanisms and functions of TGF-β signaling during tumor progression and discuss the therapeutic potentials of targeting the TGF-β pathway in cancer.

    Topics: Antineoplastic Agents; Carcinogenesis; Cell Movement; Humans; Molecular Targeted Therapy; Neoplasm Invasiveness; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2018
A Pan-Cancer Analysis Reveals High-Frequency Genetic Alterations in Mediators of Signaling by the TGF-β Superfamily.
    Cell systems, 2018, 10-24, Volume: 7, Issue:4

    We present an integromic analysis of gene alterations that modulate transforming growth factor β (TGF-β)-Smad-mediated signaling in 9,125 tumor samples across 33 cancer types in The Cancer Genome Atlas (TCGA). Focusing on genes that encode mediators and regulators of TGF-β signaling, we found at least one genomic alteration (mutation, homozygous deletion, or amplification) in 39% of samples, with highest frequencies in gastrointestinal cancers. We identified mutation hotspots in genes that encode TGF-β ligands (BMP5), receptors (TGFBR2, AVCR2A, and BMPR2), and Smads (SMAD2 and SMAD4). Alterations in the TGF-β superfamily correlated positively with expression of metastasis-associated genes and with decreased survival. Correlation analyses showed the contributions of mutation, amplification, deletion, DNA methylation, and miRNA expression to transcriptional activity of TGF-β signaling in each cancer type. This study provides a broad molecular perspective relevant for future functional and therapeutic studies of the diverse cancer pathways mediated by the TGF-β superfamily.

    Topics: Bone Morphogenetic Protein 5; DNA Methylation; Humans; MicroRNAs; Mutation Rate; Neoplasms; Receptor, Transforming Growth Factor-beta Type I; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2018
Long-Term In Vitro Expansion of Epithelial Stem Cells Enabled by Pharmacological Inhibition of PAK1-ROCK-Myosin II and TGF-β Signaling.
    Cell reports, 2018, 10-16, Volume: 25, Issue:3

    Despite substantial self-renewal capability in vivo, epithelial stem and progenitor cells located in various tissues expand for a few passages in vitro in feeder-free condition before they succumb to growth arrest. Here, we describe the EpiX method, which utilizes small molecules that inhibit PAK1-ROCK-Myosin II and TGF-β signaling to achieve over one trillion-fold expansion of human epithelial stem and progenitor cells from skin, airway, mammary, and prostate glands in the absence of feeder cells. Transcriptomic and epigenomic studies show that this condition helps epithelial cells to overcome stresses for continuous proliferation. EpiX-expanded basal epithelial cells differentiate into mature epithelial cells consistent with their tissue origins. Whole-genome sequencing reveals that the cells retain remarkable genome integrity after extensive in vitro expansion without acquiring tumorigenicity. EpiX technology provides a solution to exploit the potential of tissue-resident epithelial stem and progenitor cells for regenerative medicine.

    Topics: Animals; Cell Differentiation; Cell Proliferation; Cells, Cultured; Epithelial Cells; Feeder Cells; Gene Expression Regulation; Humans; In Vitro Techniques; Keratinocytes; Myosin Type II; Neoplasms; p21-Activated Kinases; rho-Associated Kinases; Signal Transduction; Small Molecule Libraries; Stem Cells; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2018
Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells.
    JCI insight, 2018, 10-18, Volume: 3, Issue:20

    TGF-β is a promising immunotherapeutic target. It is expressed ubiquitously in a latent form that must be activated to function. Determination of where and how latent TGF-β (L-TGF-β) is activated in the tumor microenvironment could facilitate cell- and mechanism-specific approaches to immunotherapeutically target TGF-β. Binding of L-TGF-β to integrin αvβ8 results in activation of TGF-β. We engineered and used αvβ8 antibodies optimized for blocking or detection, which - respectively - inhibit tumor growth in syngeneic tumor models or sensitively and specifically detect β8 in human tumors. Inhibition of αvβ8 potentiates cytotoxic T cell responses and recruitment of immune cells to tumor centers - effects that are independent of PD-1/PD-L1. β8 is expressed on the cell surface at high levels by tumor cells, not immune cells, while the reverse is true of L-TGF-β, suggesting that tumor cell αvβ8 serves as a platform for activating cell-surface L-TGF-β presented by immune cells. Transcriptome analysis of tumor-associated lymphoid cells reveals macrophages as a key cell type responsive to β8 inhibition with major increases in chemokine and tumor-eliminating genes. High β8 expression in tumor cells is seen in 20%-80% of various cancers, which rarely coincides with high PD-L1 expression. These data suggest tumor cell αvβ8 is a PD-1/PD-L1-independent immunotherapeutic target.

    Topics: Animals; Antineoplastic Agents, Immunological; B7-H1 Antigen; Cell Line, Tumor; Computer Simulation; Disease Models, Animal; Female; Humans; Integrins; Kaplan-Meier Estimate; Macrophages; Male; Mice; Mice, Transgenic; Neoplasms; Programmed Cell Death 1 Receptor; Signal Transduction; Transforming Growth Factor beta; Tumor Escape; Tumor Microenvironment

2018
Increased Cthrc1 Activates Normal Fibroblasts and Suppresses Keloid Fibroblasts by Inhibiting TGF-β/Smad Signal Pathway and Modulating YAP Subcellular Location.
    Current medical science, 2018, Volume: 38, Issue:5

    Keloid may induce severe impairment of life quality for the patients, although keloid is a cutaneous benign tumor. Collagen triple helix repeat containing protein 1 (Cthrc1) was identified as a novel gene that was originally found in adventitial fibroblasts after arterial injury. To address the role of Cthrc1 in keloid, the expression level of Cthrc1 was assessed in normal skin and keloid tissue, as well as in normal fibroblasts (NFs) and keloid fibroblasts (KFs) by using quantitative PCR, Western blotting and immunohistochemical analysis. The results showed that Cthrc1 was increased in keloid tissue and KFs as compared with normal skin and NFs. Meanwhile, CCK8 and Transwell assays found the cellular proliferation and migration of KFs were increased as compared with NFs. Further, to verify the function of Cthrc1 in NFs and KFs, we increased Cthrc1 expression by transfecting lentivirus (LV) vectors LV-Cthrc1. The cellular proliferation and migration, collagen synthesis and the influence on TGF-β and YAP signaling were tested. The cellular proliferation and migration were increased in NFs-Cthrc1 as compared with NFs-control. Meanwhile, YAP expression and nuclear-location was increased in NFs-Cthrc1. On the contrary, when Cthrc1 was overexpressed in KFs, the cellular migration was suppressed and YAP expression was reduced and transferred to cytoplasm in KFs-Cthrc1 as compared with KFs-control. But the expression level of collagen I was decreased and pSmad2/3 nucleus transfer was suppressed in both NFs-Cthrc1 and KFs-Cthrc1 by using Western blotting and immunofluorescence. Increased Cthrc1 activated NFs by promoting YAP nucleus translocation, whereas suppressed KFs by inhibiting YAP nucleus translocation. Enhanced Cthrc1 decreased collagen I in both NFs and KFs by inhibiting TGF-β/Smad pathway. In conclusion, Cthrc1 may play a role in the pathogenesis of keloid by inhibiting collagen synthesis and fibroblasts migration via suppressing TGF-β/Smad pathway and YAP nucleus translocation.

    Topics: Adaptor Proteins, Signal Transducing; Adventitia; Extracellular Matrix Proteins; Fibroblasts; Gene Expression Regulation, Neoplastic; Genetic Vectors; Humans; Keloid; Lentivirus; Neoplasms; Phosphoproteins; Signal Transduction; Skin Neoplasms; Smad Proteins; Transcription Factors; Transfection; Transforming Growth Factor beta; YAP-Signaling Proteins

2018
The four-dimensional Kirschner-Panetta type cancer model: How to obtain tumor eradication?
    Mathematical biosciences and engineering : MBE, 2018, 10-01, Volume: 15, Issue:5

    In this paper we examine ultimate dynamics of the four-dimensional model describing interactions between tumor cells, effector immune cells, interleukin -2 and transforming growth factor-beta. This model was elaborated by Arciero et al. and is obtained from the Kirschner-Panetta type model by introducing two various treatments. We provide ultimate upper bounds for all variables of this model and two lower bounds and, besides, study when dynamics of this model possesses a global attracting set. The nonexistence conditions of compact invariant sets are derived. We obtain bounds for treatment parameters s₁₂ under which all trajectories in the positive orthant tend to the tumor-free equilibrium point. Conditions imposed on s₁₂ under which the tumor population persists are presented as well. Finally, we compare tumor eradication/ persistence bounds and discuss our results.

    Topics: Humans; Immunotherapy; Interleukin-2; Mathematical Concepts; Models, Biological; Neoplasms; Systems Biology; Transforming Growth Factor beta; Tumor Escape

2018
The Dynamics of TGF-β Signaling Are Dictated by Receptor Trafficking via the ESCRT Machinery.
    Cell reports, 2018, 11-13, Volume: 25, Issue:7

    Signal transduction pathways stimulated by secreted growth factors are tightly regulated at multiple levels between the cell surface and the nucleus. The trafficking of cell surface receptors is emerging as a key step for regulating appropriate cellular responses, with perturbations in this process contributing to human diseases, including cancer. For receptors recognizing ligands of the transforming growth factor β (TGF-β) family, little is known about how trafficking is regulated or how this shapes signaling dynamics. Here, using whole genome small interfering RNA (siRNA) screens, we have identified the ESCRT (endosomal sorting complex required for transport) machinery as a crucial determinant of signal duration. Downregulation of ESCRT components increases the outputs of TGF-β signaling and sensitizes cells to low doses of ligand in their microenvironment. This sensitization drives an epithelial-to-mesenchymal transition (EMT) in response to low doses of ligand, and we demonstrate a link between downregulation of the ESCRT machinery and cancer survival.

    Topics: Activins; Animals; Bone Morphogenetic Proteins; Cell Line; Down-Regulation; Endosomal Sorting Complexes Required for Transport; Epithelial-Mesenchymal Transition; Genome, Human; Humans; Lysosomes; Mice; Multivesicular Bodies; Neoplasms; Phosphorylation; Prognosis; Protein Transport; Proteolysis; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad2 Protein; Survival Analysis; Transforming Growth Factor beta; Ubiquitin-Protein Ligases; Up-Regulation

2018
TGF-β downregulation-induced cancer cell death is finely regulated by the SAPK signaling cascade.
    Experimental & molecular medicine, 2018, 12-06, Volume: 50, Issue:12

    Transforming growth factor (TGF)-β signaling is increasingly recognized as a key driver in cancer. In progressive cancer tissues, TGF-β promotes tumor formation, and its increased expression often correlates with cancer malignancy. In this study, we utilized adenoviruses expressing short hairpin RNAs against TGF-β1 and TGF-β2 to investigate the role of TGF-β downregulation in cancer cell death. We found that the downregulation of TGF-β increased the phosphorylation of several SAPKs, such as p38 and JNK. Moreover, reactive oxygen species (ROS) production was also increased by TGF-β downregulation, which triggered Akt inactivation and NOX4 increase-derived ROS in a cancer cell-type-specific manner. We also revealed the possibility of substantial gene fluctuation in response to TGF-β downregulation related to SAPKs. The expression levels of Trx and GSTM1, which encode inhibitory proteins that bind to ASK1, were reduced, likely a result of the altered translocation of Smad complex proteins rather than from ROS production. Instead, both ROS and ROS-mediated ER stress were responsible for the decrease in interactions between ASK1 and Trx or GSTM1. Through these pathways, ASK1 was activated and induced cytotoxic tumor cell death via p38/JNK activation and (or) induction of ER stress.

    Topics: A549 Cells; Cell Death; Down-Regulation; Endoplasmic Reticulum Stress; Humans; Intracellular Signaling Peptides and Proteins; MAP Kinase Kinase 4; MAP Kinase Kinase Kinase 5; Membrane Proteins; NADPH Oxidase 4; Neoplasms; Oncogene Protein v-akt; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Reactive Oxygen Species; RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta

2018
Tranilast-induced stress alleviation in solid tumors improves the efficacy of chemo- and nanotherapeutics in a size-independent manner.
    Scientific reports, 2017, 04-10, Volume: 7

    Accumulation of mechanical stresses during cancer progression can induce blood and lymphatic vessel compression, creating hypo-perfusion, hypoxia and interstitial hypertension which decrease the efficacy of chemo- and nanotherapies. Stress alleviation treatment has been recently proposed to reduce mechanical stresses in order to decompress tumor vessels and improve perfusion and chemotherapy. However, it remains unclear if it improves the efficacy of nanomedicines, which present numerous advantages over traditional chemotherapeutic drugs. Furthermore, we need to identify safe and well-tolerated pharmaceutical agents that reduce stress levels and may be added to cancer patients' treatment regimen. Here, we show mathematically and with a series of in vivo experiments that stress alleviation improves the delivery of drugs in a size-independent manner. Importantly, we propose the repurposing of tranilast, a clinically approved anti-fibrotic drug as stress-alleviating agent. Using two orthotopic mammary tumor models, we demonstrate that tranilast reduces mechanical stresses, decreases interstitial fluid pressure (IFP), improves tumor perfusion and significantly enhances the efficacy of different-sized drugs, doxorubicin, Abraxane and Doxil, by suppressing TGFβ signaling and expression of extracellular matrix components. Our findings strongly suggest that repurposing tranilast could be directly used as a promising strategy to enhance, not only chemotherapy, but also the efficacy of cancer nanomedicine.

    Topics: Animals; Antineoplastic Agents; Cell Line, Tumor; Collagen; Down-Regulation; Doxorubicin; Drug Delivery Systems; Extracellular Fluid; Extracellular Matrix; Gene Expression Regulation, Neoplastic; Humans; Hyaluronic Acid; Mice, Nude; Models, Biological; Nanomedicine; Neoplasms; ortho-Aminobenzoates; Particle Size; Perfusion; Signal Transduction; Stress, Mechanical; Transforming Growth Factor beta; Treatment Outcome; Tumor Microenvironment

2017
Mathematical model of TGF-βsignalling: feedback coupling is consistent with signal switching.
    BMC systems biology, 2017, 04-13, Volume: 11, Issue:1

    Transforming growth factor β (TGF-β) signalling regulates the development of embryos and tissue homeostasis in adults. In conjunction with other oncogenic changes, long-term perturbation of TGF-β signalling is associated with cancer metastasis. Although TGF-β signalling can be complex, many of the signalling components are well defined, so it is possible to develop mathematical models of TGF-β signalling using reduction and scaling methods. The parameterization of our TGF-β signalling model is consistent with experimental data.. We developed our mathematical model for the TGF-β signalling pathway, i.e. the RF- model of TGF-β signalling, using the "rapid equilibrium assumption" to reduce the network of TGF-β signalling reactions based on the time scales of the individual reactions. By adding time-delayed positive feedback to the inherent time-delayed negative feedback for TGF-β signalling. We were able to simulate the sigmoidal, switch-like behaviour observed for the concentration dependence of long-term (> 3 hours) TGF-β stimulation. Computer simulations revealed the vital role of the coupling of the positive and negative feedback loops on the regulation of the TGF-β signalling system. The incorporation of time-delays for the negative feedback loop improved the accuracy, stability and robustness of the model. This model reproduces both the short-term and long-term switching responses for the intracellular signalling pathways at different TGF-β concentrations. We have tested the model against experimental data from MEF (mouse embryonic fibroblasts) WT, SV40-immortalized MEFs and Gp130. Signalling feedback loops are required to model TGF-β signal transduction and its effects on normal and cancer cells. We focus on the effects of time-delayed feedback loops and their coupling to ligand stimulation in this system. The model was simplified and reduced to its key components using standard methods and the rapid equilibrium assumption. We detected differences in short-term and long-term signal switching. The results from the RF- model compare well with experimental data and predict the dynamics of TGF-β signalling in cancer cells with different mutations.

    Topics: Animals; Feedback, Physiological; Kinetics; Mice; Models, Biological; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2017
Tumor-fibroblast interactions stimulate tumor vascularization by enhancing cytokine-driven production of MMP9 by tumor cells.
    Oncotarget, 2017, May-30, Volume: 8, Issue:22

    Advance-stage breast carcinomas include significant amounts of fibroblasts and infiltrating immune cells which have been implicated in tumor growth, recurrence, and response to therapy. The present study investigated the contribution of fibroblasts to tumor growth using direct tumor-fibroblast co-cultures and tumor xenograft models. Our findings revealed that fibroblasts enhance breast carcinoma growth by promoting the tumor vasculature via the MMP9-dependent mechanism. In tumor-fibroblast co-cultures, fibroblasts increased expression of TGF-β, TNF, and IL-1β cytokines in tumor cells. These cytokines cooperatively induced expression of matrix metalloproteinase MMP9 in tumor cells. Knockdown of MMP9 by shRNA significantly reduced tumor vascularization induced by fibroblasts. Mechanistically, our findings argue that expression of MMP9 in tumor cellsis regulated by crosstalk of TGF-β with TNF and/or IL-1β cytokines. The mechanism of this cooperative response did not involve cross-activation of the canonical signaling pathways as TGF-β did not activate RELA/p65 signaling, while TNF did not affect SMAD signaling. Instead, TGF-β and TNF cytokines co-stimulated MAP kinases and expression of JUN and JUNB, AP1 transcription factor subunits, which together with RELA/p65 were essential for the regulation of MMP9. Depletion of JUN and JUNB or RELA in tumor cells blocked the cooperative induction of MMP9 by the cytokines. Thus, our studies uncovered a previously unappreciated role of tumor-fibroblast interactions in the stimulation of tumor angiogenesis, and an essential role of the MAPK-AP1 axis in the cooperative up-regulation of the angiogenic driver MMP9 by cytokine crosstalk.

    Topics: Animals; Apoptosis; Cancer-Associated Fibroblasts; Cell Communication; Cell Line, Tumor; Cell Proliferation; Cells, Cultured; Coculture Techniques; Cytokines; Disease Models, Animal; Female; Humans; Inflammation Mediators; MAP Kinase Kinase Kinases; Matrix Metalloproteinase 9; Mice; Neoplasms; Neovascularization, Pathologic; Rats; Signal Transduction; Transcription Factor RelA; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Xenograft Model Antitumor Assays

2017
Combined delivery of a TGF-β inhibitor and an adenoviral vector expressing interleukin-12 potentiates cancer immunotherapy.
    Acta biomaterialia, 2017, 10-01, Volume: 61

    Cancer immunotherapy appears to have a promising future, but it can be thwarted by secretion of immunosuppressive factors, such as transforming growth factor-β (TGF-β), which inhibits local immune responses to tumors. To weaken immune resistance of tumors and simultaneously strengthen immune responses, we developed a multifunctional polymer that could co-deliver hydrophobic TGF-β inhibitor and an adenovirus gene vector to tumor sites. This co-delivery system sustainably released TGF-β inhibitor SB-505124 and effectively transferred the adenovirus vector carrying the interleukin-12 gene. In addition, it significantly delayed growth of B16 melanoma xenografts in mice and increased animal survival. Mechanistic studies showed that this combination therapy enhanced anti-tumor immune response by activating CD4. To weaken immune resistance of tumors and simultaneously strengthen tumors' immune responses, we synthesized a structurally simple, low-toxic but functional polymer β-cyclodextrin-PEI to encapsulate a hydrophobic TGF-β inhibitor SB-505124 and to complex adenovirus vectors expressing IL-12. This is the first report demonstrating that combining TGF-β inhibitor with IL-12 could provide effective immunotherapy against melanoma by the sustainable release of SB-505124 and the effectible transduction of IL-12 gene in tumor cells. The rational delivery system presented a comprehensive and valued platform to be a candidate vector for co-delivering hydrophobic small-molecule drugs and therapeutic genes for treating cancer, providing a new approach for cancer immunotherapy.

    Topics: Adenoviridae; Animals; Antineoplastic Agents; Benzodioxoles; beta-Cyclodextrins; Cell Line, Tumor; Cell Movement; Combined Modality Therapy; Drug Delivery Systems; Female; Genetic Vectors; Imidazoles; Immunotherapy; Interleukin-12; Melanoma, Experimental; Mice, Inbred C57BL; Neoplasm Invasiveness; Neoplasms; Polyethyleneimine; Pyridines; Transduction, Genetic; Transforming Growth Factor beta

2017
Incidence of Cancer in Spinal Deformity Patients Receiving High-Dose (≥40 mg) Bone Morphogenetic Protein (rhBMP-2).
    Spine, 2017, Dec-01, Volume: 42, Issue:23

    Level III, Retrospective observational study.. To determine if there is an increased risk of developing cancer after exposure to high-dose recombinant human bone morphogenetic protein-2 (rhBMP-2) and if risk is dose and/or exposure-dependent.. Concerns have been raised regarding a relationship between rhBMP-2 and cancer.. A total of 642 adult deformity patients from a single institution receiving a cumulative rhBMP-2 dose ≥40 mg from July, 2002 to July, 2009 were identified. Patients with a history of surveillance, epidemiology, and end result (SEER) cancer before rhBMP-2 exposure were excluded. To determine the occurrence of a cancer event, questionnaires were mailed and telephone follow up attempted for nonresponders. Only cancers tracked by the National Cancer Institute (NCI) SEER registry were included. Observed cancer counts were compared to expected cancer counts based on general population incidence rates within 5-year age strata. Cumulative incidence competing risk (CICR) modeling was used to evaluate the association between rhBMP-2 exposure and cancer controlling for potential confounding variables.. Forty-nine patients were lost-to-follow up, leaving 593 patients (92.4%; 138 males/455 females) available for analysis. Mean age was 52.8 years at the time of first exposure. Mean cumulative rhBMP-2 dose was 113.5 mg with 85% having one exposure (range: 1-8). Mean follow-up [date of exposure to date of death (regardless of cause) or returned completed questionnaire] was 5.6 ± 1.9 years; median follow up was 5.4 years. A total of 342 patients have greater than 5-year follow up. Minimum follow up was 2.0 years or until occurrence of a SEER cancer. Our total 8-year cumulative incidence of new SEER cancer accounting for the competing risk of death was 7.4% for 30 cancers in 593 patients. Fewer cancers were observed than expected based on general population rates, though the difference was not statistically significant (expected = 34; standardized incidence ratio = 0.88, 95% confidence interval, CI = 0.60-1.26). CICR found neither cumulative rhBMP-2 dose (hazard ratio, HR = 0.995, 95% CI 0.988-1.003; P = 0.249) nor number of exposures (HR = 0.776, 95% CI 0.359-1.677; P = 0.519) increased the risk of developing a postexposure cancer after controlling for known cancer risk factors.. The incidence of a SEER cancer after rhBMP-2 exposure was similar to incidence reported by the NCI. There were no significant rhBMP-2 dose or multi-exposure related risks of developing a life-threatening cancer.. 3.

    Topics: Adult; Aged; Bone Morphogenetic Protein 2; Female; Follow-Up Studies; Humans; Incidence; Male; Middle Aged; Neoplasms; Recombinant Proteins; Registries; Retrospective Studies; Risk Factors; SEER Program; Spinal Diseases; Transforming Growth Factor beta

2017
Pattern formation in a nonlocal mathematical model for the multiple roles of the TGF-β pathway in tumour dynamics.
    Mathematical biosciences, 2017, Volume: 289

    The growth and invasion of cancer cells are very complex processes, which can be regulated by the cross-talk between various signalling pathways, or by single signalling pathways that can control multiple aspects of cell behaviour. TGF-β is one of the most investigated signalling pathways in oncology, since it can regulate multiple aspects of cell behaviour: cell proliferation and apoptosis, cell-cell adhesion and epithelial-to-mesenchimal transition via loss of cell adhesion. In this study, we use a mathematical modelling approach to investigate the complex roles of TGF-β signalling pathways on the inhibition and growth of tumours, as well as on the epithelial-to-mesenchimal transition involved in the metastasis of tumour cells. We show that the nonlocal mathematical model derived here to describe repulsive and adhesive cell-cell interactions can explain the formation of new tumour cell aggregations at positions in space that are further away from the main aggregation. Moreover, we show that the increase in cell-cell adhesion leads to fewer but larger aggregations, and the increase in TGF-β molecules - whose late-stage effect is to decrease cell adhesion - leads to many small cellular aggregations. Finally, we perform a sensitivity analysis on some parameters associated with TGF-β dynamics, and use it to investigate the relation between the tumour size and its metastatic spread.

    Topics: Animals; Cell Adhesion; Cell Aggregation; Cell Proliferation; Epithelial-Mesenchymal Transition; Humans; Mice; Models, Biological; Neoplasm Metastasis; Neoplasms; Pattern Recognition, Automated; Signal Transduction; Transforming Growth Factor beta; Tumor Burden

2017
Autophagy-dependent regulation of tumor metastasis by myeloid cells.
    PloS one, 2017, Volume: 12, Issue:7

    Autophagy is a vital process controlling the lysosomal degradation of cellular organelles and thereby regulating tissue homeostasis in an environment-dependent fashion. Recent studies have unveiled the critical role of tumor cell-derived autophagy in regulating pro-tumor and anti-tumor processes depending on different stages and tumor microenvironments. However, the precise mechanism whereby autophagy regulates tumor progression remains largely unclear. Since myeloid cells contribute to tumor progression and metastasis, we evaluated the role of myeloid cell-specific autophagy in the regulation of tumor progression. We found that the number and size of metastatic lesions were smaller in myeloid cell-specific autophagy-deficient mice. Furthermore, autophagy-mediated regulation of TGF-β in myeloid cells was associated with the induction of epithelial-mesenchymal transition (EMT), which increases the invasive and metastatic potentials of tumor cells. Myeloid-derived autophagy also plays a critical role in impairing antitumor immune responses and promoting the survival and accumulation of M2 macrophages in tumor tissues in a CSF-1 and TGF-β-dependent manner. Taken together, our findings elucidate previously unrecognized mechanisms by which myeloid cells promote tumor progression through autophagy-mediated regulation of malignancy and immune tolerance.

    Topics: Animals; Autophagy; Epithelial-Mesenchymal Transition; Humans; Lysosomes; Macrophage Colony-Stimulating Factor; Macrophages; Mice; Myeloid Cells; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2017
Stereotactic Radiotherapy Increases Functionally Suppressive Regulatory T Cells in the Tumor Microenvironment.
    Cancer immunology research, 2017, Volume: 5, Issue:11

    Radiotherapy (RT) enhances innate and adaptive antitumor immunity; however, the effects of radiation on suppressive immune cells, such as regulatory T cells (Treg), in the tumor microenvironment (TME) are not fully elucidated. Although previous reports suggest an increased Treg infiltration after radiation, whether these Tregs are functionally suppressive remains undetermined. To test the hypothesis that RT enhances the suppressive function of Treg in the TME, we selectively irradiated implanted tumors using the small animal radiation research platform (SARRP), which models stereotactic radiotherapy in human patients. We then analyzed tumor-infiltrating lymphocytes (TIL) with flow-cytometry and functional assays. Our data showed that RT significantly increased tumor-infiltrating Tregs (TIL-Treg), which had higher expression of CTLA-4, 4-1BB, and Helios compared with Tregs in nonirradiated tumors. This observation held true across several tumor models (B16/F10, RENCA, and MC38). We found that TIL-Tregs from irradiated tumors had equal or improved suppressive capacity compared with nonirradiated tumors. Our data also indicated that after RT, Tregs proliferated more robustly than other T-cell subsets in the TME. In addition, after RT, expansion of Tregs occurred when T-cell migration was inhibited using Fingolimod, suggesting that the increased Treg frequency was likely due to preferential proliferation of intratumoral Treg after radiation. Our data also suggested that Treg expansion after irradiation was independent of TGFβ and IL33 signaling. These data demonstrate that RT increased phenotypically and functionally suppressive Tregs in the TME. Our results suggest that RT might be combined effectively with Treg-targeting agents to maximize antitumor efficacy.

    Topics: Animals; Cell Line, Tumor; Female; Fingolimod Hydrochloride; Immunosuppressive Agents; Interleukin-33; Lymphocytes, Tumor-Infiltrating; Mice, Inbred BALB C; Mice, Inbred C57BL; Neoplasms; Radiosurgery; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Burden; Tumor Microenvironment

2017
Association between malignancies and Marfan syndrome: a population-based, nested case-control study in Taiwan.
    BMJ open, 2017, Oct-16, Volume: 7, Issue:10

    Marfan syndrome (MFS) involves a deficiency of the structural extracellular matrix component fibrillin-1 and overactivation of the transforming growth factor-β (TGF-β) signalling pathway. The TGF-β signalling pathway also actively participates in malignant transformation. Although anecdotal case reports have suggested associations between MFS/MFS-like conditions and several haematological and solid malignancies, such associations have not been thoroughly evaluated in large-scale studies. We sought to use a nationwide healthcare insurance claim database to evaluate whether patients with MFS are at increased risk of malignancy.. We conducted a nested case-control analysis using a database extracted from Taiwan's National Health Insurance Research Database. All medical conditions for each case and control were categorised using the International Classification of Diseases, 9th Revision classifications. ORs and 95% CIs for associations between MFS and malignancies were estimated using conditional logistic regression and adjusted for comorbidities.. Our analyses included 1 153 137 cancer cases and 1 153 137 propensity score-matched controls. Relative to other subjects, patients with MFS had a significantly higher risk of having a malignancy (adjusted OR 3.991) and hypertension (adjusted OR 1.964) and were significantly more likely to be men. Malignancies originating from the head and neck and the urinary tract were significantly more frequent among patients with MFS than among subjects without MFS.. Patients with MFS are at increased risk of developing various malignancies. Healthcare professionals should be aware of this risk when treating such patients, and increased cancer surveillance may be necessary for these patients.

    Topics: Aged; Aged, 80 and over; Case-Control Studies; Comorbidity; Female; Fibrillin-1; Humans; Logistic Models; Male; Marfan Syndrome; Middle Aged; Multivariate Analysis; Neoplasms; Taiwan; Transforming Growth Factor beta

2017
The prognostic importance of TGF-β, TGF-β receptor, and fascin in childhood solid tumors.
    Pediatric hematology and oncology, 2017, Volume: 34, Issue:4

    Fascin plays a role in tumor metastasis under the influence of TGF-β, each potentiating the effect of the other. We retrospectively investigated whether there was a prognostic relationship between TGF-β and fascin, and disease stage, local recurrence, metastasis tendency, and response to treatment. Twelve neuroblastomas, 17 osteosarcomas, 14 Ewing's sarcomas, 15 rhabdomyosarcoma cases, and 8 rare solid tumors were included. Serum TGF-β levels were high at the time of diagnosis in all groups (p = .015) and decreased significantly during remission (p = .008). Serum TGF-β values in the relapse period rarely reached high levels at the time of diagnosis and even stayed under the control group values (p = .017). When TGF-β receptor expression in tumor tissues was evaluated, the association of TGF-β receptor positivity with metastatic disease and advanced stage was striking. We found that 88% of rhabdomyosarcoma cases with alveolar histopathology expressed the TGF-β receptor, and the association between TGF-β receptor positivity and alveolar histopathology seemed to be a negative prognostic marker. When fascin levels were evaluated in childhood solid tumor tissue, the risk of relapse increased when the fascin total score at diagnosis was >4. This is one of the few studies including prognostic markers such as serum TGF-β, tissue TGF-β, TGF-β receptor, and fascin in pediatric solid tumors. Considering the poor prognosis of advanced stage pediatric solid tumors and the need for biomarkers to predict which patient might need more intensive therapy or warrant closer follow-up afterward, we think that TGF-β, TGF-β receptor, and fascin expression have an important prognostic role.

    Topics: Adolescent; Carrier Proteins; Child; Child, Preschool; Disease-Free Survival; Female; Humans; Infant; Male; Microfilament Proteins; Neoplasm Proteins; Neoplasms; Receptors, Transforming Growth Factor beta; Survival Rate; Transforming Growth Factor beta

2017
Between biology and medicine: perspectives on the use of dendritic cells in anticancer therapy.
    Postepy higieny i medycyny doswiadczalnej (Online), 2017, Nov-19, Volume: 71, Issue:0

    Dendritic cells (DCs), as a link between innate and adaptive immunity, play a pivotal role in maintaining homeostasis of the immune system. The DC population is characterized by heterogeneity; it consists of many subpopulations which, despite their phenotypic and localization differences, play an essential function - they are professional antigen presenting cells. Due to their role, DCs can be utilized in a new cancer treatment strategy. Their main purpose is to generate an anticancer response leading to the elimination of cancer cells. The tumor microenvironment, abundant in immunosuppressive factors (e.g. IL-10, TGF-β, Arg1, IDO), impairs the proper function of DCs. For this reason, various strategies are necessary for ex vivo preparation of DC-based vaccines and for the support of in vivo DCs to fight against tumors. DC-based vaccines are combined with other forms of immunotherapy (e.g. blockade of immune checkpoint molecules, e.g. PD-1 or CTLA-4) or conventional types of therapies (e.g. chemotherapy). Despite the enormous progress that has been made in anticancer therapy in the past two decades, there are still many unresolved issues regarding the effectiveness of the DCs usage. In this paper we described, in both a mouse and a human subject, a series of DC subpopulations, differentiating in normal conditions or under the influence of cancer microenvironment. We listed factors affecting the quality of the in vivo and ex vivo generations of antitumoral responses, significant from a therapeutic point of view. Moreover, the most important strategies for the use of DCs in anticancer therapies, as well as further developments on this field, have been discussed.

    Topics: Adaptive Immunity; Animals; Dendritic Cells; Humans; Immunotherapy; Interleukin-10; Mice; Neoplasms; Transforming Growth Factor beta

2017
Synergistic Chemopreventive and Therapeutic Effects of Co-drug UA-Met: Implication in Tumor Metastasis.
    Journal of agricultural and food chemistry, 2017, Dec-20, Volume: 65, Issue:50

    The anticancer properties of ursolic acid (UA) and metformin (Met) have been well demonstrated. However, whether these compounds can act synergistically to prevent and treat cancer is not known. We present in this study, the synergism between UA and Met, and that of a new codrug made of UA and Met (UA-Met) against several cancer cell lines. The combination of high concentration of UA (25, 50, 75, 100 μM) and Met (5, 10, 20, 40 mM) resulted in synergetic cytotoxicity on MDA-MB-231 and MCF-7 cells (CI < 0.8). Molecular and cellular studies showed that codrug UA-Met significantly inhibited the invasion (∼55.3 ± 2.74%) and migration (∼52.4 ± 1.57%) of TGF-β induced breast cancer MDA-MB-231 and MCF-7 cells in vitro at low concentration of 10 μM. These effects were accompanied by down-regulation of CXCR4, uPA, vimentin, E-cadherin, N-cadherin, and MMP-2/9 proteins expression and regulation of the AMPK/m-TOR signaling pathways as expected from UA and Met. Moreover, UA-Met could reduce the progression of pulmonary metastasis by 4T1 cells (63.4 ± 3.52%) without influencing the glucose blood level in mice. Our study suggests that the codrug UA-Met is safe and effective in preventing cancer metastasis and possibly treatment of cancer.

    Topics: Animals; Cadherins; Cell Line, Tumor; Drug Synergism; Female; Humans; Metformin; Mice; Neoplasm Metastasis; Neoplasms; Rats; Rats, Sprague-Dawley; Transforming Growth Factor beta; Triterpenes; Ursolic Acid; Vimentin; Xenograft Model Antitumor Assays

2017
The TGFβ superfamily in Lisbon: navigating through development and disease.
    Development (Cambridge, England), 2017, 12-15, Volume: 144, Issue:24

    The 10th FASEB meeting 'The TGFβ Superfamily: Signaling in Development and Disease' took place in Lisbon, Portugal, in July 2017. As we review here, the findings presented at the meeting highlighted the important contributions of TGFβ family signaling to normal development, adult homeostasis and disease, and also revealed novel mechanisms by which TGFβ signals are transduced.

    Topics: Animals; Cell Transformation, Neoplastic; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2017
A TGFβ-PRMT5-MEP50 axis regulates cancer cell invasion through histone H3 and H4 arginine methylation coupled transcriptional activation and repression.
    Oncogene, 2017, 01-19, Volume: 36, Issue:3

    Protein arginine methyltransferase 5 (PRMT5) complexed with MEP50/WDR77 catalyzes arginine methylation on histones and other proteins. PRMT5-MEP50 activity is elevated in cancer cells and its expression is highly correlated with poor prognosis in many human tumors. We demonstrate that PRMT5-MEP50 is essential for transcriptional regulation promoting cancer cell invasive phenotypes in lung adenocarcinoma, lung squamous cell carcinoma and breast carcinoma cancer cells. RNA-Seq transcriptome analysis demonstrated that PRMT5 and MEP50 are required to maintain expression of metastasis and Epithelial-to-mesenchymal transition (EMT) markers and to potentiate an epigenetic mechanism of the TGFβ response. We show that PRMT5-MEP50 activity both positively and negatively regulates expression of a wide range of genes. Exogenous TGFβ promotes EMT in a unique pathway of PRMT5-MEP50 catalyzed histone mono- and dimethylation of chromatin at key metastasis suppressor and EMT genes, defining a new mechanism regulating cancer invasivity. PRMT5 methylation of histone H3R2me1 induced transcriptional activation by recruitment of WDR5 and concomitant H3K4 methylation at targeted genes. In parallel, PRMT5 methylation of histone H4R3me2s suppressed transcription at distinct genomic loci. Our decoding of histone methylarginine at key genes supports a critical role for complementary PRMT5-MEP50 transcriptional activation and repression in cancer invasion pathways and in response to TGFβ stimulation and therefore orients future chemotherapeutic opportunities.

    Topics: A549 Cells; Adaptor Proteins, Signal Transducing; Adenocarcinoma; Adenocarcinoma of Lung; Arginine; Breast Neoplasms; Carcinoma, Squamous Cell; Epigenesis, Genetic; Epithelial-Mesenchymal Transition; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Histones; Humans; Lung Neoplasms; MCF-7 Cells; Methylation; Neoplasm Invasiveness; Neoplasms; Prognosis; Protein-Arginine N-Methyltransferases; Sequence Analysis, RNA; Transcription, Genetic; Transforming Growth Factor beta

2017
M2-polarized and tumor-associated macrophages alter NK cell phenotype and function in a contact-dependent manner.
    Journal of leukocyte biology, 2017, Volume: 101, Issue:1

    The crosstalk between NK cells and M1 macrophages has a vital role in the protection against infections and tumor development. However, macrophages in the tumor resemble an M2 phenotype, and, at present, their effect on NK cells is less clear. This study investigated whether tumor-associated macrophages (TAMs) have a role in altering NK cell function and phenotype using in vitro cocultures of murine NK cells with peritoneal or bone marrow-derived, M2-polarized macrophages or TAMs isolated from spontaneous mouse breast tumors. We report here that both peritoneal and bone marrow-derived M2 macrophages, as well as TAMs, substantially inhibit NK cell activation and concordant cytotoxicity against tumor cells. The mechanism for this inhibition was found to require contact between the respective cell types. Both M2 macrophages and TAMs are producers of the immunosuppressive cytokine TGF-β. The inhibition of TGF-β restored the cytotoxicity of NK cells in contact with M2 macrophages, implicating TGF-β in the mechanism for NK cell inhibition. In addition to affecting NK cell function, TAMs also induced a CD27

    Topics: Animals; Bone Marrow Cells; Cell Communication; Cell Line, Tumor; Cell Polarity; Cell Survival; Coculture Techniques; Cytotoxicity, Immunologic; Killer Cells, Natural; Lysosomal-Associated Membrane Protein 1; Macrophages, Peritoneal; Mice, Inbred C57BL; Neoplasms; Phenotype; Transforming Growth Factor beta; Tumor Necrosis Factor Receptor Superfamily, Member 7

2017
TGF-β upregulates the translation of USP15 via the PI3K/AKT pathway to promote p53 stability.
    Oncogene, 2017, 05-11, Volume: 36, Issue:19

    Crosstalk between transforming growth factor beta (TGF-β) signaling and p53 has a critical role in cancer progression. TGF-β signals via Smad and non-Smad pathways. Under normal conditions, wild-type p53 forms a complex with Smad2/3 and co-activates transcription of a variety of tumor suppressor genes, resulting in tumor suppressive effects. Thus, p53 stability is essential in progression of tumor suppressive responses mediated by TGF-β signaling. However, it remains unknown whether p53 stability is regulated by TGF-β. In the current study, we identify that USP15 binds to and stabilizes p53 through deubiquitination in U2OS and HEK293 cells. TGF-β promotes the translation of USP15 through activation of mammalian target of rapamycin by the phosphoinositide 3-kinase/AKT pathway. Upregulation of USP15 translation links the crosstalk between TGF-β signaling and p53 stability, allowing this cytokine to have a critical role in cancer progression.

    Topics: Apoptosis; Cell Proliferation; Gene Expression Regulation, Neoplastic; HEK293 Cells; Humans; Neoplasms; Phosphatidylinositol 3-Kinases; Protein Binding; Protein Stability; Proto-Oncogene Proteins c-akt; Signal Transduction; Smad2 Protein; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Ubiquitin-Specific Proteases

2017
Intercellular Adhesion Molecule-1 and Vascular Cell Adhesion Molecule Are Induced by Ionizing Radiation on Lymphatic Endothelium.
    International journal of radiation oncology, biology, physics, 2017, 02-01, Volume: 97, Issue:2

    The goal of this study was to assess the effects of ionizing radiation on the expression of the integrin ligands ICAM-1 and VCAM that control leucocyte transit by lymphatic endothelial cells.. Confluent monolayers of primary human lymphatic endothelial cells (LEC) were irradiated with single dose of 2, 5, 10 or 20 Gy, with 6 MeV-x-rays using a Linear-Accelerator. ICAM-1 and VCAM expression was determined by flow cytometry. Human tissue specimens received a single dose of 20 Gy with 15 MeV-x-rays. MC38, B16-OVA or B16-VEGF-C tumors grown in C57BL/6 mice were irradiated with single dose of 20Gy using a Linear-Accelerator fitted with a 10mm Radiosurgery collimator. Clinical samples were obtained from patients previous and 4 weeks after complete standard radiotherapy. ICAM-1 and VCAM expression was detected in all tissue specimens by confocal microscopy. To understand the role of TGFβ in this process anti-TGFβ blocking mAb were injected i.p. 30min before radiotherapy. Cell adhesion to irradiated LEC was analyzed in adhesion experiments performed in the presence or in the absence of anti- TGFβ and /or anti-ICAM1 blocking mAb.. We demonstrate that lymphatic endothelial cells in tumor samples experience induction of surface ICAM-1 and VCAM when exposed to ionizing radiation in a dose- and time-dependent manner. These effects can be recapitulated in cultured LEC, and are in part mediated by TGFβ. These data are consistent with increases in ICAM-1 and VCAM expression on LYVE-1+ endothelial cells in freshly explanted human tumor tissue and in mouse transplanted tumors after radiotherapy. Finally, ICAM-1 and VCAM expression accounts for enhanced adherence of human T lymphocytes to irradiated LEC.. Our results show induction of ICAM-1 and VCAM on LVs in irradiated lesions and offer a starting point for elucidating the biological and therapeutic implications of targeting leukocyte traffic in combination to immunotherapy.

    Topics: Animals; Cell Adhesion; Cell Line, Tumor; Cell Movement; Dose-Response Relationship, Radiation; Endothelium, Lymphatic; Flow Cytometry; Fluoroimmunoassay; Humans; Intercellular Adhesion Molecule-1; Mice; Mice, Inbred C57BL; Neoplasms; Particle Accelerators; Radiation Dosage; Random Allocation; T-Lymphocytes; Time Factors; Transforming Growth Factor beta; Vascular Cell Adhesion Molecule-1

2017
TGF-β reduces DNA ds-break repair mechanisms to heighten genetic diversity and adaptability of CD44+/CD24- cancer cells.
    eLife, 2017, 01-16, Volume: 6

    Many lines of evidence have indicated that both genetic and non-genetic determinants can contribute to intra-tumor heterogeneity and influence cancer outcomes. Among the best described sub-population of cancer cells generated by non-genetic mechanisms are cells characterized by a CD44+/CD24- cell surface marker profile. Here, we report that human CD44+/CD24- cancer cells are genetically highly unstable because of intrinsic defects in their DNA-repair capabilities. In fact, in CD44+/CD24- cells, constitutive activation of the TGF-beta axis was both necessary and sufficient to reduce the expression of genes that are crucial in coordinating DNA damage repair mechanisms. Consequently, we observed that cancer cells that reside in a CD44+/CD24- state are characterized by increased accumulation of DNA copy number alterations, greater genetic diversity and improved adaptability to drug treatment. Together, these data suggest that the transition into a CD44+/CD24- cell state can promote intra-tumor genetic heterogeneity, spur tumor evolution and increase tumor fitness.

    Topics: CD24 Antigen; Cell Line, Tumor; DNA Breaks, Double-Stranded; DNA Repair; Gene Dosage; Genetic Variation; Humans; Hyaluronan Receptors; Mutation; Neoplasms; Transforming Growth Factor beta

2017
A Transcriptional Program for Detecting TGFβ-Induced EMT in Cancer.
    Molecular cancer research : MCR, 2017, Volume: 15, Issue:5

    Most cancer deaths are due to metastasis, and epithelial-to-mesenchymal transition (EMT) plays a central role in driving cancer cell metastasis. EMT is induced by different stimuli, leading to different signaling patterns and therapeutic responses. TGFβ is one of the best-studied drivers of EMT, and many drugs are available to target this signaling pathway. A comprehensive bioinformatics approach was employed to derive a signature for TGFβ-induced EMT which can be used to score TGFβ-driven EMT in cells and clinical specimens. Considering this signature in pan-cancer cell and tumor datasets, a number of cell lines (including basal B breast cancer and cancers of the central nervous system) show evidence for TGFβ-driven EMT and carry a low mutational burden across the TGFβ signaling pathway. Furthermore, significant variation is observed in the response of high scoring cell lines to some common cancer drugs. Finally, this signature was applied to pan-cancer data from The Cancer Genome Atlas to identify tumor types with evidence of TGFβ-induced EMT. Tumor types with high scores showed significantly lower survival rates than those with low scores and also carry a lower mutational burden in the TGFβ pathway. The current transcriptomic signature demonstrates reproducible results across independent cell line and cancer datasets and identifies samples with strong mesenchymal phenotypes likely to be driven by TGFβ.

    Topics: Cell Line, Tumor; Computational Biology; Epithelial-Mesenchymal Transition; Gene Expression Profiling; Gene Regulatory Networks; Humans; Mutation; Neoplasms; Signal Transduction; Survival Analysis; Transforming Growth Factor beta

2017
Driving cancer evolution.
    eLife, 2017, 03-10, Volume: 6

    Tumor-growth-factor-beta signaling helps cancer cells to evolve and become resistant to drugs by down-regulating accurate DNA repair.

    Topics: CD24 Antigen; DNA; DNA Repair; Genetic Variation; Humans; Hyaluronan Receptors; Neoplasms; Transforming Growth Factor beta

2017
Adipose tissue fibrosis in human cancer cachexia: the role of TGFβ pathway.
    BMC cancer, 2017, 03-14, Volume: 17, Issue:1

    Cancer cachexia is a multifactorial syndrome that dramatically decreases survival. Loss of white adipose tissue (WAT) is one of the key characteristics of cachexia. WAT wasting is paralleled by microarchitectural remodeling in cachectic cancer patients. Fibrosis results from uncontrolled ECM synthesis, a process in which, transforming growth factor-beta (TGFβ) plays a pivotal role. So far, the mechanisms involved in adipose tissue (AT) re-arrangement, and the role of TGFβ in inducing AT remodeling in weight-losing cancer patients are poorly understood. This study examined the modulation of ECM components mediated by TGFβ pathway in fibrotic AT obtained from cachectic gastrointestinal cancer patients.. After signing the informed consent form, patients were enrolled into the following groups: cancer cachexia (CC, n = 21), weight-stable cancer (WSC, n = 17), and control (n = 21). The total amount of collagen and elastic fibers in the subcutaneous AT was assessed by histological analysis and by immunohistochemistry. TGFβ isoforms expression was analyzed by Multiplex assay and by immunohistochemistry. Alpha-smooth muscle actin (αSMA), fibroblast-specific protein (FSP1), Smad3 and 4 were quantified by qPCR and/or by immunohistochemistry. Interleukin (IL) 2, IL5, IL8, IL13 and IL17 content, cytokines known to be associated with fibrosis, was measured by Multiplex assay.. There was an accumulation of collagen and elastic fibers in the AT of CC, as compared with WSC and controls. Collagens type I, III, VI, and fibronectin expression was enhanced in the tissue of CC, compared with both WSC and control. The pronounced expression of αSMA in the surrounding of adipocytes, and the increased mRNA content for FSP1 (20-fold) indicate the presence of activated myofibroblasts; particularly in CC. TGFβ1 and TGFβ3 levels were up-regulated by cachexia in AT, as well in the isolated adipocytes. Smad3 and Smad4 labeling was found to be more evident in the fibrotic areas of CC adipose tissue.. Cancer cachexia promotes the development of AT fibrosis, in association with altered TGFβ signaling, compromising AT organization and function.

    Topics: Actins; Adipose Tissue; Adult; Aged; Cachexia; Calcium-Binding Proteins; Female; Fibrosis; Gene Expression; Humans; Male; Middle Aged; Neoplasms; Protein Isoforms; S100 Calcium-Binding Protein A4; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2017
[113th Scientific Meeting of the Japanese Society of Internal Medicine: Presidential Lecture: Special Lecture: TGF-β Family and Internal Medicine].
    Nihon Naika Gakkai zasshi. The Journal of the Japanese Society of Internal Medicine, 2016, Volume: 105, Issue:9

    Topics: Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Humans; Internal Medicine; Japan; Neoplasms; Signal Transduction; Societies, Medical; Transforming Growth Factor beta

2016
Complications and cancer rates in spine fusion with recombinant human bone morphogenetic protein-2 (rhBMP-2).
    European spine journal : official publication of the European Spine Society, the European Spinal Deformity Society, and the European Section of the Cervical Spine Research Society, 2016, Volume: 25, Issue:12

    To quantitatively synthesize the available best evidence for general complications, heterotopic ossification (HO), retrograde ejaculation, cervical swelling, and cancer rates with the use of rhBMP-2 in lumbar and cervical spine fusion.. We conducted an online search for relevant controlled trials and extracted data on the abovementioned endpoints. Studies were eligible for inclusion if they reported on spinal fusion with rhBMP-2 in humans. Publication bias and heterogeneity were assessed mathematically. These data were synthesized in a meta-analysis using DerSimonian-Laird random effects modeling to calculate pooled odds ratios.. We identified 26 studies reporting on a total of 184,324 patients (28,815 experimental, 155,509 controls) with a mean age of 51.1 ± 1.8 years. There was a significantly higher risk of general complications with rhBMP-2 compared to iliac crest bone graft (ICBG) with an odds ratio (OR) of 1.78 (95 %CI 1.20-2.63), (p = 0.004). The odds ratio for HO was 5.57 (95 %CI 1.90-16.36), (p = 0.002), for retrograde ejaculation 3.31 (95 %CI 1.20-9.09), (p = 0.020), and for cervical swelling 4.72 (95 %CI 1.42-15.67), (p = 0.011), all significantly higher in the rhBMP-2 group. The pooled odds ratio for new onset of tumor was 1.35 (95 %CI 0.93-1.96), which represents no statistically significant difference between the groups (p = 0.111).. rhBMP-2 is associated with a higher rate of general complications as well as retrograde ejaculation, HO, and cervical tissue swelling in spine fusion. There is a slightly increased risk of new onset of tumors, however, without statistical significance.

    Topics: Bone Morphogenetic Protein 2; Humans; Middle Aged; Neoplasms; Ossification, Heterotopic; Recombinant Proteins; Spinal Fusion; Transforming Growth Factor beta

2016
HMGB1 enhances the protumoral activities of M2 macrophages by a RAGE-dependent mechanism.
    Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine, 2016, Volume: 37, Issue:3

    The monocyte-macrophage lineage shows a high degree of diversity and plasticity. Once they infiltrate tissues, they may acquire two main functional phenotypes, being known as the classically activated type 1 macrophages (M1) and the alternative activated type 2 macrophages (M2). The M1 phenotype can be induced by bacterial products and interferon-γ and exerts a cytotoxic effect on cancer cells. Conversely, the alternatively activated M2 phenotype is induced by Il-4/IL13 and promotes tumor cell growth and vascularization. Although receptor for advanced glycation end-products (RAGE) engagement in M1 macrophages has been reported by several groups to promote inflammation, nothing is known about the functionality of RAGE in M2 macrophages. In the current study, we demonstrate that RAGE is equally expressed in both macrophage phenotypes and that RAGE activation by high-mobility group protein box1 (HMGB1) promotes protumoral activities of M2 macrophages. MKN45 cells co-cultured with M2 macrophages treated with HMGB1 at different times displayed higher invasive abilities. Additionally, conditioned medium from HMGB1-treated M2 macrophages promotes angiogenesis in vitro. RAGE-targeting knockdown abrogates these activities. Overall, the present findings suggest that HMGB1 may contribute, by a RAGE-dependent mechanism, to the protumoral activities of the M2 phenotype.

    Topics: Blotting, Western; Cell Line; Cell Line, Tumor; Cell Proliferation; Coculture Techniques; Gene Expression; HMGB1 Protein; Humans; Interleukin-10; Interleukin-1beta; Macrophage Activation; Macrophages; Neoplasms; Nitric Oxide Synthase Type II; Receptor for Advanced Glycation End Products; Reverse Transcriptase Polymerase Chain Reaction; RNA Interference; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor-alpha

2016
Imaging TGFβ Signaling in Mouse Models of Cancer Metastasis.
    Methods in molecular biology (Clifton, N.J.), 2016, Volume: 1344

    Metastatic spread of cancer cells from the primary tumors to distant vital organs, such as lung, liver, brain, and bone, is responsible for the majority of cancer-related deaths. Development of metastatic lesions is critically dependent on the interaction of tumor cells with the stromal microenvironment. As a multifunctional paracrine signaling factor that is abundantly produced by both tumor and stromal cells, TGFβ has been well established as an important mediator of tumor-stromal interaction during cancer metastasis. Imaging the in vivo dynamic of TGFβ signaling activity during cancer metastasis is critical for understanding the pathogenesis of the disease, and for the development of effective anti-metastasis treatments. In this chapter, I describe several xenograft methods to introduce human breast cancer cells into nude mice in order to generate spontaneous and experimental metastases, as well as the luciferase-based bioluminescence imaging method for quantitative imaging analysis of TGFβ signaling in tumor cells during metastasis.

    Topics: Animals; Disease Models, Animal; Heterografts; Humans; Luminescent Measurements; Mice; Molecular Imaging; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2016
TGF-β-Regulated MicroRNAs and Their Function in Cancer Biology.
    Methods in molecular biology (Clifton, N.J.), 2016, Volume: 1344

    The transforming growth factor-β (TGF-β) is known to regulate a large number of biological processes and is involved in various aspects of tumor development. Recent studies have shown that the biogenesis of miRNAs can be regulated by TGF-β signaling directly via Smad-dependent mechanisms and/or other unknown mechanisms, which may induce autoregulatory feedback loops in response to the activation of TGF-β signaling, influencing the fate of tumor cells. In this chapter, we summarize the currently described mechanisms underlying TGF-β's regulation of miRNA biogenesis, and the functional role of TGF-β-regulated miRNAs in tumor initiation, epithelial-mesenchymal transition, and tumor microenvironment modulation. Finally, we introduce methods to study TGF-β-regulated miRNAs and their functions in tumor progression and metastasis using an example of publication from our lab demonstrating the presence of a TGF-β-miR-34a-CCL22 signaling axis, which serves as a potent etiological pathway for the development of hepatocellular carcinoma venous metastases.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Epithelial-Mesenchymal Transition; Gene Expression Regulation; Humans; Mice; MicroRNAs; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2016
Dlx-2 and glutaminase upregulate epithelial-mesenchymal transition and glycolytic switch.
    Oncotarget, 2016, Feb-16, Volume: 7, Issue:7

    Most cancer cells depend on enhanced glucose and glutamine (Gln) metabolism for growth and survival. Oncogenic metabolism provides biosynthetic precursors for nucleotides, lipids, and amino acids; however, its specific roles in tumor progression are largely unknown. We previously showed that distal-less homeobox-2 (Dlx-2), a homeodomain transcription factor involved in embryonic and tumor development, induces glycolytic switch and epithelial-mesenchymal transition (EMT) by inducing Snail expression. Here we show that Dlx-2 also induces the expression of the crucial Gln metabolism enzyme glutaminase (GLS1), which converts Gln to glutamate. TGF-β and Wnt induced GLS1 expression in a Dlx-2-dependent manner. GLS1 shRNA (shGLS1) suppressed in vivo tumor metastasis and growth. Inhibition of Gln metabolism by shGLS1, Gln deprivation, and Gln metabolism inhibitors (DON, 968 and BPTES) prevented Dlx-2-, TGF-β-, Wnt-, and Snail-induced EMT and glycolytic switch. Finally, shDlx-2 and Gln metabolism inhibition decreased Snail mRNA levels through p53-dependent upregulation of Snail-targeting microRNAs. These results demonstrate that the Dlx-2/GLS1/Gln metabolism axis is an important regulator of TGF-β/Wnt-induced, Snail-dependent EMT, metastasis, and glycolytic switch.

    Topics: Adenosine Triphosphate; Apoptosis; Blotting, Western; Cell Proliferation; Chromatin Immunoprecipitation; Epithelial-Mesenchymal Transition; Fluorescent Antibody Technique; Glutaminase; Glutamine; Glycolysis; HeLa Cells; Hep G2 Cells; Homeodomain Proteins; Humans; MCF-7 Cells; Neoplasms; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta; Tumor Cells, Cultured

2016
TGF-β Signaling in Cancer.
    Journal of cellular biochemistry, 2016, Volume: 117, Issue:6

    The transforming growth factor-β (TGF-β) is a family of structurally related proteins that comprises of TGF-β, activins/inhibins, and bone morphogenic proteins (BMPs). Members of the TGF-β family control numerous cellular functions including proliferation, apoptosis, differentiation, epithelial-mesenchymal transition (EMT), and migration. The first identified member, TGF-β is implicated in several human diseases, such as vascular diseases, autoimmune disorders, and carcinogenesis. Activation of the TGF-β receptor by its ligands induces the phosphorylation of serine/threonine residues and triggers phosphorylation of the intracellular effectors, SMADs. Upon activation, SMAD proteins translocate to the nucleus and induce transcription of their target genes, regulating several cellular functions. TGF-β dysregulation has been implicated in carcinogenesis. In early stages of cancer, TGF-β exhibits tumor suppressive effects by inhibiting cell cycle progression and promoting apoptosis. However, in late stages TGF-β exerts tumor promoting effects, increasing tumor invasiveness, and metastasis. Furthermore, the TGF-β signaling pathway communicates with other signaling pathways in a synergistic or antagonistic manner and regulates cellular functions. Elevated TGF-β activity has been associated with poor clinical outcome. Given the pivotal role of TGF-β in tumor progression, this pathway is an attractive target for cancer therapy. Several therapeutic tools such as TGF-β antibodies, antisense oligonucleotides, and small molecules inhibitors of TGF-β receptor-1 (TGF-βR1) have shown immense potential to inhibit TGF-β signaling. Finally, in the interest of developing future therapies, further studies are warranted to identify novel points of convergence of TGF-β with other signaling pathways and oncogenic factors in the tumor microenvironment.

    Topics: Antineoplastic Agents; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Phosphorylation; Prognosis; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2016
TGF-β/β2-spectrin/CTCF-regulated tumor suppression in human stem cell disorder Beckwith-Wiedemann syndrome.
    The Journal of clinical investigation, 2016, Volume: 126, Issue:2

    Beckwith-Wiedemann syndrome (BWS) is a human stem cell disorder, and individuals with this disease have a substantially increased risk (~800-fold) of developing tumors. Epigenetic silencing of β2-spectrin (β2SP, encoded by SPTBN1), a SMAD adaptor for TGF-β signaling, is causally associated with BWS; however, a role of TGF-β deficiency in BWS-associated neoplastic transformation is unexplored. Here, we have reported that double-heterozygous Sptbn1+/- Smad3+/- mice, which have defective TGF-β signaling, develop multiple tumors that are phenotypically similar to those of BWS patients. Moreover, tumorigenesis-associated genes IGF2 and telomerase reverse transcriptase (TERT) were overexpressed in fibroblasts from BWS patients and TGF-β-defective mice. We further determined that chromatin insulator CCCTC-binding factor (CTCF) is TGF-β inducible and facilitates TGF-β-mediated repression of TERT transcription via interactions with β2SP and SMAD3. This regulation was abrogated in TGF-β-defective mice and BWS, resulting in TERT overexpression. Imprinting of the IGF2/H19 locus and the CDKN1C/KCNQ1 locus on chromosome 11p15.5 is mediated by CTCF, and this regulation is lost in BWS, leading to aberrant overexpression of growth-promoting genes. Therefore, we propose that loss of CTCF-dependent imprinting of tumor-promoting genes, such as IGF2 and TERT, results from a defective TGF-β pathway and is responsible at least in part for BWS-associated tumorigenesis as well as sporadic human cancers that are frequently associated with SPTBN1 and SMAD3 mutations.

    Topics: Animals; Beckwith-Wiedemann Syndrome; Carrier Proteins; CCCTC-Binding Factor; Chromosomes, Human, Pair 11; Cyclin-Dependent Kinase Inhibitor p57; Hep G2 Cells; Humans; Insulin-Like Growth Factor II; KCNQ1 Potassium Channel; Mice; Mice, Knockout; Microfilament Proteins; Neoplasm Proteins; Neoplasms; Repressor Proteins; Signal Transduction; Smad3 Protein; Telomerase; Transforming Growth Factor beta

2016
Post-Sepsis State Induces Tumor-Associated Macrophage Accumulation through CXCR4/CXCL12 and Favors Tumor Progression in Mice.
    Cancer immunology research, 2016, Volume: 4, Issue:4

    Survivors from sepsis are in an immunosuppressed state that is associated with higher long-term mortality and risk of opportunistic infections. Whether these factors contribute to neoplastic proliferation, however, remains unclear. Tumor-associated macrophages (TAM) can support malignant cell proliferation, survival, and angiogenesis. We addressed the relationship between the post-sepsis state, tumor progression and TAM accumulation, and phenotypic and genetic profile, using a mouse model of sepsis resolution and then B16 melanoma in mice. In addition, we measured the serum concentrations of TNFα, TGFβ, CCL2, and CXCL12 and determined the effect of in vivo CXCR4/CXCL12 inhibition in this context. Mice that survived sepsis showed increased tumor progression both in the short and long term, and survival times were shorter. TAM accumulation, TAM local proliferation, and serum concentrations of TGFβ, CXCL12, and TNFα were increased. Naïve mice inoculated with B16 together with macrophages from post-sepsis mice also had faster tumor progression and shorter survival. Post-sepsis TAMs had less expression of MHC-II and leukocyte activation-related genes. Inhibition of CXCR4/CXCL12 prevented the post-sepsis-induced tumor progression, TAM accumulation, and TAM in situ proliferation. Collectively, our data show that the post-sepsis state was associated with TAM accumulation through CXCR4/CXCL12, which contributed to B16 melanoma progression.

    Topics: Animals; Chemokine CXCL12; Disease Models, Animal; Disease Progression; Gene Expression; Macrophages; Male; Melanoma, Experimental; Mice; Neoplasm Metastasis; Neoplasms; Receptors, CXCR4; Sepsis; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2016
A cancer treatment based on synergy between anti-angiogenic and immune cell therapies.
    Journal of theoretical biology, 2016, Apr-07, Volume: 394

    A mathematical model integrating tumor angiogenesis and tumor-targeted cytotoxicity by immune cells was developed to identify the therapeutic window of two distinct modes to treat cancer: (1) an anti-angiogenesis treatment based on the monoclonal antibody bevacizumab that targets tumor vasculature, and (2) immunotherapy involving the injection of unlicensed dendritic cells to boost the anti-tumor adaptive response. The angiogenic cytokine Vascular Endothelial Growth Factor (VEGF) contributes to the immunosuppressive tumor microenvironment, which is responsible for the short-lived therapeutic effect of cancer-targeted immunotherapy. The effect of immunosuppression on the width of the therapeutic window of each treatment was quantified. Experimental evidence has shown that neutralizing immunosuppressive cytokines results in an enhanced immune response against infections and chronic diseases. The model was used to determine treatment protocols involving the combination of anti-VEGF and unlicensed dendritic cell injections that enhance tumor regression. The model simulations predicted that the most effective method to treat tumors involves administering a series of biweekly anti-VEGF injections to disrupt angiogenic processes and limit tumor growth. The simulations also verified the hypothesis that reducing the concentration of the immunosuppressive factor VEGF prior to an injection of unlicensed dendritic cells enhances the cytotoxicity of CD8+ T cells and results in complete tumor elimination. Feasible treatment protocols for tumors that are diagnosed late and have grown to a relatively large size were identified.

    Topics: Angiogenesis Inhibitors; Antigens, Neoplasm; Cell Count; Cell Proliferation; Computer Simulation; Dendritic Cells; Humans; Immune Evasion; Immunosuppression Therapy; Immunotherapy; Models, Biological; Neoplasms; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2016
The anti-fibrotic agent pirfenidone synergizes with cisplatin in killing tumor cells and cancer-associated fibroblasts.
    BMC cancer, 2016, Mar-02, Volume: 16

    Anti-fibrotic drugs such as pirfenidone have been developed for the treatment of idiopathic pulmonary fibrosis. Because activated fibroblasts in inflammatory conditions have similar characteristics as cancer-associated fibroblasts (CAFs) and CAFs contribute actively to the malignant phenotype, we believe that anti-fibrotic drugs have the potential to be repurposed as anti-cancer drugs.. The effects of pirfenidone alone and in combination with cisplatin on human patient-derived CAF cell lines and non-small cell lung cancer (NSCLC) cell lines were examined. The impact on cell death in vitro as well as tumor growth in a mouse model was determined. Annexin V/PI staining and Western blot analysis were used to characterize cell death. Synergy was assessed with the combination index method using Calcusyn software.. Pirfenidone alone induced apoptotic cell death in lung CAFs at a high concentration (1.5 mg/mL). However, co-culture in vitro experiments and co-implantation in vivo experiments showed that the combination of low doses of cisplatin (10 μM) and low doses of pirfenidone (0.5 mg/mL), in both CAFs and tumors, lead to increased cell death and decreased tumor progression, respectively. Furthermore, the combination of cisplatin and pirfenidone in NSCLC cells (A549 and H157 cells) leads to increased apoptosis and synergistic cell death.. Our studies reveal for the first time that the combination of cisplatin and pirfenidone is active in preclinical models of NSCLC and therefore may be a new therapeutic approach in this disease.

    Topics: Animals; Antineoplastic Agents; Apoptosis; Cell Line, Tumor; Cell Proliferation; Cisplatin; Coculture Techniques; Disease Models, Animal; Drug Synergism; Fibroblasts; Humans; Neoplasms; Pyridones; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2016
Plasticity underlies tumor progression: role of Nodal signaling.
    Cancer metastasis reviews, 2016, Volume: 35, Issue:1

    The transforming growth factor beta (TGFβ) superfamily member Nodal is an established regulator of early embryonic development, with primary roles in endoderm induction, left-right asymmetry, and primitive streak formation. Nodal signals through TGFβ family receptors at the plasma membrane and induces signaling cascades leading to diverse transcriptional regulation. While conceptually simple, the regulation of Nodal and its molecular effects are profoundly complex and context dependent. Pioneering work by developmental biologists has characterized the signaling pathways, regulatory components, and provided detailed insight into the mechanisms by which Nodal mediates changes at the cellular and organismal levels. Nodal is also an important factor in maintaining pluripotency of embryonic stem cells through regulation of core transcriptional programs. Collectively, this work has led to an appreciation for Nodal as a powerful morphogen capable of orchestrating multiple cellular phenotypes. Although Nodal is not active in most adult tissues, its reexpression and signaling have been linked to multiple types of human cancer, and Nodal has emerged as a driver of tumor growth and cellular plasticity. In vitro and in vivo experimental evidence has demonstrated that inhibition of Nodal signaling reduces cancer cell aggressive characteristics, while clinical data have established associations with Nodal expression and patient outcomes. As a result, there is great interest in the potential targeting of Nodal activity in a therapeutic setting for cancer patients that may provide new avenues for suppressing tumor growth and metastasis. In this review, we evaluate our current understanding of the complexities of Nodal function in cancer and highlight recent experimental evidence that sheds light on the therapeutic potential of its inhibition.

    Topics: Gene Expression Regulation, Neoplastic; Humans; Molecular Targeted Therapy; Neoplasms; Nodal Protein; Signal Transduction; Transforming Growth Factor beta

2016
The Risk of Cancer With the Use of Recombinant Human Bone Morphogenetic Protein in Spine Fusion.
    Spine, 2016, Aug-15, Volume: 41, Issue:16

    Retrospective cohort study using the Washington State Comprehensive Hospital Abstract Reporting System, the Washington State Cancer Registry, and Washington State death certificates.. To study the possible association between recombinant human bone morphogenetic protein (rhBMP) and cancer risk.. The use of rhBMP in spine fusion surgery remains controversial with respect to its possible role in tumorigenesis.. We compared adults who underwent spine fusion for degenerative disease with and without rhBMP between 2002 and 2010. Patients were matched on the basis of age, sex, and year of treatment. We excluded patients with a diagnosis of cancer before or at the index procedure. The primary outcome was the first diagnosis of cancer as identified in the records of the cancer registry.. We included 16,914 patients who had spine fusion, of whom 4246 received rhBMP. During the study period, 449 patients received a diagnosis of cancer: 117 (2.76% of 4246) in the rhBMP group and 332 (2.62% of 12 668) in the no rhBMP group. The incidence rate was similar between the rhBMP and no rhBMP 9.5 and 9.0 per 1000 person years, respectively (hazard ratio, 1.06; 95% confidence interval, 0.86-1.30). There were no differences in the rate of cancer between the two groups in subgroups defined on the basis of site of fusion or surgical method.. There was no increase in overall cancer incidence among those receiving rhBMP. An important limitation of this and other studies of rhBMP and cancer that have been conducted to date is their relatively limited duration of follow-up. The examination of cancer incidence following rhBMP administration must continue beyond just the first several years to adequately assess the potential of rhBMP to influence the occurrence of one or more types of malignancy.. 3.

    Topics: Adult; Aged; Aged, 80 and over; Bone Morphogenetic Protein 2; Female; Humans; Incidence; Lumbar Vertebrae; Male; Middle Aged; Neoplasms; Recombinant Proteins; Retrospective Studies; Risk; Spinal Fusion; Transforming Growth Factor beta; Young Adult

2016
Naïve CD8(+) T cell derived tumor-specific cytotoxic effectors as a potential remedy for overcoming TGF-β immunosuppression in the tumor microenvironment.
    Scientific reports, 2016, 06-16, Volume: 6

    Despite of the potential implications for cancer immunotherapy, conventional approaches using in vitro expanded CD8(+) T cells have suboptimal outcomes, mostly due to loss of functionality from cellular exhaustion. We therefore investigated the phenotypic and functional differences among in vitro activated CD8(+) T cells of three different sources, namely naïve (NTeff), memory (MTeff) and tumor-infiltrating lymphocytes (TILeff) from human and mice, to better understand mechanisms behind potent effector functions and potential for overcoming current limitations. In line with the greater proliferation activity and longer telomere lengths of NTeff populations, cells of naïve origin exhibited significantly less amounts of T cell exhaustion markers than those of MTeff and TILeff, and moreover, acquired distinct expression patterns of memory-promoting transcription factors, T-bet and Eomes, induced in a rapid and sustainable manner. NTeff cells appeared to have lower expression of Foxp1 and were refractory to apoptosis upon TGF-β conditioning, implying better survival potential and resistance to tumor-induced immune suppression. Of CD8(+) T cell pools activated to tumor-specific CTLs, naïve cell generated effectors possessed the most potent cytotoxic activity, validating implications for use in rational design of adoptive immunotherapy.

    Topics: Animals; Apoptosis; Cell Line; Forkhead Transcription Factors; Humans; Immunologic Memory; Immunotherapy, Adoptive; Lymphocyte Activation; Lymphocytes, Tumor-Infiltrating; Mice; Neoplasms; Repressor Proteins; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Tumor Microenvironment

2016
Bone Morphogenetic Proteins in Lumbar Arthrodesis: What Is All the Debate About? Commentary on an article by Daniel C. Beachler, PhD, MHS, et al.: "Bone Morphogenetic Protein Use and Cancer Risk Among Patients Undergoing Lumbar Arthrodesis: A Case-Cohort
    The Journal of bone and joint surgery. American volume, 2016, 07-06, Volume: 98, Issue:13

    Topics: Arthrodesis; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Cohort Studies; Humans; Lumbar Vertebrae; Medicare; Neoplasms; Recombinant Proteins; Spinal Fusion; Transforming Growth Factor beta

2016
c-Src Suppresses Dendritic Cell Antitumor Activity via T Cell Ig and Mucin Protein-3 Receptor.
    Journal of immunology (Baltimore, Md. : 1950), 2016, 09-01, Volume: 197, Issue:5

    The enhanced expression of T cell Ig and mucin protein-3 (TIM-3) on tumor-associated dendritic cells (DCs) attenuates antitumor effects of DNA vaccines. To identify a potential target (or targets) for reducing TIM-3 expression on tumor-associated DCs, we explored the molecular mechanisms regulating TIM-3 expression. In this study, we have identified a novel signaling pathway (c-Src→Bruton's tyrosine kinase→transcription factors Ets1, Ets2, USF1, and USF2) necessary for TIM-3 upregulation on DCs. Both IL-10 and TGF-β, which are produced in the tumor microenvironment, upregulated TIM-3 expression on DCs via this pathway. Suppressed expression of c-Src or downstream Bruton's tyrosine kinase, Ets1, Ets2, USF1, or USF2 blocked IL-10- and TGF-β-induced TIM-3 upregulation on DCs. Notably, in vivo knockdown of c-Src in mice reduced TIM-3 expression on tumor-associated DCs. Furthermore, adoptive transfer of c-Src-silenced DCs in mouse tumors enhanced the in vivo antitumor effects of immunostimulatory CpG DNA; however, TIM-3 overexpression in c-Src-silenced DCs blocked this effect. Collectively, our data reveal the molecular mechanism regulating TIM-3 expression in DCs and identify c-Src as a target for improving the efficacy of nucleic acid-mediated anticancer therapy.

    Topics: Adoptive Transfer; Agammaglobulinaemia Tyrosine Kinase; Animals; Cell Differentiation; CSK Tyrosine-Protein Kinase; Dendritic Cells; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Genes, src; Hepatitis A Virus Cellular Receptor 2; Interleukin-10; Mice; Neoplasms; Oligodeoxyribonucleotides; Protein-Tyrosine Kinases; Signal Transduction; src-Family Kinases; T-Lymphocytes; Transforming Growth Factor beta; Up-Regulation

2016
Stimulation of Natural Killer Cell-Mediated Tumor Immunity by an IL15/TGFβ-Neutralizing Fusion Protein.
    Cancer research, 2016, 10-01, Volume: 76, Issue:19

    The clinical efficacy of immune cytokines used for cancer therapy is hampered by elements of the immunosuppressive tumor microenvironment such as TGFβ. Here we demonstrate that FIST15, a recombinant chimeric protein composed of the T-cell-stimulatory cytokine IL15, the sushi domain of IL15Rα and a TGFβ ligand trap, can overcome immunosuppressive TGFβ to effectively stimulate the proliferation and activation of natural killer (NK) and CD8

    Topics: Animals; CD8-Positive T-Lymphocytes; Cell Line, Tumor; Humans; Interleukin-15; Killer Cells, Natural; Lymphocyte Activation; Mice; Mice, Inbred C57BL; Neoplasms; Recombinant Fusion Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2016
Nanoroughened adhesion-based capture of circulating tumor cells with heterogeneous expression and metastatic characteristics.
    BMC cancer, 2016, 08-08, Volume: 16

    Circulating tumor cells (CTCs) have shown prognostic relevance in many cancer types. However, the majority of current CTC capture methods rely on positive selection techniques that require a priori knowledge about the surface protein expression of disseminated CTCs, which are known to be a dynamic population.. We developed a microfluidic CTC capture chip that incorporated a nanoroughened glass substrate for capturing CTCs from blood samples. Our CTC capture chip utilized the differential adhesion preference of cancer cells to nanoroughened etched glass surfaces as compared to normal blood cells and thus did not depend on the physical size or surface protein expression of CTCs.. The microfluidic CTC capture chip was able to achieve a superior capture yield for both epithelial cell adhesion molecule positive (EpCAM+) and EpCAM- cancer cells in blood samples. Additionally, the microfluidic CTC chip captured CTCs undergoing transforming growth factor beta-induced epithelial-to-mesenchymal transition (TGF-β-induced EMT) with dynamically down-regulated EpCAM expression. In a mouse model of human breast cancer using EpCAM positive and negative cell lines, the number of CTCs captured correlated positively with the size of the primary tumor and was independent of their EpCAM expression. Furthermore, in a syngeneic mouse model of lung cancer using cell lines with differential metastasis capability, CTCs were captured from all mice with detectable primary tumors independent of the cell lines' metastatic ability.. The microfluidic CTC capture chip using a novel nanoroughened glass substrate is broadly applicable to capturing heterogeneous CTC populations of clinical interest independent of their surface marker expression and metastatic propensity. We were able to capture CTCs from a non-metastatic lung cancer model, demonstrating the potential of the chip to collect the entirety of CTC populations including subgroups of distinct biological and phenotypical properties. Further exploration of the biological potential of metastatic and presumably non-metastatic CTCs captured using the microfluidic chip will yield insights into their relevant differences and their effects on tumor progression and cancer outcomes.

    Topics: A549 Cells; Animals; Cell Adhesion; Cell Line, Tumor; Cell Separation; Epithelial Cell Adhesion Molecule; Epithelial-Mesenchymal Transition; Female; Genetic Heterogeneity; Humans; MCF-7 Cells; Mice; Microfluidic Analytical Techniques; Neoplasm Metastasis; Neoplasm Transplantation; Neoplasms; Neoplastic Cells, Circulating; Transforming Growth Factor beta

2016
[Role of HIF-induced EMT in invasion and
metastasis of tumor].
    Zhong nan da xue xue bao. Yi xue ban = Journal of Central South University. Medical sciences, 2016, Volume: 41, Issue:8

    Hypoxia plays a vital role in tumor metabolism, proliferation, apoptosis, invasion and metastasis via hypoxia-inducible factor (HIF). Epithelial to mesenchymal transition (EMT) is a crucial process to metastasis, which could be triggered by hypoxia. EMT could be regulated by HIF via multiple pathways including TGF-β, Notch, and Wnt/β-catenin. It has been shown that anti-HIF drugs combined with anti-EMT therapies could be a promising strategy for tumor therapy.. 缺氧是肿瘤微环境的主要特点,通过缺氧诱导因子(hypoxia-inducible factor,HIF)激活众多靶基因,从而影响肿瘤的代谢、增殖、凋亡、侵袭转移、耐药等多个方面。上皮细胞间质化(epithelial to mesenchymal transition,EMT)是肿瘤发生侵袭转移的重要起始过程,缺氧亦是EMT的诱发因素之一并可通过多条信号转导通路包括TGF-β,Notch,Wnt/β-catenin等调节EMT。研究缺氧诱导EMT的机制并阻断其进程,可成为治疗肿瘤的新靶点。.

    Topics: Basic Helix-Loop-Helix Transcription Factors; beta Catenin; Epithelial-Mesenchymal Transition; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Transforming Growth Factor beta

2016
GADD34 Promotes Tumor Growth by Inducing Myeloid-derived Suppressor Cells.
    Anticancer research, 2016, Volume: 36, Issue:9

    Tumor hypoxia induces the expression of growth arrest and DNA damage-inducible protein (GADD34). However, the role of GADD34 in tumor growth remains unclear.. Gadd34 expression was knocked-down through lentivirus-mediated short hairpin RNA (shRNA) in tumor cells, which were subsequently injected subcutaneously into mice. Tumor volumes and myeloid-derived suppressor cells (MDSCs) were monitored. Isolated MDSCs were incubated with tumor supernatant to investigate the impact of GADD34 on cytokine secretion of MDSCs.. We observed that reduction of GADD34 expression significantly suppressed tumor, and resulted in decreased accumulation of MDSCs and T-cells, and inhibition of GADD34 reduced secretion of vascular epithelial growth factor α and transforming growth factor β by MDSCs.. These findings provide a promising strategy for targeting GADD34 activity in order to inhibit tumor growth.

    Topics: Animals; Breast Neoplasms; Carcinoma, Lewis Lung; CD11b Antigen; Culture Media, Conditioned; DNA Damage; Female; Flow Cytometry; Humans; Hypoxia; Mice; Mice, Inbred C57BL; Mice, Nude; Myeloid Cells; Myeloid-Derived Suppressor Cells; Neoplasms; Protein Phosphatase 1; Real-Time Polymerase Chain Reaction; RNA, Small Interfering; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2016
PMEPA1, a TGF-β- and hypoxia-inducible gene that participates in hypoxic gene expression networks in solid tumors.
    Biochemical and biophysical research communications, 2016, Oct-28, Volume: 479, Issue:4

    Prostate transmembrane protein, androgen induced 1 (PMEPA1) is highly expressed in various solid tumors and is known to play important roles in the transforming growth factor-β (TGF-β) signaling pathway. Here, we demonstrate a novel relationship between PMEPA1 and hypoxia, a common microenvironmental stress condition in solid tumors. We showed that induction of PMEPA1 expression occurred during hypoxia in a manner dependent on both TGF-β signaling and hypoxia-inducible factor-1 (HIF-1) pathways. Furthermore, overexpression and knockdown experiments revealed that PMEPA1 enhanced HIF-1 transcription activity. Bioinformatics analyses of PMEPA1-correlated genes using a gene expression database in clinical settings showed significant enrichment of gene sets defined by TGF-β and hypoxia and these two signaling pathways-related angiogenesis and epithelial-mesenchymal transition in many types of solid tumors. Collectively, our findings indicated that PMEPA1 participates in TGF-β- and hypoxia-regulated gene expression networks in solid tumors and thereby may contribute to tumor progression.

    Topics: Cell Line, Tumor; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Gene Regulatory Networks; Humans; Intracellular Signaling Peptides and Proteins; Membrane Proteins; Mitochondrial Proteins; Neoplasm Proteins; Neoplasms; Transcription, Genetic; Transforming Growth Factor beta; Tumor Hypoxia

2016
TGF-
    JCI insight, 2016, 06-16, Volume: 1, Issue:9

    Tregs imprint an early immunotolerant tumor environment that prevents effective antitumor immune responses. Using transcriptomics of tumor tissues, we identified early upregulation of VEGF and TGF-β pathways compatible with tolerance imprinting. Silencing of VEGF or TGF-β in tumor cells induced early and pleiotropic modulation of immune-related transcriptome signatures in tumor tissues. These were surprisingly similar for both silenced tumors and related to common downstream effects on Tregs. Silencing of VEGF or TGF-β resulted in dramatically delayed tumor growth, associated with decreased Tregs and myeloid-derived suppressor cells and increased effector T cell activation in tumor infiltrates. Strikingly, co-silencing of TGF-β and VEGF led to a substantial spontaneous tumor eradication rate and the combination of their respective inhibitory drugs was synergistic. VEGF and/or TGF-β silencing also restored tumor sensitivity to tumor-specific cell therapies and markedly improved the efficacy of anti-PD-1/anti-CTLA-4 treatment. Thus, TGF-β and VEGF cooperatively control the tolerant environment of tumors and are targets for improved cancer immunotherapies.

    Topics: Animals; Cell Line, Tumor; Female; Gene Silencing; Immune Tolerance; Immunotherapy; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2016
NKp44 and NKp30 splice variant profiles in decidua and tumor tissues: a comparative viewpoint.
    Oncotarget, 2016, Oct-25, Volume: 7, Issue:43

    NKp44 and NKp30 splice variant profiles have been shown to promote diverse cellular functions. Moreover, microenvironment factors such as TGF-β, IL-15 and IL-18 are able to influence both NKp44 and NKp30 splice variant profiles, leading to cytokine-associated profiles. Placenta and cancerous tissues have many similarities; both are immunologically privileged sites and both share immune tolerance mechanisms to support tissue development. Therefore, we studied the profiles of NKp44 and NKp30 splice variants in these states by comparing (i) decidua from pregnancy disorder and healthy gestation and (ii) matched normal and cancer tissue. Decidua samples had high incidence of both NKp44 and NKp30. In cancerous state it was different; while NKp30 expression was evident in most cancerous and matched normal tissues, NKp44 incidence was lower and was mostly associated with the cancerous tissues. A NKp44-1dominant inhibitory profile predominated in healthy pregnancy gestation. Interestingly, the NKp44-2/3 activation profile becomes the leading profile in spontaneous abortions, whereas balanced NKp44 profiles were observed in preeclampsia. In contrast, a clear preference for the NKp30a/b profile was evident in the 1st trimester decidua, yet no significant differences were observed for NKp30 profiles between healthy gestation and spontaneous abortions/preeclampsia. Both cancerous and matched normal tissues manifested balanced NKp30c inhibitory and NKp30a/b activation profiles with a NKp44-1dominant profile. However, a shift in NKp30 profiles between matched normal and cancer tissue was observed in half of the cases. To summarize, NKp44 and NKp30 splice variants profiles are tissue/condition specific and demonstrate similarity between placenta and cancerous tissues.

    Topics: Abortion, Spontaneous; Decidua; Female; Flow Cytometry; Humans; Immune Privilege; Interleukin-15; Interleukin-18; Killer Cells, Natural; Lymphocytes, Tumor-Infiltrating; Natural Cytotoxicity Triggering Receptor 2; Natural Cytotoxicity Triggering Receptor 3; Neoplasms; Pre-Eclampsia; Pregnancy; RNA Splicing; Transforming Growth Factor beta; Tumor Microenvironment

2016
Evaluation of complications associated with off-label use of recombinant human bone morphogenetic protein-2 (rhBMP-2) in pediatric orthopaedics.
    Journal of materials science. Materials in medicine, 2016, Volume: 27, Issue:12

    The off-label use of recombinant human bone morphogenetic protein-2 to promote bone healing in adults has significantly increased in recent years, while reports of recombinant human bone morphogenetic protein-2 application in children and adolescents are very rare. The aim of this study was to evaluate the safety of single and repetitive recombinant human bone morphogenetic protein-2 use in pediatric orthoapedics. Therefore we reviewed the medical records of 39 patients who had been treated with recombinant human bone morphogenetic protein-2 at our institution. Their mean age was 10.9 years. Recombinant human bone morphogenetic protein-2 was used in 17 patients for spine fusion, in 11 patients for the treatment of congenital pseudarthrosis of the tibia or tibial nonunion, in 5 patients for the management of femoral nonunion, in 5 patients for nonunions at other locations, and in 1 case for tibial shortening. Special attention was paid to identify all adverse events that may be attributed to recombinant human bone morphogenetic protein-2 use, including local inflammatory reactions, allergic reactions, systemic toxicity, excessive wound swelling, hematoma, compartment syndrome, infection, heterotopic ossification, excessive bone growth, carcinogenicity, and the consequences of repeated applications of recombinant human bone morphogenetic protein-2. Follow-up was a mean of 39 months. Forty-six operations with application of rhBMP-2 were performed. Complications that may be due to application of recombinant human bone morphogenetic protein-2 were seen after 18 operations including swelling, increase in temperature, wound secretion, redness and hyperthermia. We consider the three cases of necessary revisions, one due to hematoma, one due to development of a compartment syndrome, and one due to deep infection, to be the only complications related to the use of recombinant human bone morphogenetic protein-2. In conclusion, we found few complications attributable to application of recombinant human bone morphogenetic protein-2 in pediatric patients.

    Topics: Adolescent; Bone Morphogenetic Protein 2; Child; Cohort Studies; Collagen; Female; Humans; Intraoperative Complications; Lumbar Vertebrae; Male; Neoplasms; Off-Label Use; Orthopedics; Pediatrics; Recombinant Proteins; Reoperation; Transforming Growth Factor beta

2016
Intestinal helminths regulate lethal acute graft-versus-host disease and preserve the graft-versus-tumor effect in mice.
    Journal of immunology (Baltimore, Md. : 1950), 2015, Feb-01, Volume: 194, Issue:3

    Donor T lymphocyte transfer with hematopoietic stem cells suppresses residual tumor growth (graft-versus-tumor [GVT]) in cancer patients undergoing bone marrow transplantation (BMT). However, donor T cell reactivity to host organs causes severe and potentially lethal inflammation called graft-versus-host disease (GVHD). High-dose steroids or other immunosuppressive drugs are used to treat GVHD that have limited ability to control the inflammation while incurring long-term toxicity. Novel strategies are needed to modulate GVHD, preserve GVT, and improve the outcome of BMT. Regulatory T cells (Tregs) control alloantigen-sensitized inflammation of GVHD, sustain GVT, and prevent mortality in BMT. Helminths colonizing the alimentary tract dramatically increase the Treg activity, thereby modulating intestinal or systemic inflammatory responses. These observations led us to hypothesize that helminths can regulate GVHD and maintain GVT in mice. Acute GVHD was induced in helminth (Heligmosomoides polygyrus)-infected or uninfected BALB/c recipients of C57BL/6 donor grafts. Helminth infection suppressed donor T cell inflammatory cytokine generation and reduced GVHD-related mortality, but maintained GVT. H. polygyrus colonization promoted the survival of TGF-β-generating recipient Tregs after a conditioning regimen with total body irradiation and led to a TGF-β-dependent in vivo expansion/maturation of donor Tregs after BMT. Helminths did not control GVHD when T cells unresponsive to TGF-β-mediated immune regulation were used as donor T lymphocytes. These results suggest that helminths suppress acute GVHD using Tregs and TGF-β-dependent pathways in mice. Helminthic regulation of GVHD and GVT through intestinal immune conditioning may improve the outcome of BMT.

    Topics: Acute Disease; Adoptive Transfer; Animals; Bone Marrow Transplantation; Cytokines; Disease Models, Animal; Graft vs Host Disease; Helminthiasis, Animal; Helminths; Immunomodulation; Immunophenotyping; Intestines; Male; Mice; Neoplasms; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Transplantation Conditioning; Transplantation, Homologous

2015
Cutting edge: epigenetic regulation of Foxp3 defines a stable population of CD4+ regulatory T cells in tumors from mice and humans.
    Journal of immunology (Baltimore, Md. : 1950), 2015, Feb-01, Volume: 194, Issue:3

    CD4(+) regulatory T cells (Tregs) are critical for maintaining self-tolerance and function to prevent autoimmune disease. High densities of intratumoral Tregs are generally associated with poor patient prognosis, a correlation attributed to their broad immune-suppressive features. Two major populations of Tregs have been defined, thymically derived natural Tregs (nTregs) and peripherally induced Tregs (iTregs). However, the relative contribution of nTregs versus iTregs to the intratumoral Treg compartment remains controversial. Demarcating the proportion of nTregs versus iTregs has important implications in the design of therapeutic strategies to overcome their antagonistic effects on antitumor immune responses. We used epigenetic, phenotypic, and functional parameters to evaluate the composition of nTregs versus iTregs isolated from mouse tumor models and primary human tumors. Our findings failed to find evidence for extensive intratumoral iTreg induction. Rather, we identified a population of Foxp3-stable nTregs in tumors from mice and humans.

    Topics: Animals; Antigens, Surface; Cell Line, Tumor; CpG Islands; Disease Models, Animal; DNA Methylation; Epigenesis, Genetic; Forkhead Transcription Factors; Gene Expression Regulation, Neoplastic; Humans; Immunophenotyping; Lymphocytes, Tumor-Infiltrating; Mice; Mice, Transgenic; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2015
Genetic variants modulating CRIPTO serum levels identified by genome-wide association study in Cilento isolates.
    PLoS genetics, 2015, Volume: 11, Issue:1

    Cripto, the founding member of the EGF-CFC genes, plays an essential role in embryo development and is involved in cancer progression. Cripto is a GPI-anchored protein that can interact with various components of multiple signaling pathways, such as TGF-β, Wnt and MAPK, driving different processes, among them epithelial-mesenchymal transition, cell proliferation, and stem cell renewal. Cripto protein can also be cleaved and released outside the cell in a soluble and still active form. Cripto is not significantly expressed in adult somatic tissues and its re-expression has been observed associated to pathological conditions, mainly cancer. Accordingly, CRIPTO has been detected at very low levels in the plasma of healthy volunteers, whereas its levels are significantly higher in patients with breast, colon or glioblastoma tumors. These data suggest that CRIPTO levels in human plasma or serum may have clinical significance. However, very little is known about the variability of serum levels of CRIPTO at a population level and the genetic contribution underlying this variability remains unknown. Here, we report the first genome-wide association study of CRIPTO serum levels in isolated populations (n = 1,054) from Cilento area in South Italy. The most associated SNPs (p-value<5*10-8) were all located on chromosome 3p22.1-3p21.3, in the CRIPTO gene region. Overall six CRIPTO associated loci were replicated in an independent sample (n = 535). Pathway analysis identified a main network including two other genes, besides CRIPTO, in the associated regions, involved in cell movement and proliferation. The replicated loci explain more than 87% of the CRIPTO variance, with 85% explained by the most associated SNP. Moreover, the functional analysis of the main associated locus identified a causal variant in the 5'UTR of CRIPTO gene which is able to strongly modulate CRIPTO expression through an AP-1-mediate transcriptional regulation.

    Topics: Adult; Aged; Cell Movement; Cell Proliferation; Embryonic Development; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation; Genome-Wide Association Study; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; Italy; Middle Aged; Neoplasm Proteins; Neoplasms; Transcription Factor AP-1; Transforming Growth Factor beta

2015
Dendritic cell regulation of NK-cell responses involves lymphotoxin-α, IL-12, and TGF-β.
    European journal of immunology, 2015, Volume: 45, Issue:6

    Dendritic cell (DC) vaccines induce T-cell responses in cancer patients. However, there is a paucity of data regarding the role of DC vaccines in shaping natural killer (NK) cell responses. Here, we observe that NK cells are less activated following DC vaccination. In vitro, DC-mediated inhibition of NK cells did not require cell-to-cell contact, but required increased Signal transducer and activator of transcription 3 (STAT3) phosphorylation (pSTAT3) in DCs. When phosphorylation of STAT3 was inhibited in DCs, we found that DCs did not suppress NK cells, and observed an increase in the production of lymphotoxin-alpha (LTα) and interleukin-12 (IL-12) as well as reduced release of transforming growth factor beta (TGF-β). The addition of recombinant LTα or IL-12 to the DC-NK-cell cocultures restored NK-cell activity, and neutralization of TGF-β resulted in elevated production of LTα and IL-12 from DCs. Compared with LPS, DCs matured with a cocktail of R848, poly I:C, and IFN-γ showed reduced levels of pSTAT3 and higher levels of LTα and IL-12 and did not inhibit NK-cell activity. These results show that LTα, IL-12, and TGF-β are involved in the cross-talk between NK cells and DCs. Our findings have important implications for the development of DC-based vaccination strategies to potentiate NK-cell responses in patients with cancer.

    Topics: Autocrine Communication; Cancer Vaccines; Cell Communication; Dendritic Cells; Humans; Immunomodulation; Immunotherapy; Interferon-gamma; Interleukin-12; Interleukin-2; Killer Cells, Natural; Lymphocyte Activation; Lymphotoxin-alpha; Neoplasms; Phenotype; Phosphorylation; STAT3 Transcription Factor; Transforming Growth Factor beta

2015
Editorial: Food and Drug Administration Safety Communication on rhBMP-2 use.
    Journal of neurosurgery. Pediatrics, 2015, Volume: 16, Issue:1

    Topics: Bone Morphogenetic Protein 2; Female; Humans; Male; Neoplasms; Spinal Fusion; Transforming Growth Factor beta

2015
Routine use of recombinant human bone morphogenetic protein-2 in posterior fusions of the pediatric spine and incidence of cancer.
    Journal of neurosurgery. Pediatrics, 2015, Volume: 16, Issue:1

    OBJECT The aim of this study was to determine the safety of recombinant human bone morphogenetic protein-2 (rhBMP-2) use in posterior instrumented fusions in the pediatric population, focusing on cancer risk. In a previous study, the authors reported the short-term (mean follow-up of 11 months) safety and efficacy of rhBMP-2 in the pediatric age group. The present study reports their results with a minimum of 24 months' follow-up. METHODS The authors retrospectively reviewed 57 consecutive cases involving pediatric patients who underwent posterior occiptocervical, cervical, thoracic, lumbar, or lumbosacral spine fusion from October 1, 2007, to June 30, 2011, at Texas Children's Hospital. Seven cases were excluded from further analysis because of loss to follow-up. Three patients died during the follow-up period and were placed in a separate cohort. RESULTS The patients' average age at the time of surgery was 11 years, 4 months (range 9 months to 20 years). The mean duration of follow-up was 48.4 months (range 24-70 months). Cancer status was determined at the most recent encounter with the patient and/or caretaker(s) in person, or in telephone follow-up. Twenty-four or more months after administration of rhBMP-2, there were no cases of new malignancy, degeneration, or metastasis of existing tumors. The cause of death of the patients who died during the study period was not related to BMP or to the development, degeneration, or metastasis of cancer. CONCLUSIONS Despite the large number of adult studies reporting increased cancer risk associated with BMP use, the authors' outcomes with rhBMP-2 in the pediatric population suggest that it is a safe adjunct to posterior spine fusions of the occipitocervical, cervical, thoracic, lumbar, and lumbosacral spine. There were no new cases of cancer, or degeneration or metastasis of existing malignancies in this series.

    Topics: Adolescent; Adult; Atlanto-Occipital Joint; Bone Morphogenetic Protein 2; Cervical Vertebrae; Child; Child, Preschool; Female; Follow-Up Studies; Humans; Incidence; Infant; Lumbar Vertebrae; Male; Neoplasms; Recombinant Proteins; Retrospective Studies; Risk Assessment; Risk Factors; Sacrum; Spinal Fusion; Texas; Thoracic Vertebrae; Transforming Growth Factor beta; Young Adult

2015
Cancer: Antibodies regulate antitumour immunity.
    Nature, 2015, May-07, Volume: 521, Issue:7550

    Topics: Animals; Antibodies, Neoplasm; B-Lymphocytes; Humans; Immunoglobulin A; Immunoglobulin G; Models, Immunological; Neoplasm Transplantation; Neoplasms; Organoplatinum Compounds; Oxaliplatin; T-Lymphocytes; Transforming Growth Factor beta

2015
SPSB1, a Novel Negative Regulator of the Transforming Growth Factor-β Signaling Pathway Targeting the Type II Receptor.
    The Journal of biological chemistry, 2015, Jul-17, Volume: 290, Issue:29

    Appropriate cellular signaling is essential to control cell proliferation, differentiation, and cell death. Aberrant signaling can have devastating consequences and lead to disease states, including cancer. The transforming growth factor-β (TGF-β) signaling pathway is a prominent signaling pathway that has been tightly regulated in normal cells, whereas its deregulation strongly correlates with the progression of human cancers. The regulation of the TGF-β signaling pathway involves a variety of physiological regulators. Many of these molecules act to alter the activity of Smad proteins. In contrast, the number of molecules known to affect the TGF-β signaling pathway at the receptor level is relatively low, and there are no known direct modulators for the TGF-β type II receptor (TβRII). Here we identify SPSB1 (a Spry domain-containing Socs box protein) as a novel regulator of the TGF-β signaling pathway. SPSB1 negatively regulates the TGF-β signaling pathway through its interaction with both endogenous and overexpressed TβRII (and not TβRI) via its Spry domain. As such, TβRII and SPSB1 co-localize on the cell membrane. SPSB1 maintains TβRII at a low level by enhancing the ubiquitination levels and degradation rates of TβRII through its Socs box. More importantly, silencing SPSB1 by siRNA results in enhanced TGF-β signaling and migration and invasion of tumor cells.

    Topics: Animals; Cell Line; Cell Line, Tumor; Cell Movement; Gene Silencing; HEK293 Cells; Humans; Mice; Neoplasm Invasiveness; Neoplasms; NIH 3T3 Cells; Protein Interaction Maps; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Suppressor of Cytokine Signaling Proteins; Transcriptional Activation; Transforming Growth Factor beta; Ubiquitination

2015
Design and Selection of Antisense Oligonucleotides Targeting Transforming Growth Factor Beta (TGF-β) Isoform mRNAs for the Treatment of Solid Tumors.
    Methods in molecular biology (Clifton, N.J.), 2015, Volume: 1317

    Transforming growth factor beta isoforms (TGF-β1, -β2, and -β3) are cytokines associated with a wide range of biological processes in oncology including tumor cell invasion and migration, angiogenesis, immunosuppression, as well as regulation of tumor stem cell properties. Hence, blocking the TGF-β signaling pathways may have a multifold therapeutic benefit for the treatment of solid tumors. Here, we describe the identification and selection processes for the development of highly potent and selective chemically modified antisense oligodeoxynucleotides (fully phosphorothioate locked nucleic acid gapmers) allowing effective and selective suppression of TGF-β isoform expression in cell-based assays and in vivo preclinical models.

    Topics: Alanine Transaminase; Animals; Base Sequence; Cell Line, Tumor; DNA; Down-Regulation; Enzyme-Linked Immunosorbent Assay; Genetic Therapy; Humans; Liver; Mice; Neoplasms; Oligonucleotides, Antisense; Protein Isoforms; RNA, Messenger; Transforming Growth Factor beta

2015
Inhibition of FOXP3/NFAT Interaction Enhances T Cell Function after TCR Stimulation.
    Journal of immunology (Baltimore, Md. : 1950), 2015, Oct-01, Volume: 195, Issue:7

    Regulatory T cell (Treg) activity is modulated by a cooperative complex between the transcription factor NFAT and FOXP3, a lineage specification factor for Tregs. FOXP3/NFAT interaction is required to repress expression of IL-2, upregulate expression of the Treg markers CTLA4 and CD25, and confer suppressor function to Tregs. However, FOXP3 is expressed transiently in conventional CD4(+) T cells upon TCR stimulation and may lead to T cell hyporesponsiveness. We found that a short synthetic peptide able to inhibit FOXP3/NFAT interaction impaired suppressor activity of conventional Tregs in vitro. Specific inhibition of FOXP3/NFAT interaction with this inhibitory peptide revealed that FOXP3 downregulates NFAT-driven promoter activity of CD40L and IL-17. Inhibition of FOXP3/NFAT interaction upregulated CD40L expression on effector T cells and enhanced T cell proliferation and IL-2, IFN-γ, IL-6, or IL-17 production in response to TCR stimulation. The inhibitory peptide impaired effector T cell conversion into induced Tregs in the presence of TGF-β. Moreover, in vivo peptide administration showed antitumor efficacy in mice bearing Hepa129 or TC1 tumor cells when combined with sorafenib or with an antitumor vaccine, respectively. Our results suggest that inhibition of NFAT/FOXP3 interaction might improve antitumor immunotherapies.

    Topics: Animals; Antineoplastic Agents; CD40 Ligand; Cell Proliferation; CTLA-4 Antigen; Female; Forkhead Transcription Factors; Humans; Immunotherapy; Interferon-gamma; Interleukin-17; Interleukin-2; Interleukin-2 Receptor alpha Subunit; Interleukin-6; Jurkat Cells; Lymphocyte Activation; Male; Mice; Mice, Inbred BALB C; Mice, Inbred C3H; Mice, Inbred C57BL; Mice, Transgenic; Neoplasms; NFATC Transcription Factors; Niacinamide; Ovalbumin; Peptide Fragments; Phenylurea Compounds; Promoter Regions, Genetic; Receptors, Antigen, T-Cell; Sorafenib; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2015
Excess TGF-β mediates muscle weakness associated with bone metastases in mice.
    Nature medicine, 2015, Volume: 21, Issue:11

    Cancer-associated muscle weakness is a poorly understood phenomenon, and there is no effective treatment. Here we find that seven different mouse models of human osteolytic bone metastases-representing breast, lung and prostate cancers, as well as multiple myeloma-exhibited impaired muscle function, implicating a role for the tumor-bone microenvironment in cancer-associated muscle weakness. We found that transforming growth factor (TGF)-β, released from the bone surface as a result of metastasis-induced bone destruction, upregulated NADPH oxidase 4 (Nox4), resulting in elevated oxidization of skeletal muscle proteins, including the ryanodine receptor and calcium (Ca(2+)) release channel (RyR1). The oxidized RyR1 channels leaked Ca(2+), resulting in lower intracellular signaling, which is required for proper muscle contraction. We found that inhibiting RyR1 leakage, TGF-β signaling, TGF-β release from bone or Nox4 activity improved muscle function in mice with MDA-MB-231 bone metastases. Humans with breast- or lung cancer-associated bone metastases also had oxidized skeletal muscle RyR1 that is not seen in normal muscle. Similarly, skeletal muscle weakness, increased Nox4 binding to RyR1 and oxidation of RyR1 were present in a mouse model of Camurati-Engelmann disease, a nonmalignant metabolic bone disorder associated with increased TGF-β activity. Thus, pathological TGF-β release from bone contributes to muscle weakness by decreasing Ca(2+)-induced muscle force production.

    Topics: Absorptiometry, Photon; Animals; Bone Neoplasms; Breast Neoplasms; Calcium; Calcium Signaling; Camurati-Engelmann Syndrome; Cell Line, Tumor; Disease Models, Animal; Female; Humans; Lung Neoplasms; Male; MCF-7 Cells; Mice; Mice, Nude; Mice, SCID; Multiple Myeloma; Muscle Contraction; Muscle Proteins; Muscle Strength; Muscle Weakness; Muscle, Skeletal; NADPH Oxidase 4; NADPH Oxidases; Neoplasms; Osteolysis; Oxidation-Reduction; Prostatic Neoplasms; Ryanodine Receptor Calcium Release Channel; Transforming Growth Factor beta; Up-Regulation; X-Ray Microtomography

2015
Impact of Diverse Immune Evasion Mechanisms of Cancer Cells on T Cells Engaged by EpCAM/CD3-Bispecific Antibody Construct AMG 110.
    PloS one, 2015, Volume: 10, Issue:10

    Bispecific T cell engager (BiTE®) are single-chain bispecific antibody constructs with dual specificity for CD3 on T cells and a surface antigen on target cells. They can elicit a polyclonal cytotoxic T cell response that is not restricted by T cell receptor (TCR) specificity, and surface expression of MHC class I/peptide antigen complexes. Using human EpCAM/CD3-bispecific BiTE® antibody construct AMG 110, we here assessed to what extent surface expression of PD-L1, cytoplasmic expression of indoleamine-2,3-deoxygenase type 1, Bcl-2 and serpin PI-9, and the presence of transforming growth factor beta (TGF-β), interleukin-10 (IL-10) and adenosine in culture medium can impact redirected lysis by AMG 110-engaged T cells.. The seven factors, which are all involved in inhibiting T cell functions by cancer cells, were tested with human EpCAM-expressing Chinese hamster ovary (CHO) target cells at levels that in most cases exceeded those observed in a number of human cancer cell lines. Co-culture experiments were used to determine the impact of the evasion mechanisms on EC50 values and amplitude of redirected lysis by AMG 110, and on BiTE®-induced proliferation of previously resting human peripheral T cells.. An inhibitory effect on redirected lysis by AMG 110-engaged T cells was seen upon overexpression of serpin PI-9, Bcl-2, TGF-β and PD-L1. An inhibitory effect on induction of T cell proliferation was only seen with CHO cells overexpressing IDO. In no case, a single evasion mechanism rendered target cells completely resistant to BiTE®-induced lysis, and even various combinations could not.. Our data suggest that diverse mechanisms employed by cancer cells to fend off T cells cannot inactivate AMG 110-engaged T cells, and that inhibitory effects observed in vitro may be overcome by increased concentrations of the BiTE® antibody construct.

    Topics: Animals; Antibodies, Bispecific; Antigens, Neoplasm; B7-H1 Antigen; CD3 Complex; Cell Adhesion Molecules; Cell Line, Tumor; CHO Cells; Cricetulus; Cytotoxicity, Immunologic; Epithelial Cell Adhesion Molecule; Gene Expression; Genetic Vectors; Humans; Immune Evasion; Lymphocyte Activation; Neoplasms; Proto-Oncogene Proteins c-bcl-2; Serpins; T-Lymphocytes; Transfection; Transforming Growth Factor beta

2015
A TGF-β pathway associated with cancer cachexia.
    Nature medicine, 2015, Volume: 21, Issue:11

    Topics: Animals; Bone Neoplasms; Bone Resorption; Cachexia; Calcium; Humans; Mice; Muscle Weakness; Muscle, Skeletal; NADPH Oxidases; Neoplasms; Ryanodine Receptor Calcium Release Channel; Sarcoplasmic Reticulum; Signal Transduction; Smad3 Protein; Transforming Growth Factor beta; Troponin

2015
Comprehensive functional characterization of cancer-testis antigens defines obligate participation in multiple hallmarks of cancer.
    Nature communications, 2015, Nov-16, Volume: 6

    Tumours frequently activate genes whose expression is otherwise biased to the testis, collectively known as cancer-testis antigens (CTAs). The extent to which CTA expression represents epiphenomena or confers tumorigenic traits is unknown. In this study, to address this, we implemented a multidimensional functional genomics approach that incorporates 7 different phenotypic assays in 11 distinct disease settings. We identify 26 CTAs that are essential for tumor cell viability and/or are pathological drivers of HIF, WNT or TGFβ signalling. In particular, we discover that Foetal and Adult Testis Expressed 1 (FATE1) is a key survival factor in multiple oncogenic backgrounds. FATE1 prevents the accumulation of the stress-sensing BH3-only protein, BCL-2-Interacting Killer (BIK), thereby permitting viability in the presence of toxic stimuli. Furthermore, ZNF165 promotes TGFβ signalling by directly suppressing the expression of negative feedback regulatory pathways. This action is essential for the survival of triple negative breast cancer cells in vitro and in vivo. Thus, CTAs make significant direct contributions to tumour biology.

    Topics: Adenocarcinoma; Adenocarcinoma of Lung; Animals; Antigens, Neoplasm; Apoptosis Regulatory Proteins; Carcinogenesis; Cell Line; Cell Line, Tumor; Cell Survival; Colorectal Neoplasms; DNA-Binding Proteins; Fluorescent Antibody Technique; Gene Expression Regulation, Neoplastic; HCT116 Cells; HEK293 Cells; Humans; Immunoblotting; In Vitro Techniques; Lung Neoplasms; Membrane Proteins; Mice, Inbred NOD; Mitochondrial Proteins; Neoplasm Transplantation; Neoplasms; Prognosis; Proportional Hazards Models; Real-Time Polymerase Chain Reaction; Signal Transduction; Smad7 Protein; Transcription Factors; Transforming Growth Factor beta; Triple Negative Breast Neoplasms; Ubiquitin-Protein Ligases; Wnt Signaling Pathway

2015
Bone Morphogenetic Proteins in Spinal Surgery: What Is the Fusion Rate and Do They Cause Cancer?
    Spine, 2015, Volume: 40, Issue:22

    A retrospective cohort study.. The aim of this study was to determine the fusion rate using recombinant human bone morphogenetic protein (rhBMP) in spinal surgery and to estimate the risk of cancer subsequent to their use.. rhBMP may obviate the need for iliac crest bone graft harvest and provides similar or higher fusion rates than autologous bone graft. Recently, there are concerns that rhBMPs may either cause cancer or accelerate progression.. Patients were treated by 2 spine surgeons between 2002 and 2012. Inclusion criteria were patients who resided in the state of Victoria, Australia, undergoing lumbar fusion (anterior, lateral, posterior, and posterolateral) with rhBMP [either rhBMP-2 (Infuse) or rhBMP-7 (OP-1)]. Exclusion criteria were patients who reported having an invasive cancer diagnosis before the spinal fusion procedure. The occurrence of incident cancers was obtained from record linkage to the Victorian Cancer Registry.. A total of 527 patients were included in the cohort, with a mean follow-up of 4.4 years (1.8-11.5). Patients received Infuse in 77% of cases and OP-1 in 23%. The mean Infuse does was 10.2  mg (2.5-48.0) and 3.3  mg (1.7-6.6) for OP-1. There was no significant difference in fusion rates between Infuse (90.1%) and OP-1 (91.9%) (P = 0.42). The overall success of interbody fusion with rhBMP was 93.5% at 12 months. Twenty-seven patients were diagnosed with an invasive cancer since treatment (20 Infuse and 7 OP-1 patients). Comparing the observed numbers in our study cohort with those expected on the basis of the Victorian population's age and sex-specific rates, we observed that the study cohort was not at a significantly increased risk of cancer. The standardized incidence ratio for cancer overall (of any type) was 0.84 [95% confidence interval (95% CI) 0.56-1.21].. Off-label use of rhBMP provided high fusion rates with no evidence of a significantly increased risk of cancer.. 4.

    Topics: Adult; Aged; Aged, 80 and over; Australia; Bone Morphogenetic Protein 2; Bone Transplantation; Female; Humans; Incidence; Lumbar Vertebrae; Male; Middle Aged; Neoplasms; Off-Label Use; Recombinant Proteins; Registries; Retrospective Studies; Spinal Fusion; Transforming Growth Factor beta; Treatment Outcome; Young Adult

2015
CLIC4 regulates TGF-β-dependent myofibroblast differentiation to produce a cancer stroma.
    Oncogene, 2014, Feb-13, Volume: 33, Issue:7

    Cancer stroma has a profound influence on tumor development and progression. The conversion of fibroblasts to activated myofibroblasts is a hallmark of reactive tumor stroma. Among a number of factors involved in this conversion, transforming growth factor (TGF)-β has emerged as a major regulator. CLIC4, an integral protein in TGF-β signaling, is highly upregulated in stroma of multiple human cancers, and overexpression of CLIC4 in stromal cells enhances the growth of cancer xenografts. In this study, we show that conditioned media from tumor cell lines induces expression of both CLIC4 and the myofibroblast marker alpha smooth muscle actin (α-SMA) in stromal fibroblasts via TGF-β signaling. Genetic ablation of CLIC4 in primary fibroblasts prevents or reduces constitutive or TGF-β-induced expression of α-SMA and extracellular matrix components that are markers of myofibroblasts. CLIC4 is required for the activation of p38 map kinase by TGF-β, a pathway that signals myofibroblast conversion in stromal cells. This requirement involves the interaction of CLIC4 with PPM1a, the selective phosphatase of activated p38. Conditioned media from fibroblasts overexpressing CLIC4 increases tumor cell migration and invasion in a TGF-β-dependent manner and promotes epithelial to mesenchymal transition indicating that high stromal CLIC4 serves to enhance tumor invasiveness and progression. Thus, CLIC4 is significantly involved in the development of a nurturing tumor microenvironment by enhancing TGF-β signaling in a positive feedback loop. Targeting CLIC4 in tumor stroma should be considered as a strategy to mitigate some of the tumor enhancing effects of the cancer stroma.

    Topics: Animals; Cell Differentiation; Cell Line, Tumor; Cell Movement; Chloride Channels; Humans; MAP Kinase Signaling System; Mice; Mice, Inbred C57BL; Myofibroblasts; Neoplasm Invasiveness; Neoplasms; Phosphorylation; Protein Processing, Post-Translational; Stromal Cells; Transforming Growth Factor beta

2014
Camel milk inhibits inflammatory angiogenesis via downregulation of proangiogenic and proinflammatory cytokines in mice.
    APMIS : acta pathologica, microbiologica, et immunologica Scandinavica, 2014, Volume: 122, Issue:7

    Camel milk has traditionally been used to treat cancer, but this practice awaits scientific scrutiny, in particular its role in tumor angiogenesis, the key step involved in tumor growth and metastasis. We aimed to investigate the effects of camel milk on key components of inflammatory angiogenesis in sponge implant angiogenesis model. Polyester-polyurethane sponges, used as a framework for fibrovascular tissue growth, were implanted in Swiss albino mice and camel milk (25, 50 and 100 mg/kg/day) was administered for 14 days through installed cannula. The implants collected at day 14 post-implantation were processed for the assessment of hemoglobin (Hb), myeloperoxidase (MPO), N-acetylglucosaminidase (NAG), and collagen, which were used as indices for angiogenesis, neutrophil, and macrophage accumulation and extracellular matrix deposition, respectively. Relevant inflammatory, angiogenic, and fibrogenic cytokines were also determined. Camel milk treatment attenuated the main components of the fibrovascular tissue, wet weight, vascularization (Hb content), macrophage recruitment (NAG activity), collagen deposition and the levels of vascular endothelial growth factor (VEGF), interleukin (IL)-1β, IL-6, IL-17, tumor necrosis factor-α, and transforming growth factor-β. A regulatory function of camel milk on multiple parameters of the main components of inflammatory angiogenesis has been revealed, giving insight into the potential therapeutic benefit underlying the anti-cancer actions of camel milk.

    Topics: Acetylglucosaminidase; Angiogenesis Inhibitors; Animals; Camelus; Chemoprevention; Collagen; Cytokines; Disease Models, Animal; Hemoglobins; Inflammation; Interleukin-17; Interleukin-1beta; Interleukin-6; Lactation; Macrophages; Male; Mice; Milk; Neoplasms; Neovascularization, Pathologic; Neutrophils; Peroxidase; Polyesters; Polyurethanes; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Vascular Endothelial Growth Factor A

2014
In silico identification of potential therapeutic targets in the TGF-β signal transduction pathway.
    Molecular bioSystems, 2014, Mar-04, Volume: 10, Issue:3

    The transforming growth factor-β (TGF-β) superfamily of cytokines controls fundamental cellular processes, such as proliferation, motility, differentiation, and apoptosis. This fundamental role is emphasized by the widespread presence of mutations of the core components of the TGF-β signal transduction pathway in a number of human diseases. Therefore, there is an increasing interest in the development of therapies to specifically target this pathway. Here we develop a computational approach to identify potential intervention points that are capable of restoring the normal signaling dynamics to the mutated system while maintaining the behavior of normal cells substantially unperturbed. We apply this approach explicitly to the TGF-β pathway to study the signaling dynamics of mutated and normal cells treated with inhibitory drugs and identify the processes in the pathway that are most susceptible to therapeutic intervention.

    Topics: Cell Line, Tumor; Cluster Analysis; Computational Biology; Computer Simulation; Drug Discovery; Humans; Models, Biological; Mutation; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2014
Protein kinase inhibitor SU6668 attenuates positive regulation of Gli proteins in cancer and multipotent progenitor cells.
    Biochimica et biophysica acta, 2014, Volume: 1843, Issue:4

    Observations that Glioma-associated transcription factors Gli1 and Gli2 (Gli1/2), executers of the Sonic Hedgehog (Shh) signaling pathway and targets of the Transforming Growth Factor β (TGF-β) signaling axis, are involved in numerous developmental and pathological processes unveil them as attractive pharmaceutical targets. Unc-51-like serine/threonine kinase Ulk3 has been suggested to play kinase activity dependent and independent roles in the control of Gli proteins in the context of the Shh signaling pathway. This study aimed at investigating whether the mechanism of generation of Gli1/2 transcriptional activators has similarities regardless of the signaling cascade evoking their activation. We also elucidate further the role of Ulk3 kinase in regulation of Gli1/2 proteins and examine SU6668 as an inhibitor of Ulk3 catalytic activity and a compound targeting Gli1/2 proteins in different cell-based experimental models. Here we demonstrate that Ulk3 is required not only for maintenance of basal levels of Gli1/2 proteins but also for TGF-β or Shh dependent activation of endogenous Gli1/2 proteins in human adipose tissue derived multipotent stromal cells (ASCs) and mouse immortalized progenitor cells, respectively. We show that cultured ASCs possess the functional Shh signaling axis and differentiate towards osteoblasts in response to Shh. Also, we demonstrate that similarly to Ulk3 RNAi, SU6668 prevents de novo expression of Gli1/2 proteins and antagonizes the Gli-dependent activation of the gene expression programs induced by either Shh or TGF-β. Our data suggest SU6668 as an efficient inhibitor of Ulk3 kinase allowing manipulation of the Gli-dependent transcriptional outcome.

    Topics: Animals; Cell Differentiation; Cells, Cultured; Gene Expression Regulation, Developmental; Hedgehog Proteins; Humans; Indoles; Kruppel-Like Transcription Factors; Leukocytes, Mononuclear; Mice; Multipotent Stem Cells; Neoplasms; Nuclear Proteins; Oxindoles; Propionates; Protein Serine-Threonine Kinases; Pyrroles; Signal Transduction; Transcription Factors; Transforming Growth Factor beta; Zinc Finger Protein GLI1; Zinc Finger Protein Gli2

2014
An evidence-based knowledgebase of pulmonary arterial hypertension to identify genes and pathways relevant to pathogenesis.
    Molecular bioSystems, 2014, Volume: 10, Issue:4

    Pulmonary arterial hypertension (PAH) is a major progressive form of pulmonary hypertension (PH) with more than 4800 patients in the United States. In the last two decades, many studies have identified numerous genes associated with this disease. However, there is no comprehensive research resource for PAH or other PH types that integrates various genetic studies and their related biological information. Thus, the number of associated genes, and their strength of evidence, is unclear. In this study, we tested the hypothesis that a web-based knowledgebase could be used to develop a biological map of highly interrelated, functionally important genes in PAH. We developed the pulmonary arterial hypertension knowledgebase (PAHKB, ), a comprehensive database with a user-friendly web interface. PAHKB extracts genetic data from all available sources, including those from association studies, genetic mutation, gene expression, animal model, supporting literature, various genomic annotations, gene networks, cellular and regulatory pathways, as well as microRNAs. Moreover, PAHKB provides online tools for data browsing and searching, data integration, pathway graphical presentation, and gene ranking. In the current release, PAHKB contains 341 human PH-related genes (293 protein coding and 48 non-coding genes) curated from over 1000 PubMed abstracts. Based on the top 39 ranked PAH-related genes in PAHKB, we constructed a core biological map. This core map was enriched with the TGF-beta signaling pathway, focal adhesion, cytokine-cytokine receptor interaction, and MAPK signaling. In addition, the reconstructed map elucidates several novel cancer signaling pathways, which may provide clues to support the application of anti-cancer therapeutics to PAH. In summary, we have developed a system for the identification of core PH-related genes and identified critical signaling pathways that may be relevant to PAH pathogenesis. This system can be easily applied to other pulmonary diseases.

    Topics: Chromosome Mapping; Data Collection; Databases, Genetic; Familial Primary Pulmonary Hypertension; Focal Adhesions; Humans; Internet; MAP Kinase Signaling System; Neoplasms; Receptors, Cytokine; Transforming Growth Factor beta

2014
Inhibition of TBK1 attenuates radiation-induced epithelial-mesenchymal transition of A549 human lung cancer cells via activation of GSK-3β and repression of ZEB1.
    Laboratory investigation; a journal of technical methods and pathology, 2014, Volume: 94, Issue:4

    Radiotherapy is an effective treatment method for lung cancer, particularly when the disease is at an advanced stage. However, previous researchers have observed that the majority of patients with conventional radiation therapy develop distant metastases and succumb to the disease. Thus, identifying and understanding novel pathways for the development of new therapeutic targets is a major goal in research on pulmonary neoplasms. Recent studies suggest that epithelial-mesenchymal transition (EMT) is the most important contributor to cancer metastasis. Induction of this complex process requires endogenously produced microRNAs; specifically, downregulation of the miRNA-200c causes an induction of EMT. We recently identified the tank-binding kinase-1 (TBK1) as a downstream effector of the miR-200c-driven pathway, but the biological function of TBK1 in EMT remains unknown. In this study, we tested whether TBK1 has a role in radiation-induced EMT and identified associated potential mechanisms. Human alveolar type II epithelial carcinoma A549 cells were irradiated with (60)Co γ-rays. Western blotting revealed a time- and dose-dependent decrease in E-cadherin with a concomitant increase in vimentin after radiation, suggesting that the epithelial cells acquired a mesenchymal-like morphology. TBK1 siRNA significantly inhibited radiation-induced suppression of the epithelial marker E-cadherin and upregulation of the mesenchymal marker vimentin. The invasion and migratory potential of lung cancer cells upon radiation treatment was also reduced by TBK1 knockdown. Furthermore, radiation-induced EMT attenuated by TBK1 depletion was partially dependent on transcriptional factor ZEB1 expression. Finally, we found glycogen synthase kinase-3β (GSK-3β) is involved in regulation of radiation-induced EMT by TBK1. Thus, our findings reveal that TBK1 signaling regulates radiation-induced EMT by controlling GSK-3β phosphorylation and ZEB1 expression. TBK1 may therefore constitute a useful target for treatment of radiotherapy-induced metastasis diseases.

    Topics: Cell Line, Tumor; Epithelial-Mesenchymal Transition; Gamma Rays; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Homeodomain Proteins; Humans; Neoplasm Metastasis; Neoplasms; NF-kappa B; Protein Serine-Threonine Kinases; Radiotherapy; Smad Proteins; Transcription Factors; Transforming Growth Factor beta; Zinc Finger E-box-Binding Homeobox 1

2014
Transforming growth factor β in cancer: Janus, the two-faced god.
    Journal of the National Cancer Institute, 2014, Volume: 106, Issue:2

    Topics: Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2014
TGFβ-mediated suppression of CD248 in non-cancer cells via canonical Smad-dependent signaling pathways is uncoupled in cancer cells.
    BMC cancer, 2014, Feb-20, Volume: 14

    CD248 is a cell surface glycoprotein, highly expressed by stromal cells and fibroblasts of tumors and inflammatory lesions, but virtually undetectable in healthy adult tissues. CD248 promotes tumorigenesis, while lack of CD248 in mice confers resistance to tumor growth. Mechanisms by which CD248 is downregulated are poorly understood, hindering the development of anti-cancer therapies.. We sought to characterize the molecular mechanisms by which CD248 is downregulated by surveying its expression in different cells in response to cytokines and growth factors.. Only transforming growth factor (TGFβ) suppressed CD248 protein and mRNA levels in cultured fibroblasts and vascular smooth muscle cells in a concentration- and time-dependent manner. TGFβ transcriptionally downregulated CD248 by signaling through canonical Smad2/3-dependent pathways, but not via mitogen activated protein kinases p38 or ERK1/2. Notably, cancer associated fibroblasts (CAF) and cancer cells were resistant to TGFβ mediated suppression of CD248.. The findings indicate that decoupling of CD248 regulation by TGFβ may contribute to its tumor-promoting properties, and underline the importance of exploring the TGFβ-CD248 signaling pathway as a potential therapeutic target for early prevention of cancer and proliferative disorders.

    Topics: Activins; Animals; Antigens, CD; Antigens, Neoplasm; Bone Morphogenetic Protein 2; Cell Line, Tumor; Cytokines; Disease Models, Animal; Gene Expression Regulation, Neoplastic; Humans; Mice; Mice, Knockout; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Neoplasms; p38 Mitogen-Activated Protein Kinases; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; RNA Stability; RNA, Messenger; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2014
TGF-β-inducible microRNA-183 silences tumor-associated natural killer cells.
    Proceedings of the National Academy of Sciences of the United States of America, 2014, Mar-18, Volume: 111, Issue:11

    Transforming growth factor β1 (TGF-β), enriched in the tumor microenvironment and broadly immunosuppressive, inhibits natural killer (NK) cell function by yet-unknown mechanisms. Here we show that TGF-β-treated human NK cells exhibit reduced tumor cytolysis and abrogated perforin polarization to the immune synapse. This result was accompanied by loss of surface expression of activating killer Ig-like receptor 2DS4 and NKp44, despite intact cytoplasmic stores of these receptors. Instead, TGF-β depleted DNAX activating protein 12 kDa (DAP12), which is critical for surface NK receptor stabilization and downstream signal transduction. Mechanistic analysis revealed that TGF-β induced microRNA (miR)-183 to repress DAP12 transcription/translation. This pathway was confirmed with luciferase reporter constructs bearing the DAP12 3' untranslated region as well as in human NK cells by use of sense and antisense miR-183. Moreover, we documented reduced DAP12 expression in tumor-associated NK cells in lung cancer patients, illustrating this pathway to be consistently perturbed in the human tumor microenvironment.

    Topics: Adaptor Proteins, Signal Transducing; Flow Cytometry; Humans; Immunoblotting; Immunohistochemistry; Killer Cells, Natural; Luciferases; Membrane Proteins; MicroRNAs; Microscopy, Fluorescence; Neoplasms; Receptors, Natural Killer Cell; Signal Transduction; Transforming Growth Factor beta

2014
Identification, mechanism of action, and antitumor activity of a small molecule inhibitor of hippo, TGF-β, and Wnt signaling pathways.
    Molecular cancer therapeutics, 2014, Volume: 13, Issue:6

    Embryonic signaling pathways, in particular those mediated by Wnt and TGF-β, are known to play key roles in tumor progression through the induction of epithelial-mesenchymal transition (EMT). Their simultaneous targeting could therefore represent a desirable anticancer strategy. On the basis of recent findings that both Wnt and TGF-β-associated pathways are regulated by Hippo signaling in mammalian cells, we reasoned that targeting the latter would be more effective in inhibiting EMT. In a search for such inhibitors, we identified a small molecule (C19) with remarkable inhibitory activity not only against Hippo, but also against Wnt and TGF-β pathways. C19 inhibited cancer cell migration, proliferation, and resistance to doxorubicin in vitro, and exerted strong antitumor activity in a mouse tumor model. Mechanistically, C19 induced GSK3-β-mediated degradation of the Hippo transducer TAZ, through activation of the Hippo kinases Mst/Lats and the tumor suppressor kinase AMPK upstream of the degradation complex. Overall, this study identified C19 as a multi-EMT pathway inhibitor with a unique mechanism of action. The findings that both AMPK and Mst/Lats mediate the antitumor activity of C19 shed light on a potential cross-talk between metabolic and organ size control pathways in regulating cancer progression. By simultaneously targeting these two pathways, C19 may represent a new type of agents to suppress cancer progression and/or its recurrence.

    Topics: Acyltransferases; AMP-Activated Protein Kinases; Animals; Antineoplastic Agents; Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Glycogen Synthase Kinase 3; Hippo Signaling Pathway; Humans; Mice; Neoplasm Recurrence, Local; Neoplasms; Propanols; Protein Serine-Threonine Kinases; Thiadiazoles; Transcription Factors; Transforming Growth Factor beta; Wnt Signaling Pathway

2014
Identification of novel non-coding RNA-based negative feedback regulating the expression of the oncogenic transcription factor GLI1.
    Molecular oncology, 2014, Volume: 8, Issue:5

    Non-coding RNAs are a complex class of nucleic acids, with growing evidence supporting regulatory roles in gene expression. Here we identify a non-coding RNA located head-to-head with the gene encoding the Glioma-associated oncogene 1 (GLI1), a transcriptional effector of multiple cancer-associated signaling pathways. The expression of this three-exon GLI1 antisense (GLI1AS) RNA in cancer cells was concordant with GLI1 levels. siRNAs knockdown of GLI1AS up-regulated GLI1 and increased cellular proliferation and tumor growth in a xenograft model system. Conversely, GLI1AS overexpression decreased the levels of GLI1, its target genes PTCH1 and PTCH2, and cellular proliferation. Additionally, we demonstrate that GLI1 knockdown reduced GLI1AS, while GLI1 overexpression increased GLI1AS, supporting the role of GLI1AS as a target gene of the GLI1 transcription factor. Activation of TGFβ and Hedgehog signaling, two known regulators of GLI1 expression, conferred a concordant up-regulation of GLI1 and GLI1AS in cancer cells. Finally, analysis of the mechanism underlying the interplay between GLI1 and GLI1AS indicates that the non-coding RNA elicits a local alteration of chromatin structure by increasing the silencing mark H3K27me3 and decreasing the recruitment of RNA polymerase II to this locus. Taken together, the data demonstrate the existence of a novel non-coding RNA-based negative feedback loop controlling GLI1 levels, thus expanding the repertoire of mechanisms regulating the expression of this oncogenic transcription factor.

    Topics: Cell Line, Tumor; Chromatin; Gene Expression Regulation, Neoplastic; Hedgehog Proteins; Humans; Neoplasms; Oncogene Proteins; RNA Polymerase II; RNA, Untranslated; Signal Transduction; Trans-Activators; Transcriptional Activation; Transforming Growth Factor beta; Zinc Finger Protein GLI1

2014
Comparing stochastic differential equations and agent-based modelling and simulation for early-stage cancer.
    PloS one, 2014, Volume: 9, Issue:4

    There is great potential to be explored regarding the use of agent-based modelling and simulation as an alternative paradigm to investigate early-stage cancer interactions with the immune system. It does not suffer from some limitations of ordinary differential equation models, such as the lack of stochasticity, representation of individual behaviours rather than aggregates and individual memory. In this paper we investigate the potential contribution of agent-based modelling and simulation when contrasted with stochastic versions of ODE models using early-stage cancer examples. We seek answers to the following questions: (1) Does this new stochastic formulation produce similar results to the agent-based version? (2) Can these methods be used interchangeably? (3) Do agent-based models outcomes reveal any benefit when compared to the Gillespie results? To answer these research questions we investigate three well-established mathematical models describing interactions between tumour cells and immune elements. These case studies were re-conceptualised under an agent-based perspective and also converted to the Gillespie algorithm formulation. Our interest in this work, therefore, is to establish a methodological discussion regarding the usability of different simulation approaches, rather than provide further biological insights into the investigated case studies. Our results show that it is possible to obtain equivalent models that implement the same mechanisms; however, the incapacity of the Gillespie algorithm to retain individual memory of past events affects the similarity of some results. Furthermore, the emergent behaviour of ABMS produces extra patters of behaviour in the system, which was not obtained by the Gillespie algorithm.

    Topics: Algorithms; Computer Simulation; Humans; Interleukin-2; Models, Biological; Neoplasm Staging; Neoplasms; Regression Analysis; Stochastic Processes; Transforming Growth Factor beta

2014
Acquisition of useful sero-diagnostic autoantibodies using the same patients'sera and tumor tissues.
    Biomedical research (Tokyo, Japan), 2014, Volume: 35, Issue:2

    Cancer tissues are comprised of various components including tumor cells and the surrounding tumor stroma, which consists of the extracellular matrix and inflammatory cells. Since the tumor stroma plays critical roles in tumor development, investigation of the tumor stroma in addition to tumor cells is important to identify useful tumor-associated markers. To discover novel and useful sero-diagnostic markers, a comparative study of tumor-associated autoantibodies (AAbs) in sera from lung adenocarcinoma (AC) patients was investigated by two-dimensional immunoblotting with AC cell lines or each autologous AC tissues. Autoantigens identified from tissue and cell line samples comprised 58 (45 antigens) and 53 spots (41 antigens), respectively. Thirty-six proteins including Transforming growth factor-beta-induced protein ig-h3 (BIGH3) and Hyaluronan and proteoglycan link protein 1 (HAPLN1) were detected only from tissues, 32 proteins only from cell lines, and 9 proteins from both. BIGH3 and HAPLN1 expressions were confirmed in the tumor stroma, but not in AC cell lines by immunostaining and immunoblotting. These data suggest that autologous tumor tissue and serum are important to coincidently detect AAbs derived from the tumor stroma in addition to tumor cells.

    Topics: Adenocarcinoma; Adenocarcinoma of Lung; Aged; Annexin A2; Antigens, Neoplasm; Autoantibodies; Autoantigens; Biomarkers, Tumor; Cell Line, Tumor; Electrophoresis, Gel, Two-Dimensional; Extracellular Matrix Proteins; Female; Humans; Immunohistochemistry; Lung Neoplasms; Male; Microfilament Proteins; Middle Aged; Neoplasms; Nuclear Proteins; Proteoglycans; Transforming Growth Factor beta

2014
Aberrant miRNA profiles associated with chronic benzene poisoning.
    Experimental and molecular pathology, 2014, Volume: 96, Issue:3

    Chronic occupational benzene exposure is associated with an increased risk of hematological malignancies. To gain an insight into the new biomarkers and molecular mechanisms of chronic benzene poisoning, miRNA profiles and mRNA expression pattern from the peripheral blood mononuclear cells of chronic benzene poisoning patients and health controls matched age and gender without benzene exposure were performed using the Exiqon miRNA PCR ARRAY and Gene Chip Human Gene 2.0ST Arrays, respectively. Totally, 6 up-regulated miRNAs (miR-34a, miR-205, miR-10b, let-7d, miR-185 and miR-423-5p-2) and 7 down-regulated miRNAs (miR-133a, miR-543, hsa-miR-130a, miR-27b,miR-223, miR-142-5p and miR-320b) were found in chronic benzene poisoning group compared to health controls (P ≤ 0.05). By integrating miRNA and mRNA expression data, these differential miRNAs were mainly involved in regulation of transcription from RNA polymerase II promoter, axon guidance, regulation of transcription, DNA-dependent, nervous system development, and regulation of actin cytoskeleton organization. Further, pathway analysis indicated that SMAD4, PLCB1, NFAT5, GNAI2, PTEN, VEGFA, BCL2, CTNNB1 and CCND1 were key target genes of differential miRNAs which were implicated in Adherens junction, TGF-beta signaling pathway, Wnt signaling pathway, tight junction and Pathways in cancer. In conclusion, the aberrant miRNAs might be a potential biomarker of chronic benzene poisoning.

    Topics: Benzene; Case-Control Studies; Down-Regulation; Genetic Markers; Humans; Leukocytes, Mononuclear; MicroRNAs; Neoplasms; Oligonucleotide Array Sequence Analysis; Phospholipase C beta; Promoter Regions, Genetic; RNA, Messenger; Smad4 Protein; Transcription Factors; Transcriptome; Transforming Growth Factor beta; Up-Regulation; Wnt Signaling Pathway

2014
A comparative study of the effects of inhibitory cytokines on human natural killer cells and the mechanistic features of transforming growth factor-beta.
    Cellular immunology, 2014, Volume: 290, Issue:1

    The major factors and mechanisms by which natural killer (NK) cells are inhibited in cancer patients have not yet been well defined. In this study, we conducted a comparative analysis of the effects of TGF-β, IL-10, and IL-4 on primary NK cells, and it was demonstrated that (1) TGF-β most potently inhibited the overall function of NK cells. (2) It appears that TGF-β reduced the tyrosine phosphorylation of Syk and the expression of c-myc. (3) It was also found that the IL-2-induced promoter-binding activities of C-myb, AP-1, CREB, and AR were also completely suppressed upon TGF-β treatment. Interestingly, TGF-β also completely suppressed other transcription factors, which are constitutively activated. Among these factors, we further confirmed roles of AP-1 in NK-92 cell activation through c-jun and MEK1 inhibitor assay. Our study provides insight into the effects of TGF-β in modulating NK cell functions.

    Topics: Anthracenes; Cell Line; Cell Proliferation; Cyclic AMP Response Element-Binding Protein; Humans; Interferon-gamma; Interleukin-10; Interleukin-2; Interleukin-4; Intracellular Signaling Peptides and Proteins; JNK Mitogen-Activated Protein Kinases; Killer Cells, Natural; Lymphocyte Activation; MAP Kinase Kinase 1; Neoplasms; Phosphorylation; Promoter Regions, Genetic; Protein-Tyrosine Kinases; Proto-Oncogene Proteins c-myb; Proto-Oncogene Proteins c-myc; Receptors, Androgen; Signal Transduction; Syk Kinase; Transcription Factor AP-1; Transforming Growth Factor beta; Tumor Escape

2014
Increased antitumor effects using IL-2 with anti-TGF-β reveals competition between mouse NK and CD8 T cells.
    Journal of immunology (Baltimore, Md. : 1950), 2014, Aug-15, Volume: 193, Issue:4

    Because of increasing interest in the removal of immunosuppressive pathways in cancer, the combination of IL-2 with Abs to neutralize TGF-β, a potent immunosuppressive cytokine, was assessed. Combination immunotherapy resulted in significantly greater antitumor effects. These were correlated with significant increases in the numbers and functionality of NK cells, NK cell progenitors, and activated CD8 T cells, resulting in the observed antitumor effects. Combination immunotherapy also was accompanied by lesser toxicities than was IL-2 therapy alone. Additionally, we observed a dual competition between NK cells and activated CD8 T cells such that, after immunotherapy, the depletion of either effector population resulted in the increased total expansion of the other population and compensatory antitumor effects. This study demonstrates the efficacy of this combination immunotherapeutic regimen as a promising cancer therapy and illustrates the existence of potent competitive regulatory pathways between NK cells and CD8 T cells in response to systemic activation.

    Topics: Animals; Antibodies, Neutralizing; CD8-Positive T-Lymphocytes; Cell Line, Tumor; Fas Ligand Protein; fas Receptor; Female; Immunotherapy; Interleukin-2; Killer Cells, Natural; Lymphocyte Activation; Mice; Mice, Inbred C57BL; Mice, Knockout; Neoplasms; NK Cell Lectin-Like Receptor Subfamily A; NK Cell Lectin-Like Receptor Subfamily C; NK Cell Lectin-Like Receptor Subfamily K; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2014
Tumor-derived transforming growth factor-β is critical for tumor progression and evasion from immune surveillance.
    Asian Pacific journal of cancer prevention : APJCP, 2014, Volume: 15, Issue:13

    Tumors have evolved numerous mechanisms by which they can escape from immune surveillance. One of these is to produce immunosuppressive cytokines. Transforming growth factor-β(TGF-β) is a pleiotropic cytokine with a crucial function in mediating immune suppression, especially in the tumor microenvironment. TGF-β produced by T cells has been demonstrated as an important factor for suppressing antitumor immune responses, but the role of tumor-derived TGF-β in this process is poorly understood. In this study, we demonstrated that knockdown of tumor-derived TGF-β using shRNA resulted in dramatically reduced tumor size, slowing tumor formation, prolonging survival rate of tumor-bearing mice and inhibiting metastasis. We revealed possible underlying mechanisms as reducing the number of myeloid-derived suppressor cells (MDSC) and CD4+Foxp3+ Treg cells, and consequently enhanced IFN-γ production by CTLs. Knockdown of tumor-derived TGF-β also significantly reduced the conversion of naive CD4+ T cells into Treg cells in vitro. Finally, we found that knockdown of TGF-β suppressed cell migration, but did not change the proliferation and apoptosis of tumor cells in vitro. In summary, our study provided evidence that tumor-derived TGF-β is a critical factor for tumor progression and evasion of immune surveillance, and blocking tumor-derived TGF-β may serve as a potential therapeutic approach for cancer.

    Topics: Animals; Apoptosis; CD4-Positive T-Lymphocytes; Cell Line, Tumor; Cell Proliferation; Disease Progression; Interferon-gamma; Male; Melanoma, Experimental; Mice; Mice, Inbred C57BL; Monitoring, Immunologic; Myeloid Cells; Neoplasms; Survival Rate; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2014
SMAD4 suppresses AURKA-induced metastatic phenotypes via degradation of AURKA in a TGFβ-independent manner.
    Molecular cancer research : MCR, 2014, Volume: 12, Issue:12

    SMAD4 has been suggested to inhibit the activity of the WNT/β-catenin signaling pathway in cancer. However, the mechanism by which SMAD4 antagonizes WNT/β-catenin signaling in cancer remains largely unknown. Aurora A kinase (AURKA), which is frequently overexpressed in cancer, increases the transcriptional activity of β-catenin/T-cell factor (TCF) complex by stabilizing β-catenin through the inhibition of GSK-3β. Here, SMAD4 modulated AURKA in a TGFβ-independent manner. Overexpression of SMAD4 significantly suppressed AURKA function, including colony formation, migration, and invasion of cell lines. In addition, SMAD4 bound to AURKA induced degradation of AURKA by the proteasome. A luciferase activity assay revealed that the transcriptional activity of the β-catenin/TCF complex was elevated by AURKA, but decreased by SMAD4 overexpression. Moreover, target gene analysis showed that SMAD4 abrogated the AURKA-mediated increase of β-catenin target genes. However, this inhibitory effect of SMAD4 was abolished by overexpression of AURKA or silencing of AURKA in SMAD4-overexpressed cells. Meanwhile, the SMAD4-mediated repression of AURKA and β-catenin was independent of TGFβ signaling because blockage of TGFβR1 or restoration of TGFβ signaling did not prevent suppression of AURKA and β-catenin signaling by SMAD4. These results indicate that the tumor-suppressive function of SMAD4 is mediated by downregulation of β-catenin transcriptional activity via AURKA degradation in a TGFβ-independent manner.. SMAD4 interacts with AURKA and antagonizes its tumor-promoting potential, thus demonstrating a novel mechanism of tumor suppression.

    Topics: Animals; Aurora Kinase A; beta Catenin; Cell Line, Tumor; Cell Movement; HeLa Cells; Humans; Mice; Mutation; Neoplasm Metastasis; Neoplasms; NIH 3T3 Cells; Proteasome Endopeptidase Complex; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta

2014
Inhibition of IL-17A suppresses enhanced-tumor growth in low dose pre-irradiated tumor beds.
    PloS one, 2014, Volume: 9, Issue:9

    Ionizing radiation induces modification of the tumor microenvironment such as tumor surrounding region, which is relevant to treatment outcome after radiotherapy. In this study, the effects of pre-irradiated tumor beds on the growth of subsequently implanted tumors were investigated as well as underlying mechanism. The experimental model was set up by irradiating the right thighs of C3H/HeN mice with 5 Gy, followed by the implantation of HCa-I and MIH-2. Both implanted tumors in the pre-irradiated bed showed accelerated-growth compared to the control. Tumor-infiltrated lymphocyte (TIL) levels were increased, as well as pro-tumor factors such as IL-6 and transforming growth factor-beta1 (TGF-β1) in the pre-irradiated group. In particular, the role of pro-tumor cytokine interleukin-17A (IL-17A) was investigated as a possible target mechanism because IL-6 and TGF-β are key factors in Th17 cells differentiation from naïve T cells. IL-17A expression was increased not only in tumors, but also in CD4+ T cells isolated from the tumor draining lymph nodes. The effect of IL-17A on tumor growth was confirmed by treating tumors with IL-17A antibody, which abolished the acceleration of tumor growth. These results indicate that the upregulation of IL-17A seems to be a key factor for enhancing tumor growth in pre-irradiated tumor beds.

    Topics: Animals; Antibodies, Neutralizing; Cell Differentiation; Cell Proliferation; Disease Progression; Dose-Response Relationship, Radiation; Interleukin-17; Interleukin-6; Lymph Nodes; Lymphocytes, Tumor-Infiltrating; Male; Mice, Inbred C3H; Neoplasm Transplantation; Neoplasms; Neutralization Tests; Radiation, Ionizing; Th17 Cells; Transforming Growth Factor beta

2014
Cancer risk from bone morphogenetic protein exposure in spinal arthrodesis.
    The Journal of bone and joint surgery. American volume, 2014, Sep-03, Volume: 96, Issue:17

    The U.S. Food and Drug Administration reported a higher incidence of cancer in patients who had spinal arthrodesis and were exposed to a high dose of recombinant human bone morphogenetic protein-2 (rhBMP-2) compared with the control group in a randomized controlled trial. The purpose of this study was to determine the risk of cancer after spinal arthrodesis with BMP.. We retrospectively analyzed the incidence of cancer in 467,916 Medicare patients undergoing spinal arthrodesis from 2005 to 2010. Patients with a preexisting diagnosis of cancer were excluded. The average follow-up duration was 2.85 years for the BMP group and 2.94 years for the control group. The main outcome measure was the relative risk of developing new malignant lesions after spinal arthrodesis with or without exposure to BMP.. The relative risk of developing cancer after BMP exposure was 0.938 (95% confidence interval [95% CI]: 0.913 to 0.964), which was significant. In the BMP group, 5.9% of the patients developed an invasive cancer compared with 6.5% of the patients in the control group. The relative risk of developing cancer after BMP exposure was 0.98 in males (95% CI: 0.94 to 1.02) and 0.93 (95% CI: 0.90 to 0.97) in females. The control group showed a higher incidence of each type of cancer except pancreatic cancer.. Recent clinical use of BMP was not associated with a detectable increase in the risk of cancer within a mean 2.9-year time window.. Therapeutic Level III. See Instructions for Authors for a complete description of levels of evidence.

    Topics: Age Distribution; Aged; Aged, 80 and over; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Case-Control Studies; Confidence Intervals; Dose-Response Relationship, Drug; Drug Administration Schedule; Female; Follow-Up Studies; Humans; Incidence; Male; Medicare; Middle Aged; Neoplasms; Predictive Value of Tests; Recombinant Proteins; Retrospective Studies; Risk Assessment; SEER Program; Sex Distribution; Spinal Diseases; Spinal Fusion; Time Factors; Transforming Growth Factor beta; Treatment Outcome; United States

2014
Reducing CD73 expression by IL1β-Programmed Th17 cells improves immunotherapeutic control of tumors.
    Cancer research, 2014, Nov-01, Volume: 74, Issue:21

    T cells of the T helper (Th)17 subset offer promise in adoptive T-cell therapy for cancer. However, current protocols for ex vivo programming of Th17 cells, which include TGFβ exposure, increase the expression of CD39 and CD73, two cell surface ATP ectonucleotidases that reduce T-cell effector functions and promote immunosuppression. Here, we report that ATP-mediated suppression of IFNγ production by Th17 cells can be overcome by genetic ablation of CD73 or by using IL1β instead of TGFβ to program Th17 cells ex vivo. Th17 cells cultured in IL1β were also highly polyfunctional, expressing high levels of effector molecules and exhibiting superior short-term control of melanoma in mice, despite reduced stem cell-like properties. TGFβ addition at low doses that did not upregulate CD73 expression but induced stemness properties drastically improved the antitumor effects of IL1β-cultured Th17 cells. Effector properties of IL1β-dependent Th17 cells were likely related to their high glycolytic capacity, since ex vivo programming in pyruvate impaired glycolysis and antitumor effects. Overall, we show that including TGFβ in ex vivo cultures used to program Th17 cells blunts their immunotherapeutic potential and demonstrate how this potential can be more fully realized for adoptive T-cell therapy.

    Topics: 5'-Nucleotidase; Animals; Antigens, CD; Apyrase; Cell Line, Tumor; Gene Expression Regulation, Neoplastic; GPI-Linked Proteins; Humans; Immune Tolerance; Immunotherapy, Adoptive; Interleukin-1beta; Mice; Neoplasms; Th17 Cells; Transforming Growth Factor beta

2014
Targeting foxp1 for reinstating anticancer immunosurveillance.
    Immunity, 2014, Sep-18, Volume: 41, Issue:3

    Transforming growth factor β (TGF-β) is a canonical immunosuppressive cytokine secreted by tumors. In this issue of Immunity, Stephen et al. (2014) reveal that tumor-derived TGF-β deactivates antitumor CD8(+) T cell responses through T cell upregulation of the FoxP1 transcription factor.

    Topics: Animals; Female; Forkhead Transcription Factors; Neoplasms; Repressor Proteins; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Tumor Escape

2014
Transforming growth factor β-mediated suppression of antitumor T cells requires FoxP1 transcription factor expression.
    Immunity, 2014, Sep-18, Volume: 41, Issue:3

    Tumor-reactive T cells become unresponsive in advanced tumors. Here we have characterized a common mechanism of T cell unresponsiveness in cancer driven by the upregulation of the transcription factor Forkhead box protein P1 (Foxp1), which prevents CD8⁺ T cells from proliferating and upregulating Granzyme-B and interferon-γ in response to tumor antigens. Accordingly, Foxp1-deficient lymphocytes induced rejection of incurable tumors and promoted protection against tumor rechallenge. Mechanistically, Foxp1 interacted with the transcription factors Smad2 and Smad3 in preactivated CD8⁺ T cells in response to microenvironmental transforming growth factor-β (TGF-β), and was essential for its suppressive activity. Therefore, Smad2 and Smad3-mediated c-Myc repression requires Foxp1 expression in T cells. Furthermore, Foxp1 directly mediated TGF-β-induced c-Jun transcriptional repression, which abrogated T cell activity. Our results unveil a fundamental mechanism of T cell unresponsiveness different from anergy or exhaustion, driven by TGF-β signaling on tumor-associated lymphocytes undergoing Foxp1-dependent transcriptional regulation.

    Topics: Adoptive Transfer; Animals; Antigens, Neoplasm; CD4-Positive T-Lymphocytes; Cell Proliferation; Female; Forkhead Transcription Factors; Gene Expression Regulation; Granzymes; Interferon-gamma; JNK Mitogen-Activated Protein Kinases; Lymphocyte Activation; Mice; Mice, Inbred C57BL; Mice, Transgenic; Neoplasms; Proto-Oncogene Proteins c-myc; Repressor Proteins; Signal Transduction; Smad2 Protein; Smad3 Protein; T-Lymphocytes, Cytotoxic; Transcription, Genetic; Transcriptional Activation; Transforming Growth Factor beta; Tumor Escape; Tumor Microenvironment

2014
EED regulates epithelial-mesenchymal transition of cancer cells induced by TGF-β.
    Biochemical and biophysical research communications, 2014, Oct-10, Volume: 453, Issue:1

    Histone methylation is involved in various biological and pathological processes including cancer development. In this study, we found that EED, a component of Polycomb repressive complex-2 (PRC2) that catalyzes methylation of lysine 27 of histone H3 (H3K27), was involved in epithelial-mesenchymal transition (EMT) of cancer cells induced by Transforming Growth Factor-beta (TGF-β). The expression of EED was increased during TGF-β-induced EMT and knockdown of EED inhibited TGF-β-induced morphological conversion of the cells associated with EMT. EED knockdown antagonized TGF-β-dependent expression changes of EMT-related genes such as CDH1, ZEB1, ZEB2 and microRNA-200 (miR-200) family. Chromatin immunoprecipitation assays showed that EED was implicated in TGF-β-induced transcriptional repression of CDH1 and miR-200 family genes through the regulation of histone H3 methylation and EZH2 occupancies on their regulatory regions. Our study demonstrated a novel role of EED, which regulates PRC2 activity and histone methylation during TGF-β-induced EMT of cancer cells.

    Topics: Antigens, CD; Cadherins; Cell Line, Tumor; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Histones; Homeodomain Proteins; HT29 Cells; Humans; MicroRNAs; Neoplasms; Polycomb Repressive Complex 2; Promoter Regions, Genetic; Repressor Proteins; Transcription Factors; Transforming Growth Factor beta; Zinc Finger E-box Binding Homeobox 2; Zinc Finger E-box-Binding Homeobox 1

2014
TGFβ receptor 1: an immune susceptibility gene in HPV-associated cancer.
    Cancer research, 2014, Dec-01, Volume: 74, Issue:23

    Only a minority of those exposed to human papillomavirus (HPV) develop HPV-related cervical and oropharyngeal cancer. Because host immunity affects infection and progression to cancer, we tested the hypothesis that genetic variation in immune-related genes is a determinant of susceptibility to oropharyngeal cancer and other HPV-associated cancers by performing a multitier integrative computational analysis with oropharyngeal cancer data from a head and neck cancer genome-wide association study (GWAS). Independent analyses, including single-gene, gene-interconnectivity, protein-protein interaction, gene expression, and pathway analysis, identified immune genes and pathways significantly associated with oropharyngeal cancer. TGFβR1, which intersected all tiers of analysis and thus selected for validation, replicated significantly in the head and neck cancer GWAS limited to HPV-seropositive cases and an independent cervical cancer GWAS. The TGFβR1 containing p38-MAPK pathway was significantly associated with oropharyngeal cancer and cervical cancer, and TGFβR1 was overexpressed in oropharyngeal cancer, cervical cancer, and HPV(+) head and neck cancer tumors. These concordant analyses implicate TGFβR1 signaling as a process dysregulated across HPV-related cancers. This study demonstrates that genetic variation in immune-related genes is associated with susceptibility to oropharyngeal cancer and implicates TGFβR1/TGFβ signaling in the development of both oropharyngeal cancer and cervical cancer. Better understanding of the immunogenetic basis of susceptibility to HPV-associated cancers may provide insight into host/virus interactions and immune processes dysregulated in the minority of HPV-exposed individuals who progress to cancer.

    Topics: Female; Genome-Wide Association Study; Host-Pathogen Interactions; Humans; MAP Kinase Signaling System; Neoplasms; p38 Mitogen-Activated Protein Kinases; Papillomaviridae; Papillomavirus Infections; Polymorphism, Single Nucleotide; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

2014
S-nitrosoglutathione accelerates recovery from 5-fluorouracil-induced oral mucositis.
    PloS one, 2014, Volume: 9, Issue:12

    Mucositis induced by anti-neoplastic drugs is an important, dose-limiting and costly side-effect of cancer therapy.. To evaluate the effect of the topical application of S-nitrosoglutathione (GSNO), a nitric oxide donor, on 5-fluorouracil (5-FU)-induced oral mucositis in hamsters.. Oral mucositis was induced in male hamsters by two intraperitoneal administrations of 5-FU on the first and second days of the experiment (60 and 40 mg/kg, respectively) followed by mechanical trauma on the fourth day. Animals received saline, HPMC or HPMC/GSNO (0.1, 0.5 or 2.0 mM) 1 h prior to the 5-FU injection and twice a day for 10 or 14 days. Samples of cheek pouches were harvested for: histopathological analysis, TNF-α and IL-1β levels, immunohistochemical staining for iNOS, TNF-α, IL-1β, Ki67 and TGF-β RII and a TUNEL assay. The presence and levels of 39 bacterial taxa were analyzed using the Checkerboard DNA-DNA hybridization method. The profiles of NO released from the HPMC/GSNO formulations were characterized using chemiluminescence.. The HPMC/GSNO formulations were found to provide sustained release of NO for more than 4 h at concentration-dependent rates of 14 to 80 nmol/mL/h. Treatment with HPMC/GSNO (0.5 mM) significantly reduced mucosal damage, inflammatory alterations and cell death associated with 5-FU-induced oral mucositis on day 14 but not on day 10. HPMC/GSNO administration also reversed the inhibitory effect of 5-FU on cell proliferation on day 14. In addition, we observed that the chemotherapy significantly increased the levels and/or prevalence of several bacterial species.. Topical HPMC/GSNO accelerates mucosal recovery, reduces inflammatory parameters, speeds up re-epithelization and decreases levels of periodontopathic species in mucosal ulcers.

    Topics: Administration, Topical; Animals; Cricetinae; Disease Models, Animal; Fluorouracil; Gene Expression Regulation, Neoplastic; Humans; Inflammation; Interleukin-1beta; Male; Neoplasms; Nitric Oxide Synthase Type II; S-Nitrosoglutathione; Stomatitis; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2014
IL-6 restores dendritic cell maturation inhibited by tumor-derived TGF-β through interfering Smad 2/3 nuclear translocation.
    Cytokine, 2013, Volume: 62, Issue:3

    We previously found, in a canine transferable tumor model, that high concentration of IL-6 produced by tumor-infiltrating lymphocytes effectively restores the MHC expression of the tumor cells and T-cell activation inhibited by tumor-derived TGF-β. This tumor also significantly suppresses monocyte-derived dendritic cells (DCs) differentiation and the functions of differentiated DCs with unknown mechanisms. In this study, we have demonstrated that a strong reaction of IL-6 was present to neutralize TGF-β-down-regulated surface marker expression on DCs (MHC II, CD1a, CD40, CD80, CD83, CD86), TGF-β-hampered DC functions and DC-associated T-cell activation. Western blotting and confocal microscopy results indicated that the presence of IL-6 markedly decreased the nuclear concentration of a TGF-β signaling transducer, Smad 2/3. In addition, while Smad 7 is a potent molecule inhibiting Smad 2/3 nuclear translocation, no significant increase in Smad 7 gene expression upon addition of IL-6 in TGF-β-pretreated DCs was detected, which suggested that the blockage of Smad 2/3 nuclear translocation by IL-6 did not occur through a Smad 7-inhibitory mechanism. In conclusion, IL-6 inhibited TGF-β signaling and concomitantly antagonized the suppression activities of TGF-β on DC maturation and activity. This study enables further understandings of host/cancer interactions an also provide hints facilitating improvements of DC-based cancer immunotherapy.

    Topics: Animals; Antigens; Biomarkers, Tumor; Cell Differentiation; Cell Nucleus; Dendritic Cells; Dogs; Interleukin-6; Lymphocyte Culture Test, Mixed; Monocytes; Neoplasms; Phenotype; Protein Transport; Signal Transduction; Smad Proteins; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta

2013
Investigating mathematical models of immuno-interactions with early-stage cancer under an agent-based modelling perspective.
    BMC bioinformatics, 2013, Volume: 14 Suppl 6

    Many advances in research regarding immuno-interactions with cancer were developed with the help of ordinary differential equation (ODE) models. These models, however, are not effectively capable of representing problems involving individual localisation, memory and emerging properties, which are common characteristics of cells and molecules of the immune system. Agent-based modelling and simulation is an alternative paradigm to ODE models that overcomes these limitations. In this paper we investigate the potential contribution of agent-based modelling and simulation when compared to ODE modelling and simulation. We seek answers to the following questions: Is it possible to obtain an equivalent agent-based model from the ODE formulation? Do the outcomes differ? Are there any benefits of using one method compared to the other? To answer these questions, we have considered three case studies using established mathematical models of immune interactions with early-stage cancer. These case studies were re-conceptualised under an agent-based perspective and the simulation results were then compared with those from the ODE models. Our results show that it is possible to obtain equivalent agent-based models (i.e. implementing the same mechanisms); the simulation output of both types of models however might differ depending on the attributes of the system to be modelled. In some cases, additional insight from using agent-based modelling was obtained. Overall, we can confirm that agent-based modelling is a useful addition to the tool set of immunologists, as it has extra features that allow for simulations with characteristics that are closer to the biological phenomena.

    Topics: Computer Simulation; Humans; Interleukin-2; Models, Biological; Neoplasms; Transforming Growth Factor beta

2013
Localization, not important in all tumor-suppressing properties: a lesson learnt from scribble.
    Cells, tissues, organs, 2013, Volume: 198, Issue:1

    Aberrant localization of proteins is increasingly being suggested as a causal player in epithelial cancers. Despite this, few studies have investigated how mislocalization of a protein can alter individual biological processes that contribute to cancer progression. Using Ras as a model of transformation, we investigate how localization of the polarity protein Scribble contributes to its tumor-suppressive properties. Wild-type Scribble has been shown to modulate Ras-mitogen-activated protein kinase (MAPK) transformation both in vitro and in vivo. By utilizing a construct that carries a mutation in the LRR domain of Scribble (Scribble P305L) resulting in a cytosolic rather than the usual membrane-bound localization, we report that discrete tumor suppressive properties of Scribble are differentially sensitive to the localization of Scribble. We find that although the Scribble P305L mislocalization mutant can no longer suppress Ras-MAPK-induced invasion or epithelial to mesenchymal transition phenotypes, mislocalized Scribble can still suppress anchorage-independent cell growth. This study illustrates that the manner in which protein mislocalization contributes to cancer is likely complex and highlights the need for careful interrogation as to how cell polarity protein mislocalization, its secondary consequences, and the mutations that give rise to their mislocalization may contribute to specific aspects of cancer progression.

    Topics: Cell Line, Tumor; Cell Proliferation; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Humans; MAP Kinase Signaling System; Membrane Proteins; Mutation; Neoplasms; ras Proteins; Transforming Growth Factor beta; Tumor Suppressor Proteins

2013
Acquired expression of NFATc1 downregulates E-cadherin and promotes cancer cell invasion.
    Cancer research, 2013, Aug-15, Volume: 73, Issue:16

    NFATc1 is a transcription factor that regulates T-cell development, osteoclastogenesis, and macrophage function. Given that T cells, osteoclasts, and macrophages in the tumor microenvironment are thought to modulate tumor progression, tumor cells may acquire NFATc1 expression through fusion with these NFATc1-expressing normal cells. We here revealed that a small proportion of tumor cells in human carcinoma specimens expressed NFATc1. To investigate the consequences of NFATc1 acquisition by tumor cells, we established A549 and MCF7 cell lines expressing a constitutively active form of NFATc1 (NFATc1CA) in an inducible manner. The expression of NFATc1CA promoted cancer cell invasion in association with changes in cell morphology. Analysis of gene expression and RNA interference experiments revealed that NFATc1CA suppressed E-cadherin expression by upregulating the transcriptional repressors Snail and Zeb1 in a manner independent of TGF-β signaling. Induced expression of NFATc1CA also downregulated E-cadherin expression and increased invasive activity in tumor xenografts in vivo. Our results thus suggest that the acquisition of NFATc1 expression contributes to tumor progression.

    Topics: Animals; Antigens, CD; Cadherins; Cell Line, Tumor; Down-Regulation; Female; Gene Expression Regulation, Neoplastic; Heterografts; Homeodomain Proteins; Humans; MCF-7 Cells; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Invasiveness; Neoplasms; NFATC Transcription Factors; Transcription Factors; Transforming Growth Factor beta; Zinc Finger E-box-Binding Homeobox 1

2013
Large-scale integrative network-based analysis identifies common pathways disrupted by copy number alterations across cancers.
    BMC genomics, 2013, Jul-03, Volume: 14

    Many large-scale studies analyzed high-throughput genomic data to identify altered pathways essential to the development and progression of specific types of cancer. However, no previous study has been extended to provide a comprehensive analysis of pathways disrupted by copy number alterations across different human cancers. Towards this goal, we propose a network-based method to integrate copy number alteration data with human protein-protein interaction networks and pathway databases to identify pathways that are commonly disrupted in many different types of cancer.. We applied our approach to a data set of 2,172 cancer patients across 16 different types of cancers, and discovered a set of commonly disrupted pathways, which are likely essential for tumor formation in majority of the cancers. We also identified pathways that are only disrupted in specific cancer types, providing molecular markers for different human cancers. Analysis with independent microarray gene expression datasets confirms that the commonly disrupted pathways can be used to identify patient subgroups with significantly different survival outcomes. We also provide a network view of disrupted pathways to explain how copy number alterations affect pathways that regulate cell growth, cycle, and differentiation for tumorigenesis.. In this work, we demonstrated that the network-based integrative analysis can help to identify pathways disrupted by copy number alterations across 16 types of human cancers, which are not readily identifiable by conventional overrepresentation-based and other pathway-based methods. All the results and source code are available at http://compbio.cs.umn.edu/NetPathID/.

    Topics: Databases, Genetic; DNA Copy Number Variations; Genomics; Humans; Neoplasms; Protein Interaction Maps; Signal Transduction; Survival Analysis; Systems Biology; Transforming Growth Factor beta

2013
The timing of TGF-β inhibition affects the generation of antigen-specific CD8+ T cells.
    BMC immunology, 2013, Jul-17, Volume: 14

    Transforming growth factor (TGF)-β is a potent immunosuppressive cytokine necessary for cancer growth. Animal and human studies have shown that pharmacologic inhibition of TGF-β slows the growth rate of established tumors and occasionally eradicates them altogether. We observed, paradoxically, that inhibiting TGF-β before exposing animals to tumor cells increases tumor growth kinetics. We hypothesized that TGF-β is necessary for the anti-tumor effects of cytotoxic CD8+ T lymphocytes (CTLs) during the early stages of tumor initiation.. BALB/c mice were pretreated with a blocking soluble TGF-β receptor (sTGF-βR, TGF-β-blockade group, n=20) or IgG2a (Control group, n=20) before tumor inoculation. Tumor size was followed for 6 weeks. In vivo lymphocyte assays and depletion experiments were then performed to investigate the immunological basis of our results. Lastly, animals were pretreated with either sTGF-βR (n=6) or IgG2a (n=6) prior to immunization with an adenoviral vector encoding the human papillomavirus E7 gene (Ad.E7). One week later, flow cytometry was utilized to measure the number of splenic E7-specific CD8+ T cells.. Inhibition of TGF-β before the injection of tumor cells resulted in significantly larger average tumor volumes on days 11, 17, 22, 26 and 32 post tumor-inoculation (p < 0.05). This effect was due to the inhibition of CTLs, as it was not present in mice with severe combined immunodeficiency (SCID) or those depleted of CD8+ T cells. Furthermore, pretreatment with sTGF-βR inhibited tumor-specific CTL activity in a Winn Assay. Tumors grew to a much larger size when mixed with CD8+ T cells from mice pretreated with sTGF-βR than when mixed with CD8+ T cells from mice in the control group: 96 mm3 vs. 22.5 mm3, respectively (p < 0.05). In addition, fewer CD8+ T cells were generated in Ad.E7-immunized mice pretreated with sTGF-βR than in mice from the control group: 0.6% total CD8+ T cells vs. 1.9%, respectively (p < 0.05).. These studies provide the first in vivo evidence that TGF-β may be necessary for anti-tumor immune responses in certain cancers. This finding has important implications for our understanding of anti-tumor immune responses, the role of TGF-β in the immune system, and the future development of TGF-β inhibiting drugs.

    Topics: Animals; B7-2 Antigen; CD8-Positive T-Lymphocytes; Cell Count; Cell Line, Tumor; Cell Proliferation; Disease Models, Animal; Epitopes; Female; Histocompatibility Antigens Class I; Histocompatibility Antigens Class II; Humans; Immunization; Immunoglobulin G; Lymph Nodes; Lymphocyte Depletion; Mice; Neoplasm Metastasis; Neoplasms; Papillomavirus E7 Proteins; Receptors, Transforming Growth Factor beta; Signal Transduction; Solubility; T-Lymphocytes, Cytotoxic; Time Factors; Transforming Growth Factor beta

2013
Cadherin 6 is a new RUNX2 target in TGF-β signalling pathway.
    PloS one, 2013, Volume: 8, Issue:9

    Modifications in adhesion molecules profile may change the way tumor cells interact with the surrounding microenvironment. The Cadherin family is a large group of transmembrane proteins that dictate the specificity of the cellular interactions. The Cadherin switch that takes place during epithelial-mesenchymal transition (EMT) contributes to loosening the rigid organization of epithelial tissues and to enhancing motility and invasiveness of tumor cells. Recently, we found Cadherin-6 (CDH6, also known as K-CAD) highly expressed in thyroid tumor cells that display mesenchymal features and aggressive phenotype, following the overexpression of the transcriptional regulator Id1. In this work, we explored the possibility that CDH6 is part of the EMT program in thyroid tumors. We demonstrate that CDH6 is a new transforming growth factor-β (TGF-β) target and that its expression is modulated similarly to other EMT mesenchymal markers, both in vitro and in thyroid tumor patients. We show for the first time that CDH6 is expressed in human thyroid carcinomas and that its expression is enhanced at the invasive front of the tumor. Finally, we show that CDH6 is under the control of the transcription factor RUNX2, which we previously described as a crucial mediator of the Id1 pro-invasive function in thyroid tumor cells. Overall, these observations provide novel information on the mechanism of the EMT program in tumor progression and indicate CDH6 as a potential regulator of invasiveness in thyroid tumors.

    Topics: Alternative Splicing; Cadherins; Carcinoma; Carcinoma, Papillary; Cell Line; Core Binding Factor Alpha 1 Subunit; Epithelial-Mesenchymal Transition; Gene Expression; Gene Expression Regulation; Humans; Models, Biological; Neoplasm Invasiveness; Neoplasms; Phenotype; Signal Transduction; Thyroid Cancer, Papillary; Thyroid Neoplasms; Transforming Growth Factor beta

2013
[BMPs and cancer].
    Zhong nan da xue xue bao. Yi xue ban = Journal of Central South University. Medical sciences, 2013, Volume: 38, Issue:9

    Bone morphogenetic proteins (BMPs) were first studied as growth factors or morphogens of the transforming growth factor-beta super family. These growth molecules, originally associated with bone and cartilage development, are now known to play important roles in morphogenesis and homeostasis in many other tissues. Recently, significant contributions of BMPs, their receptors, and interacting molecules have been linked to carcinogenesis and tumor progression. BMPs can sometimes play a role as a tumor suppressor. This article explains the composition and biological characteristics of BMPs, and investigates their new roles in the pathogenesis of cancer.

    Topics: Bone Morphogenetic Proteins; Humans; Neoplasms; Transforming Growth Factor beta

2013
GRP78 secreted by tumor cells stimulates differentiation of bone marrow mesenchymal stem cells to cancer-associated fibroblasts.
    Biochemical and biophysical research communications, 2013, Nov-01, Volume: 440, Issue:4

    Cancer-associated fibroblasts (CAFs), one type of tumor-associated stromal cells, have been shown to provide a favorable environment for the malignant tumor progression. Extensive reports have demonstrated that mesenchymal stem cells (MSCs) can function as precursors for CAFs. However, the mechanisms by which tumor cells induce the transition of MSCs to CAFs have not been well established. GRP78, traditionally known as an endoplasmic reticulum (ER) chaperone, has been identified to overexpress in a variety of tumor entities and be involved in promoting survival and chemoresistance of tumor cells. Here, we interrogated the role of GRP78 in the generation of CAFs from MSCs. The results showed that GRP78 treatment induced expression of α-smooth muscle actin (α-SMA), a marker for CAFs, in human bone marrow mesenchymal stem cells (HBMSCs) as well as murine bone marrow mesenchymal stem cells (BMMSCs). This phenomenon was correlated with the stimulated phosphorylation of Smad2/3. Furthermore, the GRP78-induced α-SMA expression in HBMSCs was obviously attenuated by SB431542, a TGF-β type I receptor kinase inhibitor. Taken together, the present data suggested that tumor-derived secreted GRP78 elicited the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) to CAFs through activating TGF-β/Smad signaling pathway, which may represent a novel mechanism for transition of BMSCs to CAFs and a hitherto unknown function of GRP78 in the tumor microenvironment.

    Topics: Animals; Bone Marrow Cells; Cell Differentiation; Cell Line, Tumor; Culture Media, Conditioned; Endoplasmic Reticulum Chaperone BiP; Fibroblasts; Heat-Shock Proteins; Humans; Mesenchymal Stem Cells; Mice; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Microenvironment

2013
Twist and miR-34a are involved in the generation of tumor-educated myeloid-derived suppressor cells.
    International journal of molecular sciences, 2013, Oct-14, Volume: 14, Issue:10

    Tumors can induce the generation and accumulation of immunosuppressive cells such as myeloid-derived suppressor cells in the tumor microenvironment, contributing to tumor immunological escapes. Many studies have demonstrated that multiple factors could induce myeloid precursor cells into myeloid-derived suppressor cells, not dendritic cells. In our study, we found that tumor supernatants could induce the generation of myeloid-derived suppressor cells by disturbing the development of dendritic cells. Twist and miR-34a may regulate the effect of tumor cells inducing myeloid-derived suppressor cells via TGF-β and/or IL-10.

    Topics: Animals; Cell Differentiation; Dendritic Cells; Female; Interleukin-10; Male; Mice; Mice, Inbred C57BL; MicroRNAs; Myeloid Cells; Neoplasms; Transforming Growth Factor beta; Twist-Related Protein 1

2013
Meeting report - TGF-β superfamily: signaling in development and disease.
    Journal of cell science, 2013, Nov-01, Volume: 126, Issue:Pt 21

    The latest advances on the transforming growth factor β (TGF-β) and bone morphogenetic protein (BMP) signaling pathways were reported at the July 2013 FASEB Summer Research Conference 'The TGF-β Superfamily: Development and Disease'. The meeting was held in Steamboat Springs, Colorado, USA at 6700 feet above sea level in the Rocky Mountains. This was the seventh biannual meeting in the series. In attendance were investigators from a broad range of disciplines with a common interest in the mechanics of TGF-β and BMP signaling pathways, their normal developmental and homeostatic functions, and the diseases associated with pathway misregulation.

    Topics: Disease; Humans; Multigene Family; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2013
Identification of a pan-cancer oncogenic microRNA superfamily anchored by a central core seed motif.
    Nature communications, 2013, Volume: 4

    MicroRNAs modulate tumorigenesis through suppression of specific genes. As many tumour types rely on overlapping oncogenic pathways, a core set of microRNAs may exist, which consistently drives or suppresses tumorigenesis in many cancer types. Here we integrate The Cancer Genome Atlas (TCGA) pan-cancer data set with a microRNA target atlas composed of publicly available Argonaute Crosslinking Immunoprecipitation (AGO-CLIP) data to identify pan-tumour microRNA drivers of cancer. Through this analysis, we show a pan-cancer, coregulated oncogenic microRNA 'superfamily' consisting of the miR-17, miR-19, miR-130, miR-93, miR-18, miR-455 and miR-210 seed families, which cotargets critical tumour suppressors via a central GUGC core motif. We subsequently define mutations in microRNA target sites using the AGO-CLIP microRNA target atlas and TCGA exome-sequencing data. These combined analyses identify pan-cancer oncogenic cotargeting of the phosphoinositide 3-kinase, TGFβ and p53 pathways by the miR-17-19-130 superfamily members.

    Topics: 3' Untranslated Regions; Algorithms; Amino Acid Motifs; Carcinogenesis; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; HEK293 Cells; Humans; MicroRNAs; Multigene Family; Mutation; Neoplasms; Phosphatidylinositol 3-Kinases; Polymorphism, Single Nucleotide; RNA, Messenger; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2013
Targeting Stat3 and Smad7 to restore TGF-β cytostatic regulation of tumor cells in vitro and in vivo.
    Oncogene, 2013, May-09, Volume: 32, Issue:19

    Transforming Growth Factor-β (TGF-β) and Epidermal Growth Factor (EGF) signaling pathways are both independently implicated as key regulators in tumor formation and progression. Here, we report that the tumor-associated overexpression of epidermal growth factor receptor (EGFR) desensitizes TGF-β signaling and its cytostatic regulation through specific and persistent Stat3 activation and Smad7 induction in vivo. In human tumor cell lines, reduction of TGF-β-mediated Smad2 phosphorylation, nuclear translocation and Smad3 target gene activation were observed when EGFR was overexpressed, but not in cells that expressed EGFR at normal levels. We identified Stat3, which is activated specifically and persistently by overexpressed EGFR, as a key signaling molecule responsible for the reduced TGF-β sensitivity. Stable knockdown of Stat3 using small hairpin RNA(shRNA) in Head and Neck (HN5) and Epidermoid (A431) tumor cell lines resulted in reduced growth compared with control shRNA-transfected cells when grown as subcutaneous tumor xenografts. Furthermore, xenografts with Stat3 knockdown displayed increased Smad3 transcriptional activity, increased Smad2 phosphorylation and decreased Smad7 expression compared with control xenografts in vivo. Consistently, Smad7 mRNA and protein expression was also significantly reduced when EGFR activity was blocked by a specific tyrosine kinase inhibitor, AG1478, or in Stat3 knockdown tumors. Similarly, Smad7 knockdown also resulted in enhanced Smad3 transcriptional activity in vivo. Importantly, there was no uptake of subcutaneous HN5 xenografts with Smad7 knockdown. Taken together, we demonstrate here that targeting Stat3 or Smad7 for knockdown results in resensitization of TGF-β's cytostatic regulation in vivo. Overall, these results establish EGFR/Stat3/Smad7/TGF-β signaling axis driving tumor growth, which can be targeted therapeutically.

    Topics: Animals; Cell Line, Tumor; ErbB Receptors; Female; Gene Knockdown Techniques; HEK293 Cells; Humans; Mice; Mice, Inbred BALB C; Neoplasms; Phosphorylation; RNA, Small Interfering; Signal Transduction; Smad7 Protein; STAT3 Transcription Factor; Transfection; Transforming Growth Factor beta

2013
A human monoclonal antibody 264RAD targeting αvβ6 integrin reduces tumour growth and metastasis, and modulates key biomarkers in vivo.
    Oncogene, 2013, Sep-12, Volume: 32, Issue:37

    αvβ6 integrin expression is upregulated on a wide range of epithelial tumours, and is thought to play a role in modulating tumour growth. Here we describe a human therapeutic antibody 264RAD, which binds and inhibits αvβ6 integrin function. 264RAD cross-reacts with human, mouse and cynomolgus monkey αvβ6, and inhibits binding to all ligands including the latency-associated peptide of TGF-β. Screening across a range of integrins revealed that 264RAD also binds and inhibits the related integrin αvβ8, but not the integrins α5β1, αvβ3, αvβ5 and α4β1. In vitro 264RAD inhibited invasion of VB6 and Detroit 562 cells in a Matrigel invasion assay and αvβ6 mediated production of matrix metalloproteinase-9 in Calu-3 cells. It inhibited TGF-β-mediated activation of dermal skin fibroblasts by preventing local activation of TGF-β by NCI-H358 tumour cells in a tumour cell-fibroblast co-culture assay. In vivo 264RAD showed dose-dependent inhibition of Detroit 562 tumour growth, regressing established tumours when dosed at 20 mg/kg once weekly. The reduction in growth associated with 264RAD was related to a dose-dependent inhibition of Ki67 and phospho-ERK and a reduction of αvβ6 expression in the tumour cells, coupled to a reduction in fibronectin and alpha smooth muscle actin expression in stromal fibroblasts. 264RAD also reduced the growth and metastasis of orthotopic 4T1 tumours. At 20 mg/kg growth of both the primary tumour and the number of metastatic deposits in lung were reduced. The data support the conclusion that 264RAD is a potent inhibitor of αvβ6 integrin, with some activity against αvβ8 integrin, that reduces both tumour growth and metastasis.

    Topics: Animals; Antibodies, Monoclonal, Humanized; Antigens, Neoplasm; Biomarkers; Cell Line; Cell Movement; Cell Proliferation; Coculture Techniques; Female; Humans; Integrins; Lung Neoplasms; Macaca fascicularis; Matrix Metalloproteinase 9; Mice; Neoplasms; Protein Binding; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2013
Regulation of tumor dormancy and role of microenvironment: a mathematical model.
    Advances in experimental medicine and biology, 2013, Volume: 734

    Herein, a mathematical model of a molecular control system for the regulation of secondary tumors is formulated and analyzed to explore how secondary tumors can be controlled by a primary tumor with/without a surgery and the microenvironment. This control system is composed of fibroblast growth factor-2 (FGF2), urokinase-type plasminogen activator (uPA), plasmin, transforming growth factor-beta (TGFβ), latent TGFβ (LTGFβ), and tumor density. The control of secondary tumors by primary tumors was first modeled by Boushaba, Nilsen-Hamiton and Levine in [46]. The model is based on the idea that the vascularization of a secondary tumor can be suppressed by inhibitors from a larger primary tumor. The emergence of tumors at secondary sites 5-7 cm from a primary site was observed after surgical removal of the primary tumor in silico. The model supports the notion that the fate of secondary tumors after surgery depends on the distance from the primary tumor and the surrounding microenvironment. As such, the primary tumor did not influence the growth of remote secondary tumors, but it could effectively suppress the growth of the secondary tumors if they were too close to the primary tumor, even after it was removed. Thus, the model predicts the emergence of secondary tumors after the excision of the primary tumor when the distance between these tumors is in the "distance window." It also predicts that the growth behaviors of the secondary tumors depend on the local microenvironment. Based on these findings, we propose several treatment options for better clinical outcomes.

    Topics: Animals; Antineoplastic Agents; Cell Count; Cell Death; Computer Simulation; Diffusion; Extracellular Matrix; Fibrinolysin; Fibroblast Growth Factor 2; Half-Life; Humans; Models, Biological; Neoplasm Metastasis; Neoplasms; Plasminogen; Time Factors; Transforming Growth Factor beta; Tumor Microenvironment; Urokinase-Type Plasminogen Activator

2013
TGF-β/Smad2/3 signaling directly regulates several miRNAs in mouse ES cells and early embryos.
    PloS one, 2013, Volume: 8, Issue:1

    The Transforming Growth Factor-β (TGF-β) signaling pathway is one of the major pathways essential for normal embryonic development and tissue homeostasis, with anti-tumor but also pro-metastatic properties in cancer. This pathway directly regulates several target genes that mediate its downstream functions, however very few microRNAs (miRNAs) have been identified as targets. miRNAs are modulators of gene expression with essential roles in development and a clear association with diseases including cancer. Little is known about the transcriptional regulation of the primary transcripts (pri-miRNA, pri-miR) from which several mature miRNAs are often derived. Here we present the identification of miRNAs regulated by TGF-β signaling in mouse embryonic stem (ES) cells and early embryos. We used an inducible ES cell system to maintain high levels of the TGF-β activated/phosphorylated Smad2/3 effectors, which are the transcription factors of the pathway, and a specific inhibitor that blocks their activation. By performing short RNA deep-sequencing after 12 hours Smad2/3 activation and after 16 hours inhibition, we generated a database of responsive miRNAs. Promoter/enhancer analysis of a subset of these miRNAs revealed that the transcription of pri-miR-181c/d and the pri-miR-341∼3072 cluster were found to depend on activated Smad2/3. Several of these miRNAs are expressed in early mouse embryos, when the pathway is known to play an essential role. Treatment of embryos with TGF-β inhibitor caused a reduction of their levels confirming that they are targets of this pathway in vivo. Furthermore, we showed that pri-miR-341∼3072 transcription also depends on FoxH1, a known Smad2/3 transcription partner during early development. Together, our data show that miRNAs are regulated directly by the TGF-β/Smad2/3 pathway in ES cells and early embryos. As somatic abnormalities in functions known to be regulated by the TGF-β/Smad2/3 pathway underlie tumor suppression and metastasis, this research also provides a resource for miRNAs involved in cancer.

    Topics: Animals; Cells, Cultured; Embryo, Mammalian; Embryonic Stem Cells; Gene Expression Regulation, Developmental; Mice; MicroRNAs; Multigene Family; Neoplasms; Signal Transduction; Smad2 Protein; Smad3 Protein; Transcription, Genetic; Transforming Growth Factor beta

2013
[TGF-beta signaling as a target of therapy].
    Nihon rinsho. Japanese journal of clinical medicine, 2012, Volume: 70 Suppl 8

    Topics: Animals; Humans; Molecular Targeted Therapy; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2012
Wild type N-ras displays anti-malignant properties, in part by downregulating decorin.
    Journal of cellular physiology, 2012, Volume: 227, Issue:6

    Previously, we have shown that wild type N-ras (wt N-ras) harbors an anti-malignant effect against mutated Ras and in tumors without Ras mutations. To investigate the molecular bases of this anti-malignant activity, we have studied the potency of this anti-malignant effect in a model system against SV40 large T antigen (SV40T). We show that wild-type N-ras (wt N-ras) counteracts the effects of SV40T in NIH3T3 cells as seen by a decrease in proliferation, anchorage independence and changes in migration. We also show that wt N-ras elicits the same anti-malignant effects in some human tumor cell lines (HT1080 and MDA-MB-231). Through mRNA and microRNA (miRNAs) expression profiling we have identified genes (decorin) and miRNAs (mir-29A, let-7b) modulated by wt N-ras potentially responsible for the anti-malignant effect. Wt N-ras appears to mediate its anti-malignant effect by downregulating some of the targets of the TGFβ pathway and decorin, which are able to reverse the inhibition of migration induced by wt N-ras. Our experiments show that the molecules that mediate the anti-malignant effect by wt N-ras appear to be different from those modulated by transforming N-ras. The components of the pathways modulated by wt N-ras mediating its anti-malignant effects are potential targets for therapeutic intervention in cancer.

    Topics: Animals; Antigens, Polyomavirus Transforming; Cell Adhesion; Cell Cycle Checkpoints; Cell Line, Transformed; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Viral; Decorin; Down-Regulation; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Genes, ras; Genotype; Humans; Male; Mice; MicroRNAs; Neoplasms; NIH 3T3 Cells; Phenotype; RNA, Messenger; Signal Transduction; Time Factors; Transfection; Transforming Growth Factor beta

2012
Differential regulation of the two RhoA-specific GEF isoforms Net1/Net1A by TGF-β and miR-24: role in epithelial-to-mesenchymal transition.
    Oncogene, 2012, Jun-07, Volume: 31, Issue:23

    In the present study we analyzed the regulation of the two isoforms of the RhoA-specific guanine nucleotide exchange factor Net1 by transforming growth factor-β (TGF-β) in keratinocytes. We report that short-term TGF-β treatment selectively induced Net1 isoform2 (Net1A) but not Net1 isoform1. This led to upregulation of cytoplasmic Net1A protein levels that were necessary for TGF-β-mediated RhoA activation. Smad signaling and the MAPK/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway were involved in Net1A upregulation by TGF-β. Interestingly, long-term TGF-β treatment resulted in Net1 mRNA downregulation and Net1A protein degradation by the proteasome. Furthermore, we identified the microRNA miR-24 as a novel post-transcriptional regulator of Net1A expression. Silencing of Net1A resulted in disruption of E-cadherin- and zonula occludens-1 (ZO-1)-mediated junctions, as well as expression of the transcriptional repressor of E-cadherin, Slug and the mesenchymal markers N-cadherin, plasminogen activator inhibitor-1 (PAI-1) and fibronectin, indicating that late TGF-β-induced downregulation of Net1A is involved in epithelial-to-mesenchymal transition (EMT). Finally, miR-24 was found to be implicated in the regulation of the EMT program in response to TGF-β and was shown to be directly involved in the TGF-β-induced breast cancer cell invasiveness through Net1A regulation. Our results emphasize the importance of Net1 isoform2 in the short- and long-term TGF-β-mediated regulation of EMT.

    Topics: Cadherins; Cells, Cultured; Epithelial-Mesenchymal Transition; Extracellular Signal-Regulated MAP Kinases; Gene Expression Regulation; Guanine Nucleotide Exchange Factors; Humans; Keratinocytes; Kidney; MicroRNAs; Neoplasms; Oncogene Proteins; Phosphorylation; Protein Isoforms; Proteolysis; rhoA GTP-Binding Protein; RNA Interference; Signal Transduction; Transforming Growth Factor beta

2012
A mathematical model of tumor-immune interactions.
    Journal of theoretical biology, 2012, Feb-07, Volume: 294

    A mathematical model of the interactions between a growing tumor and the immune system is presented. The equations and parameters of the model are based on experimental and clinical results from published studies. The model includes the primary cell populations involved in effector T-cell mediated tumor killing: regulatory T cells, helper T cells, and dendritic cells. A key feature is the inclusion of multiple mechanisms of immunosuppression through the main cytokines and growth factors mediating the interactions between the cell populations. Decreased access of effector cells to the tumor interior with increasing tumor size is accounted for. The model is applied to tumors with different growth rates and antigenicities to gauge the relative importance of various immunosuppressive mechanisms. The most important factors leading to tumor escape are TGF-β-induced immunosuppression, conversion of helper T cells into regulatory T cells, and the limitation of immune cell access to the full tumor at large tumor sizes. The results suggest that for a given tumor growth rate, there is an optimal antigenicity maximizing the response of the immune system. Further increases in antigenicity result in increased immunosuppression, and therefore a decrease in tumor killing rate. This result may have implications for immunotherapies which modulate the effective antigenicity. Simulation of dendritic cell therapy with the model suggests that for some tumors, there is an optimal dose of transfused dendritic cells.

    Topics: Dendritic Cells; Humans; Immune Tolerance; Immunity, Cellular; Interleukin-10; Models, Immunological; Neoplasms; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Escape

2012
A control engineering approach to understanding the TGF-β paradox in cancer.
    Journal of the Royal Society, Interface, 2012, Jun-07, Volume: 9, Issue:71

    TGF-β, a key cytokine that regulates diverse cellular processes, including proliferation and apoptosis, appears to function paradoxically as a tumour suppressor in normal cells, and as a tumour promoter in cancer cells, but the mechanisms underlying such contradictory roles remain unknown. In particular, given that this cytokine is primarily a tumour suppressor, the conundrum of the unusually high level of TGF-β observed in the primary cancer tissue and blood samples of cancer patients with the worst prognosis, remains unresolved. To provide a quantitative explanation of these paradoxical observations, we present, from a control theory perspective, a mechanistic model of TGF-β-driven regulation of cell homeostasis. Analysis of the overall system model yields quantitative insight into how cell population is regulated, enabling us to propose a plausible explanation for the paradox: with the tumour suppressor role of TGF-β unchanged from normal to cancer cells, we demonstrate that the observed increased level of TGF-β is an effect of cancer cell phenotypic progression (specifically, acquired TGF-β resistance), not the cause. We are thus able to explain precisely why the clinically observed correlation between elevated TGF-β levels and poor prognosis is in fact consistent with TGF-β's original (and unchanged) role as a tumour suppressor.

    Topics: Animals; Cell Transformation, Neoplastic; Computer Simulation; Feedback, Physiological; Humans; Models, Biological; Neoplasms; Transforming Growth Factor beta

2012
A taste of TGFβ in Tuscany.
    Development (Cambridge, England), 2012, Volume: 139, Issue:3

    The recent FASEB Summer Research Conference entitled 'The TGFβ Superfamily: Signaling in Development and Disease' was held in August, 2011 in the spectacular setting of Il Ciocco, Lucca, amidst the olive trees in Tuscany, Italy. The organizers assembled an amazing forum, which included 53 speakers and 67 poster presentations from laboratories around the world, to showcase recent advances made in our understanding of the transforming growth factor-β (TGFβ) signaling pathway.

    Topics: Animals; Drosophila; Drosophila Proteins; Epigenomics; Humans; Mice; Morphogenesis; Neoplasms; Protein Processing, Post-Translational; Signal Transduction; Smad Proteins; Transcription Factors; Transforming Growth Factor beta; Wings, Animal

2012
An osteoblast-derived proteinase controls tumor cell survival via TGF-beta activation in the bone microenvironment.
    PloS one, 2012, Volume: 7, Issue:1

    Breast to bone metastases frequently induce a "vicious cycle" in which osteoclast mediated bone resorption and proteolysis results in the release of bone matrix sequestered factors that drive tumor growth. While osteoclasts express numerous proteinases, analysis of human breast to bone metastases unexpectedly revealed that bone forming osteoblasts were consistently positive for the proteinase, MMP-2. Given the role of MMP-2 in extracellular matrix degradation and growth factor/cytokine processing, we tested whether osteoblast derived MMP-2 contributed to the vicious cycle of tumor progression in the bone microenvironment.. To test our hypothesis, we utilized murine models of the osteolytic tumor-bone microenvironment in immunocompetent wild type and MMP-2 null mice. In longitudinal studies, we found that host MMP-2 significantly contributed to tumor progression in bone by protecting against apoptosis and promoting cancer cell survival (caspase-3; immunohistochemistry). Our data also indicate that host MMP-2 contributes to tumor induced osteolysis (μCT, histomorphometry). Further ex vivo/in vitro experiments with wild type and MMP-2 null osteoclast and osteoblast cultures identified that 1) the absence of MMP-2 did not have a deleterious effect on osteoclast function (cd11B isolation, osteoclast differentiation, transwell migration and dentin resorption assay); and 2) that osteoblast derived MMP-2 promoted tumor survival by regulating the bioavailability of TGFβ, a factor critical for cell-cell communication in the bone (ELISA, immunoblot assay, clonal and soft agar assays).. Collectively, these studies identify a novel "mini-vicious cycle" between the osteoblast and metastatic cancer cells that is key for initial tumor survival in the bone microenvironment. In conclusion, the findings of our study suggest that the targeted inhibition of MMP-2 and/or TGFβ would be beneficial for the treatment of bone metastases.

    Topics: Animals; Bone and Bones; Bone Neoplasms; Cell Survival; Cells, Cultured; Cellular Microenvironment; Chlorocebus aethiops; COS Cells; Female; Matrix Metalloproteinase 2; Mice; Mice, Knockout; Models, Biological; Neoplasms; Osteoblasts; Transforming Growth Factor beta; X-Ray Microtomography

2012
Hsp90 inhibition differentially destabilises MAP kinase and TGF-beta signalling components in cancer cells revealed by kinase-targeted chemoproteomics.
    BMC cancer, 2012, Jan-25, Volume: 12

    The heat shock protein 90 (Hsp90) is required for the stability of many signalling kinases. As a target for cancer therapy it allows the simultaneous inhibition of several signalling pathways. However, its inhibition in healthy cells could also lead to severe side effects. This is the first comprehensive analysis of the response to Hsp90 inhibition at the kinome level.. We quantitatively profiled the effects of Hsp90 inhibition by geldanamycin on the kinome of one primary (Hs68) and three tumour cell lines (SW480, U2OS, A549) by affinity proteomics based on immobilized broad spectrum kinase inhibitors ("kinobeads"). To identify affected pathways we used the KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway classification. We combined Hsp90 and proteasome inhibition to identify Hsp90 substrates in Hs68 and SW480 cells. The mutational status of kinases from the used cell lines was determined using next-generation sequencing. A mutation of Hsp90 candidate client RIPK2 was mapped onto its structure.. We measured relative abundances of > 140 protein kinases from the four cell lines in response to geldanamycin treatment and identified many new potential Hsp90 substrates. These kinases represent diverse families and cellular functions, with a strong representation of pathways involved in tumour progression like the BMP, MAPK and TGF-beta signalling cascades. Co-treatment with the proteasome inhibitor MG132 enabled us to classify 64 kinases as true Hsp90 clients. Finally, mutations in 7 kinases correlate with an altered response to Hsp90 inhibition. Structural modelling of the candidate client RIPK2 suggests an impact of the mutation on a proposed Hsp90 binding domain.. We propose a high confidence list of Hsp90 kinase clients, which provides new opportunities for targeted and combinatorial cancer treatment and diagnostic applications.

    Topics: Benzoquinones; Cell Line, Tumor; Enzyme Inhibitors; Extracellular Signal-Regulated MAP Kinases; HSP90 Heat-Shock Proteins; Humans; Lactams, Macrocyclic; Mutation; Neoplasms; Proteomics; Receptor-Interacting Protein Serine-Threonine Kinase 2; Signal Transduction; Transforming Growth Factor beta

2012
Reprogramming of TAM toward proimmunogenic type through regulation of MAP kinases using a redox-active copper chelate.
    Journal of leukocyte biology, 2012, Volume: 91, Issue:4

    TAMs, present in the tumor microenvironment, play an immunosuppressive role, leading to tumor progression and metastasis. Recently, numerous attempts have been made to switch immunosuppressive TAMs into an immunostimulatory type. Previously, we showed that a copper chelate, viz., copper N-(2-hydroxy acetophenone) glycinate [CuNG], can reprogram TAMs toward the proimmunogenic type to mount an antitumor immune response, but the underlying molecular mechanisms of skewing TAMs toward the proimmunogenic type remain elusive. Herein, we tried to explore the signaling mechanisms responsible for the reprogramming of TAMs. We observed that CuNG-induced ROS generation triggers activation of two MAPKs, i.e., p38MAPK and ERK1/2, and also causes up-regulation of intracellular glutathione. Furthermore, activation of p38 MAPK up-regulated the initial IL-12 production and the activation of ERK1/2 in tandem with GSH, found responsible for IFN-γ production by TAMs. This IFN-γ, in turn, prolonged IL-12 production and down-regulated TGF-β production and thus, plays the decisive role in CuNG-mediated reprogramming of regulatory cytokine production by TAMs. Our work highlights that ROS-mediated activation of MAPKs can convert suppressive macrophages toward the proimmunogenic type. Thus, the present study opens the possibility of targeting TAMs by the use of redox-active compounds for designing a novel, therapeutic strategy against cancer.

    Topics: Animals; Cell Line, Tumor; Chelating Agents; Enzyme Activation; Female; Glycine; Interferon-gamma; Interleukin-12; Macrophages; MAP Kinase Signaling System; Mice; Mitogen-Activated Protein Kinase Kinases; Neoplasms; Organometallic Compounds; Reactive Oxygen Species; Transforming Growth Factor beta; Tumor Microenvironment

2012
Elevated expression of angiomodulin (AGM/IGFBP-rP1) in tumor stroma and its roles in fibroblast activation.
    Cancer science, 2012, Volume: 103, Issue:4

    Angiomodulin (AGM/IGFBP-rP1), a glycoprotein of about 30 kDa, is overexpressed in tumor vasculature as well as some human cancer cell lines, but it has been suggested to be a tumor suppressor. To elucidate roles of angiomodulin (AGM) in tumor progression, we here examined distribution of AGM in three types of human cancer tissues by immunohistochemistry. The results showed that AGM was overexpressed in the stroma as well as the vasculature surrounding tumor cells in the human cancer tissues. AGM and α-smooth muscle actin (α-SMA) as an activated fibroblast marker were often colocalized in cancer-associated fibroblasts (CAFs). In vitro analysis indicated that transforming growth factor (TGF)-β1 might be an important inducer of AGM in normal human fibroblasts. AGM strongly stimulated the expression of fibronectin and weakly that of α-SMA in normal fibroblasts. AGM significantly stimulated the proliferation and migration of fibroblasts. The AGM-induced expression of fibronectin and α-SMA was blocked by a TGF-β signal inhibitor but neither the stimulation of cell growth nor migration. These results imply that AGM activates normal fibroblasts by TGF-β-dependent and independent mechanisms. These findings also suggest that AGM and TGF-β1 cooperatively or complementarily contribute to the stromal activation and connective tissue formation in human cancer tissues, contributing to tumor progression.

    Topics: Cell Line; Cell Movement; Cell Proliferation; Disease Progression; Fibroblasts; Humans; Insulin-Like Growth Factor Binding Proteins; Neoplasms; Stromal Cells; Transforming Growth Factor beta

2012
JunB contributes to Id2 repression and the epithelial-mesenchymal transition in response to transforming growth factor-β.
    The Journal of cell biology, 2012, Mar-05, Volume: 196, Issue:5

    The process of epithelial-mesenchymal transition (EMT) in response to transforming growth factor-β (TGF-β) contributes to tissue fibrosis, wound healing, and cancer via a mechanism that is not fully understood. This study identifies a critical role of JunB in the EMT and profibrotic responses to TGF-β. Depletion of JunB by small interfering ribonucleic acid abrogates TGF-β-induced disruption of cell-cell junctions, formation of actin fibers, focal adhesions, and expression of fibrotic proteins. JunB contributes to Smad-mediated repression of inhibitor of differentiation 2 through interaction with transcription repressor activating transcription factor 3. Importantly, JunB mediates the TGF-β induction of profibrotic response factors, fibronectin, fibulin-2, tropomyosin (Tpm1), and integrin-β3, which play critical roles in matrix deposition, cell-matrix adhesion, and actin stress fibers. In summary, JunB provides important input in setting the transcriptional program of the EMT and profibrotic responses to TGF-β. Thus, JunB represents an important target in diseases associated with EMT, including cancer and fibrosis.

    Topics: Animals; Cell Line; Cell-Matrix Junctions; Epithelial Cells; Epithelial-Mesenchymal Transition; Fibrosis; Focal Adhesions; Humans; Inhibitor of Differentiation Protein 2; Kidney; Mammary Glands, Animal; Mice; Neoplasms; Proto-Oncogene Proteins c-jun; Signal Transduction; Smad4 Protein; Stress Fibers; Transforming Growth Factor beta

2012
TGF-β-induced activation of mTOR complex 2 drives epithelial-mesenchymal transition and cell invasion.
    Journal of cell science, 2012, Mar-01, Volume: 125, Issue:Pt 5

    In cancer progression, carcinoma cells gain invasive behavior through a loss of epithelial characteristics and acquisition of mesenchymal properties, a process that can lead to epithelial-mesenchymal transition (EMT). TGF-β is a potent inducer of EMT, and increased TGF-β signaling in cancer cells is thought to drive cancer-associated EMT. Here, we examine the physiological requirement for mTOR complex 2 (mTORC2) in cells undergoing EMT. TGF-β rapidly induces mTORC2 kinase activity in cells undergoing EMT, and controls epithelial cell progression through EMT. By regulating EMT-associated cytoskeletal changes and gene expression, mTORC2 is required for cell migration and invasion. Furthermore, inactivation of mTORC2 prevents cancer cell dissemination in vivo. Our results suggest that the mTORC2 pathway is an essential downstream branch of TGF-β signaling, and represents a responsive target to inhibit EMT and prevent cancer cell invasion and metastasis.

    Topics: Animals; Carcinoma, Squamous Cell; Carrier Proteins; Cell Line, Tumor; Cell Movement; Disease Progression; Epithelial Cells; Epithelial-Mesenchymal Transition; Matrix Metalloproteinase 9; Mice; Neoplasm Invasiveness; Neoplasms; Phosphorylation; Proto-Oncogene Proteins c-akt; Rapamycin-Insensitive Companion of mTOR Protein; rhoA GTP-Binding Protein; RNA Interference; RNA, Messenger; RNA, Small Interfering; Signal Transduction; Snail Family Transcription Factors; TOR Serine-Threonine Kinases; Transcription Factors; Transforming Growth Factor beta

2012
A mathematical model of the enhancement of tumor vaccine efficacy by immunotherapy.
    Bulletin of mathematical biology, 2012, Volume: 74, Issue:7

    TGF-β is an immunoregulatory protein that contributes to inadequate antitumor immune responses in cancer patients. Recent experimental data suggests that TGF-β inhibition alone, provides few clinical benefits, yet it can significantly amplify the anti-tumor immune response when combined with a tumor vaccine. We develop a mathematical model in order to gain insight into the cooperative interaction between anti-TGF-β and vaccine treatments. The mathematical model follows the dynamics of the tumor size, TGF-β concentration, activated cytotoxic effector cells, and regulatory T cells. Using numerical simulations and stability analysis, we study the following scenarios: a control case of no treatment, anti-TGF-β treatment, vaccine treatment, and combined anti-TGF-β vaccine treatments. We show that our model is capable of capturing the observed experimental results, and hence can be potentially used in designing future experiments involving this approach to immunotherapy.

    Topics: Animals; Cancer Vaccines; Computer Simulation; Dendritic Cells; Humans; Immunotherapy; Mice; Models, Biological; Neoplasms; T-Lymphocytes, Cytotoxic; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2012
A positive-margin resection model recreates the postsurgical tumor microenvironment and is a reliable model for adjuvant therapy evaluation.
    Cancer biology & therapy, 2012, Volume: 13, Issue:9

    Up to 30% of cancer patients undergoing curative surgery develop local recurrences due to positive margins. Patients typically receive adjuvant chemotherapy, immunotherapy and/or radiation to prevent such relapses. Interestingly, evidence supporting these therapies is traditionally derived in animal models of primary tumors, thus failing to consider surgically induced tumor microenvironment changes that may influence adjuvant therapy efficacy. To address this consideration, we characterized a murine model of local cancer recurrence. This model was reproducible and generated a postoperative inflammatory tumor microenvironment that resembles those observed following human cancer surgery. To further validate this model, antagonists of two pro-inflammatory mediators, TGFβ and COX-2, were tested and found to be effective in decreasing the growth of recurrent tumors. We appreciated that preoperative TGFβ inhibition led to wound dehiscence, while postoperative initiation of COX-2 inhibition resulted in a loss of efficacy. In summary, although not an exact replica of all human cancer surgeries, our proposed local recurrence approach provides a biologically relevant and reliable model useful for preclinical evaluation of novel adjuvant therapies. The use of this model yields results that may be overlooked using traditional preclinical cancer models that fail to incorporate a surgical component.

    Topics: Animals; Antibodies, Monoclonal; Antineoplastic Agents; Celecoxib; Cell Line, Tumor; Chemotherapy, Adjuvant; Cyclooxygenase 2 Inhibitors; Disease-Free Survival; Female; Humans; Immunity, Innate; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Neoplasm Recurrence, Local; Neoplasms; Postoperative Complications; Pyrazoles; Sulfonamides; Surgical Wound Dehiscence; Transforming Growth Factor beta; Tumor Burden; Tumor Microenvironment; Xenograft Model Antitumor Assays

2012
TGF-β signaling in development and disease.
    FEBS letters, 2012, Jul-04, Volume: 586, Issue:14

    Topics: Animals; Developmental Biology; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Developmental; Humans; Models, Biological; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2012
Roles of vimentin and 14-3-3 zeta/delta in the inhibitory effects of heparin on PC-3M cell proliferation and B16-F10-luc-G5 cells metastasis.
    Acta pharmacologica Sinica, 2012, Volume: 33, Issue:6

    To investigate the inhibitory effects of heparin on PC-3M cells proliferation in vitro and B16-F10-luc-G5 cells metastasis in Balb/c nude mice and identify the protein expression patterns to elucidate the action mechanism of heparin.. Human prostate cancer PC-3M cells were incubated with heparin 0.5 to 125 μg/mL for 24 h. The proliferation of PC-3M cells was assessed by MTS assay. BrdU incoporation and Ki67 expression were detected using a high content screening (HCS) assay. The cell cycle and apoptosis of PC-3M cells were tested by flow cytometry. B16-F10-luc-G5 cardinoma cells were injected into the lateral tail vein of 6-week old male Balb/c nude mice and heparin 30 mg/kg was administered iv 30 min before and 24 h after injection. The metasis of B16-F10-luc-G5 cells was detected by bioluminescence assay. Activated partial thromboplastin time (APTT) and hemorheological parameters were measured on d 14 after injection of B16-F10-luc-G5 carcinoma cells in Balb/c mice. The global protein changes in PC-3M cells and frozen lung tissues from mice burdened with B16-F10-luc-G5 cells were determined by 2-dimensional gel electrophoresis and image analysis. The protein expression of vimentin and 14-3-3 zeta/delta was measured by Western blot. The mRNA transcription of vimentin, transforming growth factor (TGF)-β, E-cadherin, and α(v)-integrin was measured by RT-PCR.. Heparin 25 and 125 μg/mL significantly inhibited the proliferation, arrested the cells in G(1) phase, and suppressed BrdU incorporation and Ki67 expression in PC-3M cells compared with the model group. But it had no significant effect on apoptosis of PC-3M cells. Heparin 30 mg/kg markedly inhibits the metastasis of B16-F10-luc-G5 cells on day 8. Additionally, heparin administration maintained relatively normal red blood hematocrit but had no influence on APTT in nude mice burdened with B16-F10-luc-G5 cells. Thirty of down-regulated protein spots were identified after heparin treatment, many of which are related to tumor development, extracellular signaling, energy metabolism, and cellular proliferation. Vimentin and 14-3-3 zeta/delta were identified in common in PC-3M cells and the lungs of mice bearing B16-F10-luc-G5 carcinoma cells. Heparin 25 and 125 μg/mL decreased the protein expression of vimentin and 14-3-3 zeta/delta and the mRNA expression of α(v)-integrin. Heparin 125 μg/mL decreased vimentin and E-cadherin mRNA transcription while increased TGF-β mRNA transcription in the PC-3M cells, but the differences were not significant. Transfection of vimentin-targeted siRNA for 48 h significantly decreased the BrdU incoporation and Ki67 expression in PC-3M cells.. Heparin inhibited PC-3M cell proliferation in vitro and B16-F10-luc-G5 cells metastasis in nude mice by inhibition of vimentin, 14-3-3 zeta/delta, and α(v)-integrin expression.

    Topics: 14-3-3 Proteins; Animals; Anticoagulants; Antineoplastic Agents; Apoptosis; Cadherins; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Gene Expression Regulation, Neoplastic; Heparin; Humans; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Metastasis; Neoplasms; Prostate; Prostatic Neoplasms; Transforming Growth Factor beta; Vimentin

2012
The human NUPR1/P8 gene is transcriptionally activated by transforming growth factor β via the SMAD signalling pathway.
    The Biochemical journal, 2012, Jul-15, Volume: 445, Issue:2

    NUPR1 (nuclear protein 1), also called P8 (molecular mass 8 kDa) or COM1 (candidate of metastasis 1), is involved in the stress response and in cancer progression. In the present study, we investigated whether human NUPR1 expression was regulated by TGFβ (transforming growth factor β), a secreted polypeptide largely involved in tumorigenesis. We demonstrate that the expression of NUPR1 was activated by TGFβ at the transcriptional level. We show that this activation is mediated by the SMAD proteins, which are transcription factors specifically involved in the signalling of TGFβ superfamily members. NUPR1 promoter analysis reveals the presence of a functional TGFβ-response element binding the SMAD proteins located in the genomic DNA region corresponding to the 5'-UTR (5'-untranslated region). Altogether, the molecular results of the present study, which demonstrate the existence of a TGFβ/SMAD/NUPR1 activation cascade, open the way to consider and investigate further a new mechanism enabling TGFβ to promote tumorigenesis by inducing stress resistance.

    Topics: Animals; Basic Helix-Loop-Helix Transcription Factors; Blotting, Western; Cells, Cultured; Chromatin Immunoprecipitation; Electrophoretic Mobility Shift Assay; Embryo, Mammalian; Fibroblasts; Gene Expression Regulation, Neoplastic; Humans; Mice; Neoplasm Proteins; Neoplasms; Phosphorylation; Promoter Regions, Genetic; Protein Binding; Real-Time Polymerase Chain Reaction; Regulatory Sequences, Nucleic Acid; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; RNA, Small Interfering; Signal Transduction; Smad Proteins; Transcriptional Activation; Transforming Growth Factor beta

2012
Lack of transforming growth factor-β signaling promotes collective cancer cell invasion through tumor-stromal crosstalk.
    Breast cancer research : BCR, 2012, Jul-02, Volume: 14, Issue:4

    Transforming growth factor beta (TGF-β) has a dual role during tumor progression, initially as a suppressor and then as a promoter. Epithelial TGF-β signaling regulates fibroblast recruitment and activation. Concurrently, TGF-β signaling in stromal fibroblasts suppresses tumorigenesis in adjacent epithelia, while its ablation potentiates tumor formation. Much is known about the contribution of TGF-β signaling to tumorigenesis, yet the role of TGF-β in epithelial-stromal migration during tumor progression is poorly understood. We hypothesize that TGF-β is a critical regulator of tumor-stromal interactions that promote mammary tumor cell migration and invasion.. Fluorescently labeled murine mammary carcinoma cells, isolated from either MMTV-PyVmT transforming growth factor-beta receptor II knockout (TβRII KO) or TβRIIfl/fl control mice, were combined with mammary fibroblasts and xenografted onto the chicken embryo chorioallantoic membrane. These combinatorial xenografts were used as a model to study epithelial-stromal crosstalk. Intravital imaging of migration was monitored ex ovo, and metastasis was investigated in ovo. Epithelial RNA from in ovo tumors was isolated by laser capture microdissection and analyzed to identify gene expression changes in response to TGF-β signaling loss.. Intravital microscopy of xenografts revealed that mammary fibroblasts promoted two migratory phenotypes dependent on epithelial TGF-β signaling: single cell/strand migration or collective migration. At epithelial-stromal boundaries, single cell/strand migration of TβRIIfl/fl carcinoma cells was characterized by expression of α-smooth muscle actin and vimentin, while collective migration of TβRII KO carcinoma cells was identified by E-cadherin+/p120+/β-catenin+ clusters. TβRII KO tumors also exhibited a twofold greater metastasis than TβRIIfl/fl tumors, attributed to enhanced extravasation ability. In TβRII KO tumor epithelium compared with TβRIIfl/fl epithelium, Igfbp4 and Tspan13 expression was upregulated while Col1α2, Bmp7, Gng11, Vcan, Tmeff1, and Dsc2 expression was downregulated. Immunoblotting and quantitative PCR analyses on cultured cells validated these targets and correlated Tmeff1 expression with disease progression of TGF-β-insensitive mammary cancer.. Fibroblast-stimulated carcinoma cells utilize TGF-β signaling to drive single cell/strand migration but migrate collectively in the absence of TGF-β signaling. These migration patterns involve the signaling regulation of several epithelial-to-mesenchymal transition pathways. Our findings concerning TGF-β signaling in epithelial-stromal interactions are important in identifying migratory mechanisms that can be targeted as recourse for breast cancer treatment.

    Topics: Animals; beta Catenin; Cadherins; Cell Communication; Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Fibroblasts; Gene Expression Regulation, Neoplastic; Gene Knockout Techniques; Humans; Intercellular Junctions; Mice; Neoplasms; Phenotype; Protein Serine-Threonine Kinases; Protein Transport; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2012
Idiopathic pulmonary fibrosis: an altered fibroblast proliferation linked to cancer biology.
    Proceedings of the American Thoracic Society, 2012, Volume: 9, Issue:3

    The fibrotic process that characterizes idiopathic pulmonary fibrosis (IPF) is commonly considered the result of a recurrent injury to the alveolar epithelium followed by an uncontrolled proliferation of fibroblasts. However, based on considerable scientific evidence, it has been recently hypothesized that IPF might be considered a neoproliferative disorder of the lung because this disease exhibits several pathogenic features similar to cancer. Indeed, epigenetic and genetic abnormalities, altered cell-to-cell communications, uncontrolled proliferation, and abnormal activation of specific signal transduction pathways are biological hallmarks that characterize the pathogenesis of IPF and cancer. IPF remains a disease marked by a survival of 3 years, and little therapeutic progress has been made in the last few years, underlining the urgent need to improve research and to change our approach to the comprehension of this disease. The concept of IPF as a cancer-like disease may be helpful in identifying new pathogenic mechanisms that can be borrowed from cancer biology, potentially leading to different and more effective therapeutic approaches. Such vision will hopefully increase the awareness of this disease among the public and the scientific community.

    Topics: Cell Communication; Cell Proliferation; Epigenesis, Genetic; Epithelial-Mesenchymal Transition; Fibroblasts; Humans; Idiopathic Pulmonary Fibrosis; Mutation; Myofibroblasts; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2012
Lysophosphatidic acid-induced ADAM12 expression mediates human adipose tissue-derived mesenchymal stem cell-stimulated tumor growth.
    The international journal of biochemistry & cell biology, 2012, Volume: 44, Issue:11

    Lysophosphatidic acid (LPA) is involved in mesenchymal stem cell-stimulated tumor growth in vivo. However, the molecular mechanism by which mesenchymal stem cells promote tumorigenesis remains elusive. In the present study, we demonstrate that conditioned medium from A549 human lung adenocarcinoma cells (A549 CM) induced the expression of ADAM12, a disintegrin and metalloproteases family member, in human adipose tissue-derived mesenchymal stem cells (hASCs). A549 CM-stimulated ADAM12 expression was abrogated by pretreatment of hASCs with the LPA receptor 1 inhibitor Ki16425 or by small interfering RNA-mediated silencing of LPA receptor 1, suggesting a key role for the LPA-LPA receptor 1 signaling axis in A549 CM-stimulated ADAM12 expression. Silencing of ADAM12 expression using small interfering RNA or short hairpin RNA abrogated LPA-induced expression of both α-smooth muscle actin, a marker of carcinoma-associated fibroblasts, and ADAM12 in hASCs. Using a xenograft transplantation model of A549 cells, we demonstrated that silencing of ADAM12 inhibited the hASC-stimulated in vivo growth of A549 xenograft tumors and the differentiation of transplanted hASCs to α-smooth muscle actin-positive carcinoma-associated fibroblasts. LPA-conditioned medium from hASCs induced the adhesion of A549 cells and silencing of ADAM12 inhibited LPA-induced expression of extracellular matrix proteins, periostin and βig-h3, in hASCs and LPA-conditioned medium-stimulated adhesion of A549 cells. These results suggest a pivotal role for LPA-stimulated ADAM12 expression in tumor growth and the differentiation of hASCs to carcinoma-associated fibroblasts expressing α-smooth muscle actin, periostin, and βig-h3.

    Topics: Actins; ADAM Proteins; ADAM12 Protein; Adenocarcinoma; Adenocarcinoma of Lung; Adipose Tissue; Animals; Cell Adhesion; Cell Adhesion Molecules; Cell Differentiation; Cell Line, Tumor; Cell Proliferation; Culture Media, Conditioned; Extracellular Matrix Proteins; Fibroblasts; Gene Silencing; Humans; Lung Neoplasms; Lysophospholipids; Membrane Proteins; Mesenchymal Stem Cells; Mice; Mice, Nude; Neoplasms; Receptors, Lysophosphatidic Acid; RNA, Small Interfering; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2012
Metabolic remodeling of the tumor microenvironment: migration stimulating factor (MSF) reprograms myofibroblasts toward lactate production, fueling anabolic tumor growth.
    Cell cycle (Georgetown, Tex.), 2012, Sep-15, Volume: 11, Issue:18

    Migration stimulating factor (MSF) is a genetically truncated N-terminal isoform of fibronectin that is highly expressed during mammalian development in fetal fibroblasts, and during tumor formation in human cancer-associated myofibroblasts. However, its potential functional role in regulating tumor metabolism remains unexplored. Here, we generated an immortalized fibroblast cell line that recombinantly overexpresses MSF and studied their properties relative to vector-alone control fibroblasts. Our results indicate that overexpression of MSF is sufficient to confer myofibroblastic differentiation, likely via increased TGF-b signaling. In addition, MSF activates the inflammation-associated transcription factor NFκB, resulting in the onset of autophagy/mitophagy, thereby driving glycolytic metabolism (L-lactate production) in the tumor microenvironment. Consistent with the idea that glycolytic fibroblasts fuel tumor growth (via L-lactate, a high-energy mitochondrial fuel), MSF fibroblasts significantly increased tumor growth, by up to 4-fold. Mechanistic dissection of the MSF signaling pathway indicated that Cdc42 lies downstream of MSF and fibroblast activation. In accordance with this notion, Cdc42 overexpression in immortalized fibroblasts was sufficient to drive myofibroblast differentiation, to provoke a shift towards glycolytic metabolism and to promote tumor growth by up to 2-fold. In conclusion, the MSF/Cdc42/NFκB signaling cascade may be a critical druggable target in preventing "Warburg-like" cancer metabolism in tumor-associated fibroblasts. Thus, MSF functions in the metabolic remodeling of the tumor microenvironment by metabolically reprogramming cancer-associated fibroblasts toward glycolytic metabolism.

    Topics: Actins; Animals; Autophagy; Biomarkers, Tumor; cdc42 GTP-Binding Protein; Cell Cycle Checkpoints; Cell Hypoxia; Cell Movement; Cell Proliferation; Chemotactic Factors; Cytokines; Fibronectins; Glycolysis; Humans; Lactic Acid; Mice; Mitochondria; Models, Biological; Myofibroblasts; Neoplasms; Neovascularization, Pathologic; NF-kappa B; Phenotype; rac1 GTP-Binding Protein; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2012
Toxicarioside A inhibits tumor growth and angiogenesis: involvement of TGF-β/endoglin signaling.
    PloS one, 2012, Volume: 7, Issue:11

    Toxicarioside A is a cardenolide isolated mainly from plants and animals. Emerging evidence demonstrate that cardenolides not only have cardiac effects but also anticancer effects. In this study, we used in vivo models to investigate the antitumor activities of toxicarioside A and the potential mechanisms behind them. Murine colorectal carcinoma (CT26) and Lewis lung carcinoma (LL/2) models were established in syngeneic BALB/c and C57BL/6 mice, respectively. We found that the optimum effective dose of toxicarioside A treatment significantly suppressed tumor growth and angiogenesis in CT and LL/2 tumor models in vivo. Northern and Western blot analysis showed significant inhibition of endoglin expression in toxicarioside A-treated human umbilical vein endothelial cells (HUVECs) in vitro and tumor tissues in vivo. Toxicarioside A treatment significantly inhibited cell proliferation, migration and invasion, but did not cause significant cell apoptosis and affected other membrane protein (such as CD31 and MHC I) expression. In addition, TGF-β expression was also significantly inhibited in CT26 and LL/2 tumor cells treated with toxicarioside A. Western blot analysis indicated that Smad1 and phosphorylated Smad1 but not Smad2/3 and phosphorylated Smad2/3 were attenuated in HUVECs treated with toxicarioside A. Smad1 and Smad2/3 signaling remained unchanged in CT26 and LL/2 tumor cells treated with toxicarioside A. Endoglin knockout by small interfering RNA against endoglin induced alternations in Smad1 and Smad2/3 signaling in HUVECs. Our results indicate that toxicarioside A suppresses tumor growth through inhibition of endoglin-related tumor angiogenesis, which involves in the endoglin/TGF-β signal pathway.

    Topics: Alginates; Animals; Apoptosis; Cardiac Glycosides; Cell Line, Tumor; Cell Movement; Cell Proliferation; Dose-Response Relationship, Drug; Drug Screening Assays, Antitumor; Endoglin; Endothelial Cells; Female; Glucuronic Acid; Hexuronic Acids; Intracellular Signaling Peptides and Proteins; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Microcirculation; Models, Chemical; Neoplasm Invasiveness; Neoplasm Transplantation; Neoplasms; Neovascularization, Pathologic; Phosphorylation; RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta

2012
Nupr1: the Swiss-knife of cancer.
    Journal of cellular physiology, 2011, Volume: 226, Issue:6

    Nupr1 is a small, highly basic and loosely folded multifunctional protein whose expression is induced by several stresses. Its relation to cancer was first suggested by its overexpression in several human malignancies and the association of its expression with breast cancer metastasis. Accordingly, Nupr1 is structurally related to the high-mobility group (HMG) of transcriptional regulators, which play a key role in the stress response and in cancer progression. Nupr1 interacts with numerous partners to regulate cell cycle, programmed cell death, autophagy, chromatin accessibility, and transcription, and its expression is required for regulation of TGFβ activity. Pleiotropic functions accomplished by Nupr1 depend on its molecular partners, its location into the cell, its expression level and on the cell-type. Nupr1 might be a new drug-targetable protein whose blockade would prevent cancer progression and metastasis development.

    Topics: Animals; Apoptosis; Autophagy; Cell Cycle; Chromatin; Disease Progression; DNA-Binding Proteins; Humans; Neoplasms; Transforming Growth Factor beta

2011
TGFbeta activates mitogen- and stress-activated protein kinase-1 (MSK1) to attenuate cell death.
    The Journal of biological chemistry, 2011, Feb-18, Volume: 286, Issue:7

    Transforming growth factor-β (TGFβ) binding to its receptor leads to intracellular phosphorylation of Smad2 and Smad3, which oligomerize with Smad4. These complexes accumulate in the nucleus and induce gene transcription. Here we describe mitogen- and stress-activated kinase 1 (MSK1) as an antagonist of TGFβ-induced cell death. Induction of MSK1 activity by TGFβ depends on Smad4 and p38 MAPK activation. Knockdown of GADD45, a Smad4-induced upstream regulator of p38 MAPK prevents TGFβ-induced p38 and MSK1 activity. MSK1 functionally regulates pro-apoptotic BH3-only BCL2 proteins, as MSK1 knockdown reduces Bad phosphorylation and enhances Noxa and Bim expression, leading to enhanced TGFβ-induced caspase-3 activity and cell death. This finding suggests that MSK1 represents a pro-survival pathway bifurcating downstream of p38 and antagonizes the established pro-apoptotic p38 MAPK function. Furthermore, EGF could reverse all the effects observed after MSK1 knockdown. Monitoring the status of MSK1 activity in cancer promises new therapeutic targets as inactivating both MSK1 and EGF signaling may (re)-sensitize cells to TGFβ-induced cell death.

    Topics: Animals; Apoptosis Regulatory Proteins; Bcl-2-Like Protein 11; bcl-Associated Death Protein; Cell Cycle Proteins; Cell Death; Cell Line; Cell Survival; Enzyme Activation; Gene Expression Regulation; Gene Knockdown Techniques; Membrane Proteins; Mice; Neoplasms; Nuclear Proteins; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-bcl-2; Ribosomal Protein S6 Kinases, 90-kDa; Smad4 Protein; Transforming Growth Factor beta

2011
TGF-β regulates isoform switching of FGF receptors and epithelial-mesenchymal transition.
    The EMBO journal, 2011, Feb-16, Volume: 30, Issue:4

    The epithelial-mesenchymal transition (EMT) is a crucial event in wound healing, tissue repair, and cancer progression in adult tissues. Here, we demonstrate that transforming growth factor (TGF)-β induced EMT and that long-term exposure to TGF-β elicited the epithelial-myofibroblastic transition (EMyoT) by inactivating the MEK-Erk pathway. During the EMT process, TGF-β induced isoform switching of fibroblast growth factor (FGF) receptors, causing the cells to become sensitive to FGF-2. Addition of FGF-2 to TGF-β-treated cells perturbed EMyoT by reactivating the MEK-Erk pathway and subsequently enhanced EMT through the formation of MEK-Erk-dependent complexes of the transcription factor δEF1/ZEB1 with the transcriptional corepressor CtBP1. Consequently, normal epithelial cells that have undergone EMT as a result of combined TGF-β and FGF-2 stimulation promoted the invasion of cancer cells. Thus, TGF-β and FGF-2 may cooperate with each other and may regulate EMT of various kinds of cells in cancer microenvironment during cancer progression.

    Topics: Actins; Alcohol Oxidoreductases; Alternative Splicing; Cell Differentiation; Cells, Cultured; DNA-Binding Proteins; Epithelial-Mesenchymal Transition; Fibroblast Growth Factor 2; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Models, Biological; Myofibroblasts; Neoplasm Invasiveness; Neoplasms; Protein Binding; Protein Isoforms; Receptors, Fibroblast Growth Factor; Signal Transduction; Transcription Factors; Transforming Growth Factor beta; Zinc Finger E-box-Binding Homeobox 1

2011
Identification of an mRNP complex regulating tumorigenesis at the translational elongation step.
    Molecular cell, 2011, Feb-18, Volume: 41, Issue:4

    Transcript-selective translational regulation of epithelial-mesenchymal transition (EMT) by transforming growth factor-β (TGF-β) is directed by the hnRNP E1-containing TGF-β-activated-translational (BAT) mRNP complex. Herein, eukaryotic elongation factor-1 A1 (eEF1A1) is identified as an integral component of the BAT complex. Translational silencing of Dab2 and ILEI, two EMT transcripts, is mediated by the binding of hnRNP E1 and eEF1A1 to their 3'UTR BAT element, whereby hnRNP E1 stalls translational elongation by inhibiting the release of eEF1A1 from the ribosomal A site. TGF-β-mediated hnRNP E1 phosphorylation, through Akt2, disrupts the BAT complex, thereby restoring translation of target EMT transcripts. Attenuation of hnRNP E1 expression in two noninvasive breast epithelial cells (NMuMG and MCF-7) not only induced EMT but also enabled cells to form metastatic lesions in vivo. Thus, translational regulation by TGF-β at the elongation stage represents a critical checkpoint coordinating the expression of EMT transcripts required during development and in tumorigenesis and metastatic progression.

    Topics: Animals; Cell Line, Tumor; Epithelial-Mesenchymal Transition; Eukaryotic Initiation Factor-1; Mice; Mice, Inbred BALB C; Neoplasms; Peptide Biosynthesis, Nucleic Acid-Independent; Peptide Chain Elongation, Translational; Ribonucleoproteins; Signal Transduction; Transforming Growth Factor beta

2011
InACTIVatINg cancer cachexia.
    Disease models & mechanisms, 2011, Volume: 4, Issue:3

    Topics: Activin Receptors, Type II; Activins; Animals; Cachexia; Disease Models, Animal; Humans; Mice; Muscle, Skeletal; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2011
Cancer susceptibility polymorphism of p53 at codon 72 affects phosphorylation and degradation of p53 protein.
    The Journal of biological chemistry, 2011, May-20, Volume: 286, Issue:20

    The common polymorphism of p53 at codon 72, either encoding proline or arginine, has drawn attention as a genetic factor associated with clinical outcome or cancer risk for the last 2 decades. We now show that these two polymorphic variants differ in protein structure, especially within the N-terminal region and, as a consequence, differ in post-translational modification at the N terminus. The arginine form (p53-72R) shows significantly enhanced phosphorylation at Ser-6 and Ser-20 compared with the proline form (p53-72P). We also show diminished Mdm2-mediated degradation of p53-72R compared with p53-72P, which is at least partly brought about by higher levels of phosphorylation at Ser-20 in p53-72R. Furthermore, enhanced p21 expression in p53-72R-expressing cells, which is dependent on phosphorylation at Ser-6, was demonstrated. Differential p21 expression between the variants was also observed upon activation of TGF-β signaling. Collectively, we demonstrate a novel molecular difference and simultaneously suggest a difference in the tumor-suppressing function of the variants.

    Topics: Cell Line; Codon; Genetic Predisposition to Disease; Humans; Neoplasms; Phosphorylation; Polymorphism, Genetic; Protein Processing, Post-Translational; Proto-Oncogene Proteins c-mdm2; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2011
FBXW7 mutations typically found in human cancers are distinct from null alleles and disrupt lung development.
    The Journal of pathology, 2011, Volume: 224, Issue:2

    FBXW7 is the substrate recognition component of a SCF-type E3 ubiquitin ligase. It has multiple targets such as Notch1, c-Jun, and cyclin E that function in critical developmental and signalling pathways. Mutations in FBXW7 are often found in many types of cancer. In most cases, these mutations do not inactivate the protein, but are mono-allelic missense changes at specific arginine resides involved in substrate binding. We have hypothesized that FBXW7 mutations are selected in cancers for reasons other than haploinsufficiency or full loss-of-function. Given that the existing mutant Fbxw7 mice carry null alleles, we created a mouse model carrying one of the commonly occurring point mutations (Fbxw7(R482Q)) in the WD40 substrate recognition domain of Fbxw7. Mice heterozygous for this mutation apparently developed normally in utero, died perinatally due to a defect in lung development, and in some cases showed cleft palate and eyelid fusion defects. By comparison, Fbxw7(+/-) mice were viable and developed normally. Fbxw7(-/-) animals died of vascular abnormalities at E10.5. We screened known FBXW7 targets for changes in the lungs of the Fbxw7(R482Q/+) mice and found Tgif1 and Klf5 to be up-regulated. Fbxw7(R482Q) alleles are not functionally equivalent to heterozygous or homozygous null alleles, and we propose that they are selected in tumourigenesis because they cause a selective or partial loss of FBXW7 function.

    Topics: Abnormalities, Multiple; Alleles; Amino Acid Sequence; Animals; Arginine; Cell Cycle Proteins; Disease Models, Animal; F-Box Proteins; F-Box-WD Repeat-Containing Protein 7; Humans; Lung; Mice; Molecular Sequence Data; Neoplasm Proteins; Neoplasms; Point Mutation; Pulmonary Alveoli; Sequence Alignment; Transforming Growth Factor beta; Ubiquitin-Protein Ligases

2011
Novel TGF-beta antagonist inhibits tumor growth and angiogenesis by inducing IL-2 receptor-driven STAT1 activation.
    Journal of immunology (Baltimore, Md. : 1950), 2011, Jun-15, Volume: 186, Issue:12

    Carcinoma derived TGF-β acts as a potent pro-oncogenic factor and suppresses antitumor immunity. To antagonize TGF-β-mediated effects in tandem with a proinflammatory immune stimulus, we generated a chimeric protein borne of the fusion of IL-2 and the soluble extracellular domain of TGF-βR II (FIST). FIST acts as a decoy receptor trapping active TGF-β in solution and interacts with IL-2-responsive lymphoid cells, inducing a distinctive hyperactivation of STAT1 downstream of IL-2R, which in turn promotes SMAD7 overexpression. Consequently, FIST-stimulated lymphoid cells are resistant to TGF-β-mediated suppression and produce significant amounts of proinflammatory cytokines. STAT1 hyperactivation further induces significant secretion of angiostatic CXCL10. Moreover, FIST upregulates T-bet expression in NK cells promoting a potent Th1-mediated antitumor response. As a result, FIST stimulation completely inhibits pancreatic cancer (PANC02) and melanoma (B16) tumor growth in immunocompetent C57BL/6 mice. In addition, melanoma cells expressing FIST fail to form tumors in CD8(-/-), CD4(-/-), B cell-deficient (μMT), and beige mice, but not in NOD-SCID and Rag2/γc knockout mice, consistent with the pivotal role of FIST-responsive, cancer-killing NK cells in vivo. In summary, FIST constitutes a novel strategy of treating cancer that targets both the host's angiogenic and innate immune response to malignant cells.

    Topics: Animals; Antineoplastic Agents; Cell Proliferation; Interleukin-2 Receptor beta Subunit; Killer Cells, Natural; Mice; Neoplasms; Neovascularization, Pathologic; Receptors, Interleukin-2; Recombinant Fusion Proteins; STAT1 Transcription Factor; Transforming Growth Factor beta; Tumor Burden

2011
Molecular mechanisms of transforming growth factor-β signaling and disease: The 59th Fujihara International Seminar, 2010.
    Cancer science, 2011, Volume: 102, Issue:6

    The Fujihara International Seminar series is supported by the Fujihara Foundation for Science, for the purpose of organizing seminars for basic and applied science, including medical science, physics, chemistry, engineering, mathematics, geology, and biology. The 59th Fujihara International Seminar was held on July 14-17, 2010 at Tomakomai, Hokkaido, Japan, focusing on molecular mechanisms of transforming growth factor (TGF)-β signaling and disease. Recent findings on mechanisms of TGF-β signaling, the roles of TGF-β signaling in carcinogenesis and progression of tumors, and possible strategies of TGF-β-based treatment of cancer were discussed at the seminar. In particular, novel mechanisms of regulation of Smad signaling, the differential roles of Smad proteins in carcinogenesis, function of Smads in regulation of microRNA biogenesis, and treatment of cancer stem cells by targeting the TGF-β signaling pathways were discussed.

    Topics: Disease Progression; Japan; MicroRNAs; Neoplasms; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
TGF-β as target in oncology.
    Current pharmaceutical biotechnology, 2011, Volume: 12, Issue:12

    Topics: Animals; Humans; Neoplasms; Transforming Growth Factor beta

2011
Acidic preparations of lysed platelets upregulate proliferative pathways in osteoblast-like cells as demonstrated by genome-wide microarray analysis.
    Platelets, 2011, Volume: 22, Issue:6

    Platelets contain numerous growth factors essential for wound and fracture healing. We investigated the gene expression in human osteoblast-like cells stimulated with lysed platelets prepared in acidic, neutral, or alkaline buffers. Lysed platelets prepared in buffers at pH 5.4, 7.4, and 7.9, were added after neutralization to hFOB 1.19 cells. Genome-wide microarray analysis was performed using the Affymetrix GeneChip 7G Scanner. Biometric, cluster, and pathway analyses were performed with GeneSpring GX. Biometric analyses demonstrated that 53 genes were differentially regulated (p ≤ 0.005, ≥2-fold increase). Pathway analysis revealed 10 significant pathways of which eight are common ones regulating bone formation and cancer growth. Eleven genes were selected for quantitative real-time polymerase chain reaction (PCR) based on the microarray analysis of the lysed platelets prepared in the pH 5.4 experiments. In conclusion, acidic preparations of lysed platelet concentrates release factors essential for cell proliferation and particularly cell metabolism under hypoxic conditions. The genetic response from these factors was dominated by genes associated with the same pathways observed in bone formation and cancer growth. Activation of TGF-β in the acidic preparation could be a stimulatory key factor of cell proliferation. These results support the hypothesis that acidification of platelets modifies the stimulatory response of mesenchymal cells in vitro, which is analogous with the observed milieu of a low pH present in wound and fracture sites, as well as in growing tumors.

    Topics: Acids; Blood Platelets; Cell Differentiation; Cell Extracts; Cell Line; Cell Proliferation; Fracture Healing; Gene Expression Profiling; Genome-Wide Association Study; Genome, Human; Humans; Hydrogen-Ion Concentration; Neoplasms; Oligonucleotide Array Sequence Analysis; Osteoblasts; Osteogenesis; Transforming Growth Factor beta

2011
Secretome compartment is a valuable source of biomarkers for cancer-relevant pathways.
    Journal of proteome research, 2011, Sep-02, Volume: 10, Issue:9

    In principle, targeted therapies have optimal activity against a specific subset of tumors that depend upon the targeted molecule or pathway for growth, survival, or metastasis. Consequently, it is important in drug development and clinical practice to have predictive biomarkers that can reliably identify patients who will benefit from a given therapy. We analyzed tumor cell-line secretomes (conditioned cell media) to look for predictive biomarkers; secretomes represent a potential source for potential biomarkers that are expressed in intracellular signaling and therefore may reflect changes induced by targeted therapy. Using Gene Ontology, we classified by function the secretome proteins of 12 tumor cell lines of different histotypes. Representations and hierarchical relationships among the functional groups differed among the cell lines. Using bioinformatics tools, we identified proteins involved in intracellular signaling pathways. For example, we found that secretome proteins related to TGF-beta signaling in thyroid cancer cells, such as vasorin, CD109, and βIG-H3 (TGFBI), were sensitive to RPI-1 and dasatinib treatments, which have been previously demonstrated to be effective in blocking cell proliferation. The secretome may be a valuable source of potential biomarkers for detecting cancer and measuring the effectiveness of cancer therapies.

    Topics: Antineoplastic Agents; Biomarkers, Tumor; Cell Line, Tumor; Cluster Analysis; Computational Biology; Dasatinib; Databases, Protein; Humans; Intracellular Space; Models, Biological; Neoplasm Proteins; Neoplasms; Phosphotyrosine; Proteome; Pyrimidines; Signal Transduction; Thiazoles; Transforming Growth Factor beta

2011
Histone deacetylase inhibitor belinostat represses survivin expression through reactivation of transforming growth factor beta (TGFbeta) receptor II leading to cancer cell death.
    The Journal of biological chemistry, 2011, Sep-02, Volume: 286, Issue:35

    Survivin is a cancer-associated gene that functions to promote cell survival, cell division, and angiogenesis and is a marker of poor prognosis. Histone deacetylase inhibitors induce apoptosis and re-expression of epigenetically silenced tumor suppressor genes in cancer cells. In association with increased expression of the tumor suppressor gene transforming growth factor β receptor II (TGFβRII) induced by the histone deacetylase inhibitor belinostat, we observed repressed survivin expression. We investigated the molecular mechanisms involved in survivin down-regulation by belinostat downstream of reactivation of TGFβ signaling. We identified two mechanisms. At early time points, survivin protein half-life was decreased with its proteasomal degradation. We observed that belinostat activated protein kinase A at early time points in a TGFβ signaling-dependent mechanism. After longer times (48 h), survivin mRNA was also decreased by belinostat. We made the novel observation that belinostat mediated cell death through the TGFβ/protein kinase A signaling pathway. Induction of TGFβRII with concomitant survivin repression may represent a significant mechanism in the anticancer effects of this drug. Therefore, patient populations exhibiting high survivin expression with epigenetically silenced TGFβRII might potentially benefit from the use of this histone deacetylase inhibitor.

    Topics: Catalysis; Cell Death; Cell Line, Tumor; Cyclic AMP-Dependent Protein Kinases; Dose-Response Relationship, Drug; Epigenesis, Genetic; Histone Deacetylase Inhibitors; Humans; Hydroxamic Acids; Inhibitor of Apoptosis Proteins; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Small Interfering; Signal Transduction; Sulfonamides; Survivin; Transforming Growth Factor beta

2011
Atomic force microscopy and high-content analysis: two innovative technologies for dissecting the relationship between epithelial-mesenchymal transition-related morphological and structural alterations and cell mechanical properties.
    Methods in molecular biology (Clifton, N.J.), 2011, Volume: 784

    Epithelial-mesenchymal transition (EMT) is a complex series of cellular reprogramming events culminating in striking alterations in morphology towards an invasive mesenchymal phenotype. Increasingly, evidence suggests that EMT exerts a pivotal role in pathophysiological situations including fibrosis and cancer. Core to these dynamical changes in cellular polarity and plasticity is discrete modifications in cytoskeletal structure. In particular, newly established actin-stress fibres supplant a preceding system of highly organised cortical actin. Although cumulative studies have contributed to elucidation of the detailed signalling pathways that underpin this elaborate molecular process, there remains a deficiency regarding its precise contribution to cellular biomechanics. The advent of atomic force microscopy (AFM) and high-content analysis (HCA) provides two innovative technologies for dissecting the relationship between EMT-related morphological and structural alterations and cell mechanical properties. AFM permits acquisition of high resolution topographical images and detailed analysis of cellular viscoelasticity while HCA facilitates a comprehensive and perspicacious assessment of morphological changes. In combination, they offer the possibility of novel insights into the dynamic traits of transitioning cells. Herein, a detailed protocol describing AFM and HCA techniques for evaluation of transforming growth factor-β1-induced EMT of alveolar epithelial cells is provided.

    Topics: Cell Dedifferentiation; Cell Line, Tumor; Cytoskeleton; Epithelial Cells; Epithelial-Mesenchymal Transition; Fibrosis; Humans; Mesoderm; Microscopy, Atomic Force; Neoplasms; Pulmonary Alveoli; Signal Transduction; Transforming Growth Factor beta

2011
Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity.
    PloS one, 2011, Volume: 6, Issue:9

    Immune evasion is now recognized as a key feature of cancer progression. In animal models, the activity of cytotoxic lymphocytes is suppressed in the tumour microenvironment by the immunosuppressive cytokine, Transforming Growth Factor (TGF)-β. Release from TGF-β-mediated inhibition restores anti-tumour immunity, suggesting a therapeutic strategy for human cancer. We demonstrate that human natural killer (NK) cells are inhibited in a TGF-β dependent manner following chronic contact-dependent interactions with tumour cells in vitro. In vivo, NK cell inhibition was localised to the human tumour microenvironment and primary ovarian tumours conferred TGF-β dependent inhibition upon autologous NK cells ex vivo. TGF-β antagonized the interleukin (IL)-15 induced proliferation and gene expression associated with NK cell activation, inhibiting the expression of both NK cell activation receptor molecules and components of the cytotoxic apparatus. Interleukin-15 also promotes NK cell survival and IL-15 excluded the pro-apoptotic transcription factor FOXO3 from the nucleus. However, this IL-15 mediated pathway was unaffected by TGF-β treatment, allowing NK cell survival. This suggested that NK cells in the tumour microenvironment might have their activity restored by TGF-β blockade and both anti-TGF-β antibodies and a small molecule inhibitor of TGF-β signalling restored the effector function of NK cells inhibited by autologous tumour cells. Thus, TGF-β blunts NK cell activation within the human tumour microenvironment but this evasion mechanism can be therapeutically targeted, boosting anti-tumour immunity.

    Topics: Antineoplastic Agents; Cell Communication; Cell Line, Tumor; Cell Survival; Cytotoxicity, Immunologic; Gene Expression Regulation, Neoplastic; Humans; Interleukin-15; Killer Cells, Natural; Lymphocyte Activation; Models, Immunological; Neoplasms; Transforming Growth Factor beta; Tumor Escape; Tumor Microenvironment

2011
TGF-β down under 2011-the experts interviewed!
    Growth factors (Chur, Switzerland), 2011, Volume: 29, Issue:5

    Topics: Activins; Congresses as Topic; Follistatin; Humans; Inhibins; Neoplasms; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

2011
Inhibitory machinery for the TGF-β family signaling pathway.
    Growth factors (Chur, Switzerland), 2011, Volume: 29, Issue:5

    Transforming growth factor-β (TGF-β) family signaling regulates cell growth and differentiation of many different cell types and is widely involved in the regulation of homeostasis during both embryogenesis and adult life. Therefore, aberrant TGF-β family signal transduction is linked to congenital disorders, tumorigenicity, and fibrosis, which can be life-threatening. A specific receptor-ligand complex initiates transduction of TGF-β family signaling to the nucleus via intracellular signal molecules, mainly Smads, whereby a number of bioactivities such as wound healing, immunomodulation, apoptosis, and angiogenesis are controlled. To avoid an excess of TGF-β family signaling in cells, the duration and intensity of the TGF-β family signal appear to be subject to elaborate regulation. In this paper, we describe recent advances in the understanding of how TGF-β family signals are perturbed and terminated to maintain homeostasis in cells.

    Topics: Apoptosis; Cell Differentiation; Cell Enlargement; Cell Proliferation; Embryonic Development; Humans; Membrane Proteins; MicroRNAs; Neoplasms; Neovascularization, Physiologic; Proteasome Endopeptidase Complex; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
Comments on the cross-talk of TGFβ and EGF in cancer.
    Experimental oncology, 2011, Volume: 33, Issue:3

    Topics: Apoptosis; Cell Differentiation; Cell Proliferation; Cell Survival; Epidermal Growth Factor; Humans; Neoplasm Invasiveness; Neoplasms; Prognosis; Signal Transduction; Transforming Growth Factor beta

2011
Targets of the tumor suppressor miR-200 in regulation of the epithelial-mesenchymal transition in cancer.
    Cancer research, 2011, Dec-15, Volume: 71, Issue:24

    The microRNA-200 (miR-200) family restricts epithelial-mesenchymal transition (EMT) and metastasis in tumor cell lines derived from mice that develop metastatic lung adenocarcinoma. To determine the mechanisms responsible for EMT and metastasis regulated by this microRNA, we conducted a global liquid chromatography/tandem mass spectrometry analysis to compare metastatic and nonmetastatic murine lung adenocarcinoma cells which had undergone EMT because of loss of miR-200. An analysis of syngeneic tumors generated by these cells identified multiple novel proteins linked to metastasis. In particular, the analysis of conditioned media, cell surface proteins, and whole-cell lysates from metastatic and nonmetastatic cells revealed large-scale modifications in the tumor microenvironment. Specific increases were documented in extracellular matrix (ECM) proteins, peptidases, and changes in distribution of cell adhesion proteins in the metastatic cell lines. Integrating proteomic data from three subproteomes, we defined constituents of a multilayer protein network that both regulated and mediated the effects of TGFβ. Lastly, we identified ECM proteins and peptidases that were directly regulated by miR-200. Taken together, our results reveal how expression of miR-200 alters the tumor microenvironment to inhibit the processes of EMT and metastasis.

    Topics: Adenocarcinoma; Animals; Blotting, Western; Carcinoma, Non-Small-Cell Lung; Cell Line, Tumor; Chromatography, Liquid; Epithelial-Mesenchymal Transition; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Gene Regulatory Networks; Genes, Tumor Suppressor; Humans; Lung Neoplasms; Mice; MicroRNAs; Neoplasm Metastasis; Neoplasms; Oligonucleotide Array Sequence Analysis; Proteomics; Signal Transduction; Tandem Mass Spectrometry; Transforming Growth Factor beta; Tumor Microenvironment

2011
Comparative study of the immunohistochemical expression of metalloproteinases 2, 7and 9between clearly invasive carcinomas and "in situ" trophoblast invasion.
    Neoplasma, 2010, Volume: 57, Issue:1

    Matrix metalloproteinases (MMPs) are endopeptidases considered to participate in the transient invasive property of trophoblastic cells during embryo implantation and placentation. The same molecules play an important role in the invasive and metastatic potential of cancer cells. The aim of this study was to compare the immunohistochemical expression of MMP2, 7and 9between clearly invasive carcinomas and "in situ" trophoblast invasion in an effort to illuminate their distinct roles in uncontrolled and controlled invasion.
    We performed an immunohistochemical analysis of 45 clearly invasive carcinomas of various organs (colorectal, gastric, breast, pulmonary, renal) and 40 first trimester gestation specimens (before the 9th week of gestation). The markers expression was evaluated semiquantitavely, seperately in cancer parenchymal and gestational trophoblastic cells as well as cancer stromal and decidual cells, according to apercentage scale (0 %, 50% of cells) and according to staining intensity (0, +, ++, +++).
    MMP9 was expressed more often in the malignant parenchymal as well as in the malignant stromal component of carcinomas than in the trophoblastic (p=0, 0118) and decidual (p=0,017) component of gestations respectively. Although all carcinomas and almost all gestation specimens stained for MMP2 and MMP7, the immunostaining for both molecules was statistically more extensive and intense in trophoblasts and decidual cells by comparison to cancerous elements.
    In conclusion, although there seems to be adirect link between cancer invasion and MMP9 immunohistochemical expression, the role of MMP2 and MMP7 appears to be more complicated underlining the complexity of the mechanisms involved in cancer spreading.

    Topics: Female; Humans; Immunohistochemistry; Matrix Metalloproteinase 2; Matrix Metalloproteinase 7; Matrix Metalloproteinase 9; Metalloproteases; Neoplasms; Pregnancy; Transforming Growth Factor beta; Trophoblasts

2010
VEGFR2 is selectively expressed by FOXP3high CD4+ Treg.
    European journal of immunology, 2010, Volume: 40, Issue:1

    CD25+ FOXP3+CD4+ T cells (Treg) have been considered to play an important role in immune tolerance against several tumor antigens. It has also been indicated that high-level expression of FOXP3 (FOXP3high) is sufficient to confer suppressive activity to normal non-Treg. Here, we showed for the first time that vascular endothelial growth factor receptor 2 (VEGFR2) is selectively expressed by FOXP3high but not FOXP3low Treg. Such VEGFR2+ Treg exist in several tissues including PBMC and malignant effusion-derived lymphocytes. In conclusion, VEGFR2 may be a novel target for controlling Treg with highly suppressive function.

    Topics: Forkhead Transcription Factors; Humans; Immunotherapy; Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Vascular Endothelial Growth Factor Receptor-2

2010
Expression of human beta-defensin-2 in intratumoral vascular endothelium and in endothelial cells induced by transforming growth factor beta.
    Peptides, 2010, Volume: 31, Issue:2

    Human beta-defensin-2 (hBD-2) is a small cationic peptide originally identified from psoriatic skin lesions as an antimicrobial agent of the innate immune system. The expression of hBD-2 is believed to be induced exclusively in epithelial cells by microbial components and certain proinflammatory cytokines, such as interleukin-1 beta (IL-1 beta). In this study, we report, for the first time, that hBD-2 is expressed in vascular endothelial cells associated with oral squamous cell carcinoma (OSCC) and Kaposi's sarcoma lesions, but not in that of normal stroma. Expression of hBD-2 in vascular endothelial cells was further substantiated by in vitro experiments using cultured human umbilical vein endothelial cells (HUVECs). Transforming growth factor beta1 (TGF beta 1) and IL-1 beta, two well-known tumorigenic inflammatory mediators, induce hBD-2 transcript and peptide expression in HUVECs. However, TGF beta 1 does not stimulate hBD-2 expression in oral epithelial cells. In addition, proinflammatory cytokines and microbial reagents do not induce the expression of hBD-1 and hBD-3 in HUVECs. Since hBD-2 has been shown to modulate migration, proliferation, and tube formation of HUVECs in vitro and participate in immune cell trafficking, its expression in vascular endothelial cells located within malignant lesions may play a role in tumor angiogenesis and cancer metastasis.

    Topics: beta-Defensins; Carcinoma, Squamous Cell; Cytoplasm; Endothelial Cells; Endothelium, Vascular; Gene Expression; Humans; Interleukin-1beta; Mouth Neoplasms; Neoplasms; Sarcoma, Kaposi; Transforming Growth Factor beta

2010
Integrative genomic analyses of CXCR4: transcriptional regulation of CXCR4 based on TGFbeta, Nodal, Activin signaling and POU5F1, FOXA2, FOXC2, FOXH1, SOX17, and GFI1 transcription factors.
    International journal of oncology, 2010, Volume: 36, Issue:2

    CXCR4, CD133, CD44 and ABCG2 are representative transmembrane proteins expressed on the surfaces of normal and/or cancer stem cells. CXCR4 is co-expressed with POU5F1 in endodermal precursors and adult-tissue stem cells. CXCR4 is expressed in a variety of human tumors, such as breast cancer, prostate cancer, pancreatic cancer, and gastric cancer. CXCR4 is a G protein-coupled receptor (GPCR) for CXCL12 (SDF1) chemokine, and the CXCL12-CXCR4 signaling axis is involved in proliferation, survival, migration, and homing of cancer cells. Integrative genomic analyses of CXCR4 gene were carried out to elucidate the mechanisms of CXCR4 expression in stem cells, because CXCR4 is a key molecule occupying the crossroads of oncology, immunology, gerontology and regenerative medicine. Human CXCR4 promoter region with binding sites for HIF1alpha, ETS1, NF-kappaB and GLI was not conserved in mouse and rat Cxcr4 orthologs. Proximal enhancer region with palindromic Smad-binding sites, FOX-binding site, POU-binding site, triple SOX17-binding sites, bHLH-binding site, TCF/LEF-binding site, and double GFI1-binding sites was almost completely conserved among human, chimpanzee, mouse, and rat CXCR4 orthologs. TGFbeta, Nodal, and Activin signals induce CXCR4 upregulation based on Smad2/3 and FOX family members, such as FOXA2, FOXC2, and FOXH1. CXCR4 is expressed in endodermal precursors due to the existence of triple SOX17-binding sites around the POU-binding site instead of the POU5F1-SOX2 joint motif. Because CXCR4 is downregulated by p53-GFI1 signaling axis, p53 mutation in cancer stem cells leads to CXCR4 upregulation. CXCR4 is also upregulated by TGFbeta and Hedgehog signals in tumor cells at the invasion front. Small molecule compound or human antibody targeted to CXCR4 will be applied for cancer therapeutics focusing on cancer stem cells at the primary lesion as well as metastasis or recurrence niches, such as bone marrow and peritoneal cavity.

    Topics: Activins; Animals; Base Sequence; Comparative Genomic Hybridization; Conserved Sequence; DNA-Binding Proteins; Forkhead Transcription Factors; Gene Expression; Gene Expression Regulation; Hepatocyte Nuclear Factor 3-beta; Humans; Mice; Molecular Sequence Data; Neoplasms; Nodal Protein; Octamer Transcription Factor-3; Promoter Regions, Genetic; Rats; Receptors, CXCR4; Signal Transduction; SOXF Transcription Factors; Stem Cells; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta

2010
Functional interaction between Smad3 and S100A4 (metastatin-1) for TGF-beta-mediated cancer cell invasiveness.
    The Biochemical journal, 2010, Feb-24, Volume: 426, Issue:3

    TGF-beta (transforming growth factor-beta) induces a cytostatic response in most normal cell types. In cancer cells, however, it often promotes metastasis, and its high expression is correlated with poor prognosis. In the present study, we show that S100A4, a metastasis-associated protein, also called metastatin-1, can physically and functionally interact with Smad3, an important mediator of TGF-beta signalling. In agreement with its known property, S100A4 binds to Smad3 in a Ca2+-dependent manner. The S100A4-binding site is located in the N-terminal region of Smad3. S100A4 can potentiate transcriptional activity of Smad3 and the related Smad2. When exogenously expressed in MCF10CA1a.cl1, an MCF10-derived breast cancer cell line, S100A4 increases TGF-beta-induced MMP-9 (matrix metalloproteinase-9) expression. On the other hand, depletion of S100A4 by siRNA (small interfering RNA) from the MDA-MB231 cell line results in attenuation of MMP-9 induction by TGF-beta. Consistent with these observations, S100A4 increases cell invasion ability induced by TGF-beta in MCF10CA1a.cl1 cells, and depletion of the protein in MDA-MB-231 cells inhibits it. Because expression of both S100A4 and TGF-beta is highly elevated in many types of malignant tumours, S100A4 and Smad3 may co-operatively increase metastatic activity of some types of cancer cells.

    Topics: Binding Sites; Blotting, Western; Calcium; Cell Line; Cell Line, Tumor; Cell Movement; Gene Expression; Humans; Immunoprecipitation; Matrix Metalloproteinase 9; Neoplasm Invasiveness; Neoplasms; Protein Binding; Reverse Transcriptase Polymerase Chain Reaction; RNA, Small Interfering; S100 Calcium-Binding Protein A4; S100 Proteins; Smad3 Protein; Transfection; Transforming Growth Factor beta; Two-Hybrid System Techniques

2010
Processing of CD109 by furin and its role in the regulation of TGF-beta signaling.
    Oncogene, 2010, Apr-15, Volume: 29, Issue:15

    CD109 is a glycosylphosphatidylinositol (GPI)-anchored glycoprotein, whose expression is upregulated in squamous cell carcinomas of the lung, esophagus, uterus and oral cavity. CD109 negatively regulates transforming growth factor (TGF)-beta signaling in keratinocytes by directly modulating receptor activity. In this study, we further characterized CD109 regulation of TGF-beta signaling and cell proliferation. We found that CD109 is produced as a 205 kDa glycoprotein, which is then processed in the Golgi apparatus into 180 kDa and 25 kDa proteins by furin (furinase). 180 kDa CD109 associated with GPI-anchored 25 kDa CD109 on the cell surface and was also secreted into the culture medium. To investigate whether furinase cleavage of CD109 is necessary for its biological activity, we mutated arginine 1273 in the CD109 furinase cleavage motif (amino acid 1270-RRRR-1273) to serine (R1273S). Interestingly, CD109 R1273S neither significantly impaired TGF-beta signaling nor affected TGF-beta-mediated suppression of cell growth, although it was expressed on the cell surface as a 205 kDa protein. Consistent with this finding, the 180 kDa and 25 kDa CD109 complex, but not CD109 R1273S, associated with the type I TGF-beta receptor. These findings indicate that processing of CD109 into 180 kDa and 25 kDa proteins by furin, followed by complex formation with the type I TGF-beta receptor is required for the regulation of TGF-beta signaling in cancer cells and keratinocytes.

    Topics: Animals; Antibodies; Antigens, CD; Cell Line; Cell Membrane; Cell Proliferation; Culture Media; Epitopes; Furin; Glycoproteins; Glycosylation; Golgi Apparatus; GPI-Linked Proteins; Humans; Molecular Weight; Neoplasm Proteins; Neoplasms; Protein Transport; Signal Transduction; Smad Proteins; Solubility; Transforming Growth Factor beta

2010
The TGF-beta superfamily cytokine MIC-1/GDF15: secretory mechanisms facilitate creation of latent stromal stores.
    Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research, 2010, Volume: 30, Issue:6

    Macrophage inhibitory cytokine-1/growth differentiation factor 15 (MIC-1/GDF15), a divergent member of the TGF-beta superfamily is induced by a range of proinflammatory cytokines and oxidized low-density lipoprotein (oxLDL) and is highly expressed in macrophages in atherosclerotic and tumor lesions. MIC-1/GDF15, a major p53 target gene, is largely described to have anti-tumorigenic activity and more recently high MIC-1/GDF15 serum levels in late stage cancer were shown to be the major cause of cancer-associated weight loss. MIC-1/GDF15 serum levels independently predict both atherosclerotic events and severity of rheumatoid arthritis (RA), suggesting serum levels are important in modifying disease expression. Controlling serum levels is the ratio of latent unprocessed MIC-1/GDF15 stromal stores to soluble mature MIC-1/GDF15 generated by the cell. Here, we investigate MIC-1/GDF15 secretion from U937 monocytoid cells and identify novel mechanisms designed to ensure secretion of unprocessed cytokine and creation of latent stromal stores. We find that endogenous MIC-1/GDF15 is secreted as both processed and unprocessed forms. Pulse chase analysis of MIC-1/GDF15 secretion reveals that unprocessed MIC-1/GDF15 precursor is rapidly secreted, while mature MIC-1/GDF15 generated within the cell by intracellular processing is secreted much slower, possibly via an alternate secretory route. The COOH-T 47 amino acids of the propeptide are responsible for rapid secretion of MIC-1/GDF15 precursor and this effect occurs in the trans-Golgi network (TGN)/post TGN compartment. Thus, variations in MIC-1/GDF15 intracellular processing, regulating the presence or absence of propeptide, are a powerful mechanism modulating rate of MIC-1/GDF15 secretion and proMIC-1/GDF15 stromal storage, with major impact on circulating levels of mature MIC-1/GDF15.

    Topics: Arthritis, Rheumatoid; Atherosclerosis; Cell Differentiation; Cell Hypoxia; Cloning, Molecular; Cobalt; Growth Differentiation Factor 15; Humans; Immunization; Lipopolysaccharides; Macrophages; Neoplasms; Protein Processing, Post-Translational; Secretory Pathway; Transforming Growth Factor beta; Transgenes; Tretinoin; U937 Cells

2010
Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-bearing hosts.
    Cellular & molecular immunology, 2010, Volume: 7, Issue:4

    Immune dysfunction is well documented during tumor progression and likely contributes to tumor immune evasion. CD8(+) cytotoxic T lymphocytes (CTLs) are involved in antigen-specific tumor destruction and CD4(+) T cells are essential for helping this CD8(+) T cell-dependent tumor eradication. Tumors often target and inhibit T-cell function to escape from immune surveillance. This dysfunction includes loss of effector and memory T cells, bias towards type 2 cytokines and expansion of T regulatory (Treg) cells. Curcumin has previously been shown to have antitumor activity and some research has addressed the immunoprotective potential of this plant-derived polyphenol in tumor-bearing hosts. Here we examined the role of curcumin in the prevention of tumor-induced dysfunction of T cell-based immune responses. We observed severe loss of both effector and memory T-cell populations, downregulation of type 1 and upregulation of type 2 immune responses and decreased proliferation of effector T cells in the presence of tumors. Curcumin, in turn, prevented this loss of T cells, expanded central memory T cell (T(CM))/effector memory T cell (T(EM)) populations, reversed the type 2 immune bias and attenuated the tumor-induced inhibition of T-cell proliferation in tumor-bearing hosts. Further investigation revealed that tumor burden upregulated Treg cell populations and stimulated the production of the immunosuppressive cytokines transforming growth factor (TGF)-beta and IL-10 in these cells. Curcumin, however, inhibited the suppressive activity of Treg cells by downregulating the production of TGF-beta and IL-10 in these cells. More importantly, curcumin treatment enhanced the ability of effector T cells to kill cancer cells. Overall, our observations suggest that the unique properties of curcumin may be exploited for successful attenuation of tumor-induced suppression of cell-mediated immune responses.

    Topics: Adaptive Immunity; Animals; Antineoplastic Agents; CD4 Antigens; Cell Proliferation; Curcumin; Cytotoxicity, Immunologic; Down-Regulation; Drug Screening Assays, Antitumor; Forkhead Transcription Factors; Immunologic Memory; Interleukin-10; Interleukin-2 Receptor alpha Subunit; Lymphocyte Depletion; Mice; Neoplasms; Survival Analysis; T-Lymphocytes; T-Lymphocytes, Helper-Inducer; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2010
Transient tumor-fibroblast interactions increase tumor cell malignancy by a TGF-Beta mediated mechanism in a mouse xenograft model of breast cancer.
    PloS one, 2010, Mar-23, Volume: 5, Issue:3

    Carcinoma are complex societies of mutually interacting cells in which there is a progressive failure of normal homeostatic mechanisms, causing the parenchymal component to expand inappropriately and ultimately to disseminate to distant sites. When a cancer cell metastasizes, it first will be exposed to cancer associated fibroblasts in the immediate tumor microenvironment and then to normal fibroblasts as it traverses the underlying connective tissue towards the bloodstream. The interaction of tumor cells with stromal fibroblasts influences tumor biology by mechanisms that are not yet fully understood. Here, we report a role for normal stroma fibroblasts in the progression of invasive tumors to metastatic tumors. Using a coculture system of human metastatic breast cancer cells (MCF10CA1a) and normal murine dermal fibroblasts, we found that medium conditioned by cocultures of the two cell types (CoCM) increased migration and scattering of MCF10CA1a cells in vitro, whereas medium conditioned by homotypic cultures had little effect. Transient treatment of MCF10CA1a cells with CoCM in vitro accelerated tumor growth at orthotopic sites in vivo, and resulted in an expanded pattern of metastatic engraftment. The effects of CoCM on MCF10CA1a cells were dependent on small amounts of active TGF-beta1 secreted by fibroblasts under the influence of the tumor cells, and required intact ALK5-, p38-, and JNK signaling in the tumor cells. In conclusion, these results demonstrate that transient interactions between tumor cells and normal fibroblasts can modify the acellular component of the local microenvironment such that it induces long-lasting increases in tumorigenicity and alters the metastatic pattern of the cancer cells in vivo. TGF-beta appears to be a key player in this process, providing further rationale for the development of anti-cancer therapeutics that target the TGF-beta pathway.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Movement; Coculture Techniques; Culture Media, Conditioned; Female; Fibroblasts; Humans; Mice; Mice, Inbred NOD; Mice, SCID; Models, Biological; Neoplasm Metastasis; Neoplasm Transplantation; Neoplasms; Transforming Growth Factor beta

2010
Epithin, a target of transforming growth factor-beta signaling, mediates epithelial-mesenchymal transition.
    Biochemical and biophysical research communications, 2010, May-14, Volume: 395, Issue:4

    The epithelial-derived type II transmembrane serine protease epithin has been shown to be upregulated in a variety of cancer cell lines and tumor tissues, and its upregulation correlates well with tumor progression in many cases. However, little is known regarding the regulation of its expression and the mechanism of its roles in tumor progression. Here, we show that transforming growth factor-beta (TGF-beta), a potent inducer of epithelial-mesenchymal transition (EMT) in tumor progression, upregulates epithin, and that epithin plays a critical role in TGF-beta-induced EMT. Forced overexpression of epithin induced EMT to exhibit characteristic morphological changes, alternations in EMT-related proteins and enhanced cell motility. Conversely, shRNA-mediated knockdown of endogenous epithin inhibited TGF-beta-induced expression of mesenchymal markers and morphological changes. Furthermore, TGF-beta-induced cell migration and invasion were significantly impaired by epithin knockdown. In addition, we demonstrate that TGF-beta upregulates epithin transcriptionally via the Smad2/Smad4-mediated pathway. These results suggest that epithin is a key mediator of TGF-beta-induced EMT in tumor progression.

    Topics: Animals; Cell Transdifferentiation; Dogs; Epithelium; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Membrane Proteins; Mesoderm; Mice; Neoplasms; RNA, Small Interfering; Serine Endopeptidases; Signal Transduction; Smad2 Protein; Smad4 Protein; Transcriptional Activation; Transforming Growth Factor beta; Up-Regulation

2010
Regulation of autophagy by transforming growth factor-β (TGF-β) signaling.
    Autophagy, 2010, Volume: 6, Issue:5

    Transforming growth factor-β (TGF-β) has broad impacts on an array of diverse cellular functions including cell growth, differentiation, adhesion, migration, and apoptosis. Perturbations of the TGF-β signaling pathways are involved in progression of various tumors. Autophagy is a pivotal response of normal and cancer cells to environmental stresses and is induced by various stimuli. Otherwise, autophagy has an intrinsic function in tumor suppression. Recently, we demonstrated that TGF-β induces autophagy in hepatocellular carcinoma cells and mammary carcinoma cells. Autophagy activation by TGF-β is mediated through the Smad and JNK pathways. We show that siRNA-mediated knockdown of autophagy genes suppresses the growth inhibitory function of TGF-β and that autophagy activation potentiates TGF-β-mediated induction of proapoptotic genes, Bim and Bmf, in hepatoma cells. In this context, the autophagy pathway might contribute to the growth inhibitory effect of TGF-β, in conjunction with other anti-proliferative pathways downstream of TGF-β signaling. The context and manner by which the TGF-β signaling pathway regulates autophagy have implications for a better understanding of pathological and bidirectional roles of TGF-β signaling pathways in tumorigenesis.

    Topics: Animals; Apoptosis; Autophagy; Humans; Models, Biological; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2010
Immune regulatory effects of simvastatin on regulatory T cell-mediated tumour immune tolerance.
    Clinical and experimental immunology, 2010, Volume: 161, Issue:2

    Statins are potent inhibitors of hydroxyl-3-methylglutaryl co-enzyme A (HMG-CoA) reductase, and have emerged as potential anti-cancer agents based on preclinical evidence. In particular, compelling evidence suggests that statins have a wide range of immunomodulatory properties. However, little is known about the role of statins in tumour immune tolerance. Tumour immune tolerance involves the production of immunosuppressive molecules, such as interleukin (IL)-10, transforming growth factor (TGF)-beta and indoleamine-2,3-dioxygenase (IDO) by tumours, which induce a regulatory T cell (T(reg)) response. In this study, we investigated the effect of simvastatin on the production of IL-10, TGF-beta and IDO production and the proliferation of T(regs) using several cancer cell lines, and Lewis lung cancer (3LL) cells-inoculated mouse tumour model. Simvastatin treatment resulted in a decrease in the number of cancer cells (3LL, A549 and NCI-H292). The production of the immune regulatory markers IL-10, TGF-beta in 3LL and NCI-H292 cells increased after treatment with simvastatin. The expression of IDO and forkhead box P3 (FoxP3) transcription factor was also increased in the presence of simvastatin. In a murine 3LL model, there were no significant differences in tumour growth rate between untreated and simvastatin-treated mice groups. Therefore, while simvastatin had an anti-proliferative effect, it also exhibited immune tolerance-promoting properties during tumour development. Thus, due to these opposing actions, simvastatin had no net effect on tumour growth.

    Topics: Animals; Cell Count; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Cyclin D1; Cytostatic Agents; Forkhead Transcription Factors; Gene Expression; Humans; Immune Tolerance; Immunosuppressive Agents; Indoleamine-Pyrrole 2,3,-Dioxygenase; Interleukin-10; Interleukin-2 Receptor alpha Subunit; Mice; Mice, Inbred C57BL; Neoplasms; Simvastatin; Spleen; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2010
Na,K-ATPase subunits as markers for epithelial-mesenchymal transition in cancer and fibrosis.
    Molecular cancer therapeutics, 2010, Volume: 9, Issue:6

    Epithelial-to-mesenchymal transition (EMT) is an important developmental process, participates in tissue repair, and occurs during pathologic processes of tumor invasiveness, metastasis, and tissue fibrosis. The molecular mechanisms leading to EMT are poorly understood. Although it is well documented that transforming growth factor (TGF)-beta plays a central role in the induction of EMT, the targets of TGF-beta signaling are poorly defined. We have shown earlier that Na,K-ATPase beta(1)-subunit levels are highly reduced in poorly differentiated kidney carcinoma cells in culture and in patients' tumor samples. In this study, we provide evidence that Na,K-ATPase is a new target of TGF-beta(1)-mediated EMT in renal epithelial cells, a model system used in studies of both cancer progression and fibrosis. We show that following treatment with TGF-beta(1), the surface expression of the beta(1)-subunit of Na,K-ATPase is reduced, before well-characterized EMT markers, and is associated with the acquisition of a mesenchymal phenotype. RNAi-mediated knockdown confirmed the specific involvement of the Na,K-ATPase beta(1)-subunit in the loss of the epithelial phenotype and exogenous overexpression of the Na,K-ATPase beta(1)-subunit attenuated TGF-beta(1)-mediated EMT. We further show that both Na,K-ATPase alpha- and beta-subunit levels are highly reduced in renal fibrotic tissues. These findings reveal for the first time that Na,K-ATPase is a target of TGF-beta(1)-mediated EMT and is associated with the progression of EMT in cancer and fibrosis.

    Topics: Animals; Biomarkers, Tumor; Cell Differentiation; Epithelial Cells; Epithelium; Extracellular Signal-Regulated MAP Kinases; Fibrosis; Gene Knockdown Techniques; Intracellular Space; Kidney Tubules, Proximal; LLC-PK1 Cells; MAP Kinase Signaling System; Mesoderm; Neoplasms; Phenotype; Protein Subunits; Sodium; Sodium-Potassium-Exchanging ATPase; Swine; Transforming Growth Factor beta

2010
Sequential activation of NFAT and c-Myc transcription factors mediates the TGF-beta switch from a suppressor to a promoter of cancer cell proliferation.
    The Journal of biological chemistry, 2010, Aug-27, Volume: 285, Issue:35

    Transforming growth factor beta (TGF-beta) has a dual role in carcinogenesis, acting as a growth inhibitor in early tumor stages and a promoter of cell proliferation in advanced diseases. Although this cellular phenomenon is well established, the underlying molecular mechanisms remain elusive. Here, we report that sequential induction of NFAT and c-Myc transcription factors is sufficient and required for the TGF-beta switch from a cell cycle inhibitor to a growth promoter pathway in cancer cells. Mechanistically, TGF-beta induces in a calcineurin-dependent manner the expression and activation of NFAT factors, which then translocate into the nucleus to promote c-Myc expression. In response to TGF-beta, activated NFAT factors bind to and displace Smad3 repressor complexes from the previously identified TGF-beta inhibitory element (TIE) to transactivate the c-Myc promoter. c-Myc in turn stimulates cell cycle progression and growth through up-regulation of D-type cyclins. Most importantly, NFAT knockdown not only prevents c-Myc activation and cell proliferation, but also partially restores TGF-beta-induced cell cycle arrest and growth suppression. Taken together, this study provides the first evidence for a Smad-independent master regulatory pathway in TGF-beta-promoted cell growth that is defined by sequential transcriptional activation of NFAT and c-Myc factors.

    Topics: Animals; Calcineurin; Cell Cycle; Cell Line, Tumor; Humans; Mice; Neoplasms; NFATC Transcription Factors; Proto-Oncogene Proteins c-myc; Smad Proteins; Transcription, Genetic; Transcriptional Activation; Transforming Growth Factor beta

2010
Trigger found that may start cancer's spread. Research may lead to new therapies to halt cancer's progression.
    DukeMedicine healthnews, 2010, Volume: 16, Issue:5

    Topics: Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Animals; Apoptosis Regulatory Proteins; Cell Line, Transformed; Cell Transdifferentiation; Disease Progression; Epithelial Cells; Gene Expression Regulation, Neoplastic; Humans; Mice; Neoplasm Metastasis; Neoplasm Proteins; Neoplasms; Proto-Oncogene Proteins c-akt; Transforming Growth Factor beta

2010
Reduced transforming growth factor-beta receptor II expression in hepatocellular carcinoma correlates with intrahepatic metastasis.
    Laboratory investigation; a journal of technical methods and pathology, 2010, Volume: 90, Issue:9

    Hepatocellular carcinoma (HCC) occurs mainly in the liver associated with chronic hepatitis and hepatic cirrhosis as a result of prolonged viral infection. Transforming growth factor-beta (TGF-beta) induces the fibrosis in hepatic cirrhosis, although it is also an inhibitor of hepatocyte proliferation. To understand the role of TGF-beta signaling in HCC progression, we analyzed gene expression in HCC cells in relation to TGF-beta signaling using a two-way clustering algorithm. By the analysis, five HCC cell lines were classified into two groups according to their metastatic capacity. TGF-beta receptor II (TGFBR2) was downregulated in metastatic cells, which did not show a response to TGF-beta. Immunohistochemistry demonstrated clear membrane distribution of TGFBR2 in noncancerous hepatocytes, whereas reduced TGFBR2 expression was observed in 34 of 136 HCCs. In clinical cases, reduced TGFBR2 expression correlated with larger tumor size (P<0.001), poor differentiation (P<0.001), portal vein invasion (P=0.002), intrahepatic metastasis (IM) (P<0.001), and shorter recurrence-free survival (P=0.022). In conclusion, reduced TGFBR2 expression was associated with aggressive features of HCC such as IM, and may represent an immunohistochemical biomarker to detect aggressive HCC.

    Topics: Aged; Carcinoma, Hepatocellular; Down-Regulation; Female; Hepatitis, Chronic; Hepatocytes; Humans; Immunohistochemistry; Liver; Liver Cirrhosis; Liver Neoplasms; Male; Middle Aged; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2010
SnoN: bridging neurobiology and cancer biology.
    Current molecular medicine, 2010, Volume: 10, Issue:7

    The transcriptional regulator SnoN has been the subject of growing interest due to its diverse functions in normal and pathological settings. A large body of evidence has established a fundamental role for SnoN as a modulator of signaling and responses by the transforming growth beta (TGFbeta) family of cytokines, though how SnoN regulates TGFbeta responses remains incompletely understood. In accordance with the critical and complex roles of TGFbeta in tumorigenesis and metastasis, SnoN may act as a tumor promoter or suppressor depending on the stage and type of cancer. Beyond its role in cancer, SnoN has also been implicated in the control of axon morphogenesis in postmitotic neurons in the mammalian brain. Remarkably, signaling pathways that control SnoN functions in the divergent cycling cells and postmitotic neurons appear to be conserved. Identification of novel SnoN regulatory and effector mechanisms holds the promise of advances at the interface of cancer biology and neurobiology.

    Topics: Animals; Cell Differentiation; Cytokines; Gene Expression Regulation; Homeodomain Proteins; Humans; Intracellular Signaling Peptides and Proteins; Morphogenesis; Neoplasms; Neurons; Oncogenes; Proto-Oncogene Proteins; Receptors, Cytoplasmic and Nuclear; Signal Transduction; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Tumor Suppressor Proteins

2010
The roles of antigen-specificity, responsiveness to transforming growth factor-β and antigen-presenting cell subsets in tumour-induced expansion of regulatory T cells.
    Immunology, 2010, Volume: 131, Issue:4

    In this study we investigated the impact of several factors on the expansion of natural regulatory T (nTreg) cells by tumours, including antigen specificity, transforming growth factor-β (TGF-β) signalling and the antigen-presenting cell subsets responsible for expansion. We found that antigen non-specific expansion of nTreg cells is tumour cell line-dependent. Although both antigen-specific and non-specific pathways can contribute to expansion, the migration of activated nTreg cells to tumour tissues is strictly antigen-dependent. Intact TGF-β signalling on nTreg cells is also essential for tumour-induced expansion. Finally, for stimulation of resting antigen-specific CD4 T cells, CD11c(+) cells purified from tumour-draining lymph nodes were more potent than CD11b(+) cells, suggesting that dendritic cells are the key antigen-presenting cell subset involved in cross-presentation of tumour antigens. This study not only provides an in vivo system in which cross-talk between nTreg cells and tumours can be explored but also reveals novel aspects of tumour immune evasion.

    Topics: Animals; Antigen-Presenting Cells; Antigens, Neoplasm; CD11c Antigen; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cross-Priming; Lymph Nodes; Mice; Mice, Knockout; Neoplasms; Signal Transduction; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Escape

2010
NFAT transcription factors, the potion mediating "Dr. Jekill-Mr. Hyde" transformation of the TGFβ pathway in cancer cells.
    Cell cycle (Georgetown, Tex.), 2010, Oct-01, Volume: 9, Issue:19

    Topics: Cell Transformation, Neoplastic; Gene Expression Regulation; Neoplasms; NFATC Transcription Factors; Signal Transduction; Transforming Growth Factor beta

2010
Transforming growth factor β signaling inhibitor, SB-431542, induces maturation of dendritic cells and enhances anti-tumor activity.
    Oncology reports, 2010, Volume: 24, Issue:6

    The transforming growth factor β (TGFβ) stimulates tumor progression and metastasis. Secretion of TGFβ by tumor cells also suppresses an antitumor immune response in which dendritic cells (DCs) play an important role to activate cytotoxic T lymphocytes (CTLs). Herein we report that the small molecule TGFβ signaling inhibitor SB-431542, induces DC maturation in vitro and triggers antitumor activity in vivo. We added SB-431542 to cultures of murine bone-marrow derived DCs (BM-DCs) derived from BALB/c mice and human DCs generated from peripheral monocytes (human DCs) at different concentrations in triplicates and examined expression of co-stimulatory molecules by FACS and production of Interleukin-12 (IL-12) by ELISA. SB induced phenotypic maturation of BM-DCs and human DCs and improved their abilities to produce IL-12 in a dose-dependent manner. SB-431542 also augmented capacity of murine and human DCs to activate naive T cells in allogeneic mixed lymphocyte reaction. Interestingly, SB-431542 augmented the capacity of BM-DCs and human DCs to incorporate FITC-conjugated dextran. Intraperitoneal administration of SB-431542 initiated 3 and 7 days after the implantation of colon-26 cancer cells into the peritoneal cavity of BALB/c mice significantly induced CTL activity against colon-26. We incubated human DCs with SB-431542 and cell lysate of scirrhous gastric cancer cell line OCUM-8, and then examined CTL activities against OCUM-8. CD8 T cells activated by human DCs treated with SB-431542 showed modest augmentation CTL activity against cancer cells. Furthermore, pretreatment of human DCs with SB-431542 upregulated cytotoxic activity against K562 cells, suggesting SB should have potential to activate DCs to natural killer cells. In conclusion, TGFβ receptor I kinase inhibitor such as SB-431542 might induce anti-tumor immune response in immuno-tolerant patients associated with TGFβ activity.

    Topics: Animals; Antineoplastic Agents; Benzamides; Cell Differentiation; Cells, Cultured; Combined Modality Therapy; Dendritic Cells; Dioxoles; Female; Humans; Immunotherapy, Active; K562 Cells; Mice; Mice, Inbred BALB C; Neoplasms; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2010
Transforming growth factor-β signaling and cancer: the 28th Sapporo Cancer Seminar, 25-27 June 2008.
    Cancer science, 2009, Volume: 100, Issue:2

    The Sapporo Cancer Seminars have been held annually since 1981. The 28th Sapporo Cancer Seminar was held on 26-27 June 2008 at the Hokkaido University Conference Hall, focusing on transforming growth factor (TGF)-β signaling and cancer. More than 150 scientists participated in the seminar, and it provided a great deal of information on the role of TGF-β signaling in carcinogenesis and tumor metastasis. The possible use of TGF-β antagonists for treatment of cancers was also discussed at the seminar.

    Topics: Animals; Humans; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2009
Transforming growth factor-beta signaling in skin: stromal to epithelial cross-talk.
    The Journal of investigative dermatology, 2009, Volume: 129, Issue:1

    In this issue, Denton et al., describe a mouse model of postnatal deletion of the transforming growth factor (TGF)-beta receptor type II (TbetaRII) in skin fibroblasts. Using a tamoxifen-dependent inducible Cre-lox strategy, the authors demonstrate the pivotal role played by TGF-beta signaling in fibroblasts during wound healing. Healing of full-thickness wounds after fibroblast-specific deletion of TbetaRII in the skin was severely impaired and exhibited delayed re-epithelialization. This study emphasizes the importance of fibroblasts in mesenchymal-epithelial interaction in the skin during wound repair.

    Topics: Animals; Epithelium; Fibroblasts; Gene Deletion; Humans; Mice; Models, Biological; Neoplasms; Signal Transduction; Skin; Stromal Cells; Transforming Growth Factor beta; Wound Healing

2009
CD40 ligation converts TGF-beta-secreting tolerogenic CD4-8- dendritic cells into IL-12-secreting immunogenic ones.
    Biochemical and biophysical research communications, 2009, Feb-20, Volume: 379, Issue:4

    CD40L, the ligand for CD40 on dendritic cells (DCs), plays an important role in maturation and activation of DCs leading to induction of immune responses. Our previous studies showed that the mouse splenic CD4(-)8(-) DCs are tolerogenic and capable of stimulating suppressive type 1 CD4(+) regulatory T (Tr1) cell responses via TGF-beta secretion. In this study, we investigated whether CD40 ligation is able to convert tolerogenic CD4(-)8(-) DCs into immunogenic ones by in vitro treatment of DCs with anti-CD40 antibody. Our data showed that in vitro CD40 ligation with anti-CD40 antibody converted TGF-beta-secreting tolerogenic CD4(-)8(-) DCs into IL-12-secreting immunogenic ones capable of stimulating type 1 CD4(+) helper T (Th1) and CD8(+) cytotoxic T lymphocyte (CTL) responses leading to induction of antitumor immunity. In addition, in vivo CD40 ligation by intratumoral injection of adenoviral vector AdVCD40L expressing CD40 ligand also induced tumor growth inhibition and regression of established P815 tumors with infiltration of tolerogenic CD4(-)8(-) DCs. Therefore, our data provide new information for and may thus have useful impacts in CD40 ligation-based immunotherapy of cancer.

    Topics: Animals; CD4 Antigens; CD40 Antigens; CD40 Ligand; CD8 Antigens; Dendritic Cells; Immune Tolerance; Interleukin-12; Mice; Mice, Inbred C57BL; Neoplasms; T-Lymphocytes; T-Lymphocytes, Cytotoxic; Th1 Cells; Transforming Growth Factor beta; Up-Regulation

2009
Transforming growth factor beta depletion is the primary determinant of Smad signaling kinetics.
    Molecular and cellular biology, 2009, Volume: 29, Issue:9

    A cell's decision to growth arrest, apoptose, or differentiate in response to transforming growth factor beta (TGF-beta) superfamily ligands depends on the ligand concentration. How cells sense the concentration of extracellular bioavailable TGF-beta remains poorly understood. We therefore undertook a systematic quantitative analysis of how TGF-beta ligand concentration is transduced into downstream phospho-Smad2 kinetics, and we found that the rate of TGF-beta ligand depletion is the principal determinant of Smad signal duration. TGF-beta depletion is caused by two mechanisms: (i) cellular uptake of TGF-beta by a TGF-beta type II receptor-dependent mechanism and (ii) reversible binding of TGF-beta to the cell surface. Our results indicate that cells sense TGF-beta dose by depleting TGF-beta via constitutive TGF-beta type II receptor trafficking processes. Our results also have implications for the role of the TGF-beta type II receptor in disease, as tumor cells harboring TGF-beta type II receptor mutations exhibit impaired TGF-beta depletion, which may contribute to the overproduction of TGF-beta and a consequently poor prognosis in cancer.

    Topics: Animals; Cell Line; Culture Media; Humans; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad2 Protein; Transforming Growth Factor beta

2009
Hepatocyte growth factor activator inhibitor type 1 regulates epithelial to mesenchymal transition through membrane-bound serine proteinases.
    Cancer research, 2009, Mar-01, Volume: 69, Issue:5

    Hepatocyte growth factor activator inhibitor-1 (HAI-1), encoded by the serine protease inhibitor Kunitz type 1 (SPINT1) gene, is a membrane-associated proteinase inhibitor that potently inhibits a variety of serine proteinases, including those that are membrane bound. Although HAI-1/SPINT1 is widely expressed by epithelial cells and cancer cells, its functional role is still unclear, particularly in cancer. Here, we show that stable knockdown of HAI-1/SPINT1 in the human pancreatic cancer cell line SUIT-2 induces an elongated spindle-like morphology associated with accelerated invasion, thereby mimicking an epithelial to mesenchymal transition (EMT). We found that HAI-1/SPINT1 knockdown significantly reduced the expression of E-cadherin and was accompanied by up-regulation of Smad-interacting protein 1 (SIP1), an E-cadherin transcriptional repressor. In addition, matrix metalloproteinase-9 (MMP-9) was up-regulated. Similar results were obtained in the HLC-1 lung carcinoma cell line. Moreover, a metastatic variant of SUIT-2 (S2-CP8) that showed loss of E-cadherin expression also showed a significantly reduced level of HAI-1/SPINT1. Engineered overexpression of HAI-1/SPINT1 in S2-CP8 resulted in reversion of E-cadherin expression and SIP1 down-regulation, which accompanied reestablishment of epithelial morphology in culture. The EMT caused by HAI-1/SPINT1 knockdown seemed to be mediated, at least partly, by membrane-bound serine proteinases, matriptase/ST14 and TMPRSS4, as knockdown of matriptase/ST14 or TMPRSS4 in HAI-1/SPINT1 knockdown SUIT-2 cells and HLC-1 cells resulted in reversion of SIP1 and/or MMP-9 expression levels. We suggest that interactions between HAI-1/SPINT1 and membrane-bound serine proteinases contribute to transcriptional and functional changes involved in EMT in certain carcinoma cells.

    Topics: Animals; Cadherins; Cell Line, Tumor; Epithelial Cells; Humans; Male; Mesoderm; Mice; Mice, Inbred BALB C; Neoplasms; Nerve Tissue Proteins; Pancreatic Neoplasms; Proteinase Inhibitory Proteins, Secretory; RNA-Binding Proteins; Serine Endopeptidases; Transforming Growth Factor beta

2009
Induction of myeloid-derived suppressor cells by tumor exosomes.
    International journal of cancer, 2009, Jun-01, Volume: 124, Issue:11

    Myeloid-derived suppressor cells (MDSCs) promote tumor progression. The mechanisms of MDSC development during tumor growth remain unknown. Tumor exosomes (T-exosomes) have been implicated to play a role in immune regulation, however the role of exosomes in the induction of MDSCs is unclear. Our previous work demonstrated that exosomes isolated from tumor cells are taken up by bone marrow myeloid cells. Here, we extend those findings showing that exosomes isolated from T-exosomes switch the differentiation pathway of these myeloid cells to the MDSC pathway (CD11b(+)Gr-1(+)). The resulting cells exhibit MDSC phenotypic and functional characteristics including promotion of tumor growth. Furthermore, we demonstrated that in vivo MDSC mediated promotion of tumor progression is dependent on T-exosome prostaglandin E2 (PGE2) and TGF-beta molecules. T-exosomes can induce the accumulation of MDSCs expressing Cox2, IL-6, VEGF, and arginase-1. Antibodies against exosomal PGE2 and TGF-beta block the activity of these exosomes on MDSC induction and therefore attenuate MDSC-mediated tumor-promoting ability. Exosomal PGE2 and TGF-beta are enriched in T-exosomes when compared with exosomes isolated from the supernatants of cultured tumor cells (C-exosomes). The tumor microenvironment has an effect on the potency of T-exosome mediated induction of MDSCs by regulating the sorting and the amount of exosomal PGE2 and TGF-beta available. Together, these findings lend themselves to developing specific targetable therapeutic strategies to reduce or eliminate MDSC-induced immunosuppression and hence enhance host antitumor immunotherapy efficacy.

    Topics: Animals; CD11b Antigen; Cell Line, Tumor; Dinoprostone; Exosomes; Female; Mice; Mice, Inbred BALB C; Myeloid Cells; Neoplasms; Receptors, Chemokine; Transforming Growth Factor beta

2009
Quantitative modeling and analysis of the transforming growth factor beta signaling pathway.
    Biophysical journal, 2009, Mar-04, Volume: 96, Issue:5

    Transforming growth factor beta (TGF-beta) signaling, which regulates multiple cellular processes including proliferation, apoptosis, and differentiation, plays an important but incompletely understood role in normal and cancerous tissues. For instance, although TGF-beta functions as a tumor suppressor in the premalignant stages of tumorigenesis, paradoxically, it also seems to act as a tumor promoter in advanced cancer leading to metastasis. The mechanisms by which TGF-beta elicits such diverse responses during cancer progression are still not entirely clear. As a first step toward understanding TGF-beta signaling quantitatively, we have developed a comprehensive, dynamic model of the canonical TGF-beta pathway via Smad transcription factors. By describing how an extracellular signal of the TGF-beta ligand is sensed by receptors and transmitted into the nucleus through intracellular Smad proteins, the model provides quantitative insight into how TGF-beta-induced responses are modulated and regulated. Subsequent model analysis shows that mechanisms associated with Smad activation by ligand-activated receptor, nuclear complex formation among Smad proteins, and inactivation of ligand-activated Smad (e.g., degradation, dephosphorylation) may be critical for regulating TGF-beta-targeted functional responses. The model was also used to predict dynamic characteristics of the Smad-mediated pathway in abnormal cells, from which we generated four testable hypotheses regarding potential mechanisms by which TGF-beta's tumor-suppressive roles may appear to morph into tumor-promotion during cancer progression.

    Topics: Active Transport, Cell Nucleus; Algorithms; Cell Nucleus; Gene Expression; Least-Squares Analysis; Ligands; Models, Biological; Mutation; Neoplasms; Phosphorylation; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2009
Gamma irradiation alters the phenotype and function of CD4+CD25+ regulatory T cells.
    Cell biology international, 2009, Volume: 33, Issue:5

    To examine the effects of gamma irradiation on Tregs, changes in phenotype and suppression function in Tregs treated with or without gamma ray were analyzed. Purified CD4(+)CD25(+) regulatory T cells were irradiated at different dosages with a (137)Cs source gamma ray at 4.8 Gy/min. After culture, the phenotype and function changes were determined by flow cytometry and [(3)H]-thymidine incorporation, respectively. A dose-dependent reduction of Tregs proliferation in response to gamma irradiation was noted, which paralleled the apoptosis induction of Tregs. Gamma irradiation downregulated the Tregs expression of CD45RO, CD62L, FOXP3, membrane TGF-beta, but upregulated Bax and GITR. High dose gamma irradiation (30 Gy) significantly abolished the suppression of Tregs on CD4(+)CD25(-) T cells proliferation. Thus Tregs not only influences the phenotype but also alters their suppressive capacities. Our findings suggest that radiotherapy may be an important strategy to alter the immunologic balance of Tregs and effector cells in cancer therapy.

    Topics: Apoptosis; bcl-2-Associated X Protein; CD4-Positive T-Lymphocytes; Cell Proliferation; Forkhead Transcription Factors; Gamma Rays; Humans; Interleukin-2 Receptor alpha Subunit; L-Selectin; Leukocyte Common Antigens; Neoplasms; Phenotype; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2009
The c-MYC-AP4-p21 cascade.
    Cell cycle (Georgetown, Tex.), 2009, Apr-01, Volume: 8, Issue:7

    The p21 gene encodes a CDK-inhibitor, which is induced by p53 and many other anti-proliferative factors. The mechanism of transcriptional repression of p21 by c-MYC has been a subject of intensive study for several years, as it may explain how c-MYC promotes cell cycle progression. Recently, we reported a novel mechanism which allows c-MYC to repress p21: c-MYC triggers a transcriptional cascade by directly inducing the gene encoding the bHLH-LZ transcription factor AP4 (TFAP4), which binds to recognition motifs located in the vicinity of the p21 promoter and mediates transcriptional repression of p21. Thereby, AP4 interferes with induction of p21 via the DNA damage response/p53 or TGFbeta/Smad pathways and during differentiation. Intriguingly, the expression patterns of c-MYC and AP4 strictly overlap in colonic epithelium and colorectal cancer. Here we survey the recent findings and discuss the role of AP4 for c-MYC function and its potential application for cancer diagnosis and therapy.

    Topics: Animals; Apoptosis; Cell Cycle; Cell Line, Tumor; Cyclin-Dependent Kinase Inhibitor p21; DNA Damage; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Proto-Oncogene Proteins c-myc; Smad Proteins, Receptor-Regulated; Transcription Factors; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Wnt Proteins

2009
Novel therapeutic targets along the Th17 pathway.
    European journal of immunology, 2009, Volume: 39, Issue:3

    The recent discovery of IL-17-producing CD4(+) Th subset significantly revised the Th1/Th2 dichotomy model proposed by Mosmann and Coffman almost two decades ago. Th17 cells are involved in the pathogenesis of many human autoimmune diseases. Th17 cells, their developmental pathways and their effector functions, therefore, provide novel therapeutic targets.

    Topics: Animals; Antibodies, Monoclonal; Autoimmune Diseases; Clinical Trials as Topic; Cytokines; Humans; Interleukin-17; Neoplasms; Nuclear Receptor Subfamily 1, Group F, Member 3; Oligonucleotides, Antisense; Receptors, Aryl Hydrocarbon; Receptors, Retinoic Acid; Receptors, Thyroid Hormone; T-Lymphocytes, Helper-Inducer; Transforming Growth Factor beta

2009
Evaluation of immunological status in tumor-bearing dogs.
    Veterinary immunology and immunopathology, 2009, Dec-15, Volume: 132, Issue:2-4

    The purpose of this study was to evaluate immunological status in dogs with cancers at different stages, in comparison with normal dogs. The population of canine peripheral blood lymphocytes (cPBL), lymphocyte phenotypes, interleukin (IL)-6 activity and alpha 1-acid glycoprotein (alpha(1)-AG) level were analyzed. The tumor-bearing dogs had higher numbers of leukocytes than normal dogs, the count being higher in dogs with more advanced tumors. In the tumor-bearing dogs, differential leukocyte counts revealed higher percentages of inflammatory cells such as neutrophils, acidophils and monocytes, and lower numbers of CD4(+)T cells, than in normal dogs, the lymphocyte counts becoming much lower with tumor progression. In the tumor-bearing dogs, the CD8(+)T cell count at the early tumor stage was similar to that in normal dogs, but decreased with tumor progression, possibly reflecting the development of humoral immunity (Th2). Plasma IL-6 and TGF-beta activities were high in the tumor-bearing dogs. The plasma alpha(1)-AG concentration was also significantly high in the tumor-bearing dogs. Our findings suggest that assay of IL-6, TGF-beta and alpha(1)-AG may be very useful for prognostication in dogs with cancer, and that anti-tumor immunity is potently suppressed in such dogs.

    Topics: Aging; Animals; Case-Control Studies; Dog Diseases; Dogs; Female; Immune Tolerance; Interleukin-6; Lymphocyte Count; Lymphocyte Subsets; Male; Neoplasm Staging; Neoplasms; Orosomucoid; Prognosis; Transforming Growth Factor beta

2009
RUNX3 has an oncogenic role in head and neck cancer.
    PloS one, 2009, Jun-12, Volume: 4, Issue:6

    Runt-related transcription factor 3 (RUNX3) is a tumor suppressor of cancer and appears to be an important component of the transforming growth factor-beta (TGF-ss)-induced tumor suppression pathway. Surprisingly, we found that RUNX3 expression level in head and neck squamous cell carcinoma (HNSCC) tissues, which is one of the most common types of human cancer, was higher than that in normal tissues by a previously published microarray dataset in our preliminary study. Therefore, here we examined the oncogenic role of RUNX3 in HNSCC.. Frequent RUNX3 expression and its correlation with malignant behavior were observed in HNSCC. Ectopic RUNX3 overexpression promoted cell growth and inhibited serum starvation-induced apoptosis and chemotherapeutic drug induced apoptosis in HNSCC cells. These findings were confirmed by RUNX3 knockdown. Moreover, RUNX3 overexpression enhanced tumorsphere formation. RUNX3 expression level was well correlated with the methylation status in HNSCC cells. Moreover, RUNX3 expression was low due to the methylation of its promoter in normal oral epithelial cells.. Our findings suggest that i) RUNX3 has an oncogenic role in HNSCC, ii) RUNX3 expression observed in HNSCC may be caused in part by demethylation during cancer development, and iii) RUNX3 expression can be a useful marker for predicting malignant behavior and the effect of chemotherapeutic drugs in HNSCC.

    Topics: Apoptosis; Biomarkers, Tumor; Carcinoma, Squamous Cell; Cell Line, Tumor; Core Binding Factor Alpha 3 Subunit; DNA Methylation; Epithelial Cells; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Head and Neck Neoplasms; Humans; Mouth Neoplasms; Neoplasms; RNA, Small Interfering; Transforming Growth Factor beta

2009
Identification and characterization of Foxp3(+) gammadelta T cells in mouse and human.
    Immunology letters, 2009, Aug-15, Volume: 125, Issue:2

    Regulatory T cells (Tregs) expressing TCRalphabeta play a critical role in the maintenance of the immune system homeostasis. Tregs express the cell surface markers CD4 and CD25 as well as the transcription factor Foxp3. Foxp3(+)CD4(+)CD25(+)TCRalphabeta(+) Tregs can be generated from mouse and human CD4(+)CD25(-) T cells in vitro via TGF-beta induction. As growing evidences suggest that gammadelta T cells also have immunoregulatory function, we have attempted to identify and characterize Foxp3(+) cells in mouse and human gammadelta T cells. We found that freshly isolated mouse splenic gammadelta T cells did not express Foxp3. When mouse splenocytes were stimulated with anti-TCRgammadelta in the presence of TGF-beta, a population of Foxp3(+) gammadelta T cells appeared, in most of which expressed CD25 as well. Compared with CD25(-) gammadelta T cells, TGF-beta induced CD25(+) gammadelta T cells not only expressed Foxp3, but also had increased TGF-beta and GITR expression. Furthermore, the TGF-beta induced gammadelta T cells mediated a potent immunosuppressive effect on anti-CD3 stimulated T cell activation and proliferation. In contrast, although a small fraction of human peripheral blood and tumor infiltrating gammadelta T cells expressed Foxp3, similar culture condition with anti-TCRgammadelta plus TGF-beta failed to generate functional human Foxp3(+) gammadelta T cells. In conclusion, our results suggest that mouse splenic Foxp3(+) gammadelta T cells with suppressive function can be induced by TCR and TGF-beta costimulation, whereas functional human Foxp3(+) gammadelta T cells in peripheral blood could not be generated under the same condition.

    Topics: Animals; CD3 Complex; CD4 Antigens; Cell Movement; Cell Proliferation; Cells, Cultured; Female; Forkhead Transcription Factors; Glucocorticoid-Induced TNFR-Related Protein; Humans; Immune Tolerance; Interleukin-2 Receptor alpha Subunit; Lymphocyte Activation; Male; Mice; Mice, Inbred BALB C; Neoplasms; Receptors, Antigen, T-Cell, gamma-delta; Receptors, Nerve Growth Factor; Receptors, Tumor Necrosis Factor; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2009
The supernatant of apoptotic cells causes transcriptional activation of hypoxia-inducible factor-1alpha in macrophages via sphingosine-1-phosphate and transforming growth factor-beta.
    Blood, 2009, Sep-03, Volume: 114, Issue:10

    Macrophages infiltrating solid tumors exhibit a tumor-supporting phenotype and are critical for tumor development. Little is known which tumor-derived signal provokes this phenotype shift and how these signals are interpreted in macrophages to support tumor growth. We used the supernatant of apoptotic cells and noticed transcriptional, nuclear factor of activated T cells-dependent up-regulation of hypoxia-inducible factor (HIF)-1alpha mRNA, subsequent protein expression, and HIF-1 activity. Blocking calcineurin with cyclosporine A attenuated nuclear factor of activated T cells binding during electrophoretic mobility shift assay analysis and circumvented the HIF-1alpha mRNA increase. Knockdown experiments, receptor analysis, and antibody neutralization pointed to sphingosine-1-phosphate and transforming growth factor-beta as the initiators of the HIF-1 response. The use of macrophages from conditional HIF-1alpha knockout mice revealed that macrophages, under the impact of apoptotic cell supernatants, use HIF-1 to produce factors that induce CD31 expression in murine embryonic stem cells. Our study supports the notion that soluble factors produced from apoptotic tumor cells activate the HIF-1 system under normoxia in macrophages to enhance their tumor-promoting capacity by, for example, releasing vascular endothelial growth factor. This shows the importance of HIF-1-elicited responses in regulatory macrophages under normoxia.

    Topics: Animals; Apoptosis; Calcineurin; Calcineurin Inhibitors; Culture Media, Conditioned; Cyclosporine; Gene Expression Regulation; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Immunosuppressive Agents; Jurkat Cells; Lysophospholipids; Macrophages; Mice; Mice, Knockout; Neoplasms; NFATC Transcription Factors; Platelet Endothelial Cell Adhesion Molecule-1; RNA, Messenger; Sphingosine; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2009
Essential role of TGF-beta signaling in glucose-induced cell hypertrophy.
    Developmental cell, 2009, Volume: 17, Issue:1

    In multicellular organisms, cell size is tightly controlled by nutrients and growth factors. Increasing ambient glucose induces enhanced protein synthesis and cell size. Continued exposure of cells to high glucose in vivo, as apparent under pathological conditions, results in cell hypertrophy and tissue damage. We demonstrate that activation of TGF-beta signaling has a central role in glucose-induced cell hypertrophy in fibroblasts and epithelial cells. Blocking the kinase activity of the TbetaRI receptor or loss of its expression prevented the effects of high glucose on protein synthesis and cell size. Exposure of cells to high glucose induced a rapid increase in cell surface levels of the TbetaRI and TbetaRII receptors and a rapid activation of TGF-beta ligand by matrix metalloproteinases, including MMP-2 and MMP-9. The consequent autocrine TGF-beta signaling in response to glucose led to Akt-TOR pathway activation. Accordingly, preventing MMP-2/MMP-9 or TGF-beta-induced TOR activation inhibited high glucose-induced cell hypertrophy.

    Topics: Active Transport, Cell Nucleus; Animals; Cell Line; Cell Size; Diabetes Mellitus; Epithelial Cells; Fibroblasts; Glucose; Hyperglycemia; Hypertrophy; Matrix Metalloproteinase 2; Matrix Metalloproteinase 9; Mice; Mice, Knockout; Neoplasms; Protein Kinases; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-akt; Rats; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA Interference; Signal Transduction; Smad3 Protein; TOR Serine-Threonine Kinases; Transforming Growth Factor beta

2009
Multidirectional tumor-suppressive activity of AIMP2/p38 and the enhanced susceptibility of AIMP2 heterozygous mice to carcinogenesis.
    Carcinogenesis, 2009, Volume: 30, Issue:9

    Aminoacyl-transfer ribonucleic acid (tRNA) synthetases-interacting multifunctional protein (AIMP) 2 is a factor associated with the macromolecular protein synthesis machinery consisting of nine different aminoacyl-tRNA synthetases and three non-enzymatic factors. However, it was shown to work as a multifaceted regulator through the versatile interactions with diverse signal mediators. For instance, it can mediate pro-apoptotic response to DNA damage and tumor necrosis factor-alpha (TNF-alpha) stimulus and growth-arresting signal by transforming growth factor (TGF)-beta. Considering that these pathways are critically implicated in the control of tumorigenesis, AIMP2 is expected to work as a potent tumor suppressor with broad coverage against different cancer types. Here we investigated whether AIMP2 would give gene dosage effect on its pro-apoptotic and anti-proliferative activities using the wild-type, hetero- and homozygous AIMP2 cells and whether AIMP2 would be critical in preventing tumorigenesis using different in vivo tumor models. Both the apoptotic responses to DNA damage and TNF-alpha and sensitivity to growth arresting TGF-beta signal were reduced in AIMP2 hetero- and homozygous cells compared with the wild-type cells in dose-dependent manner. In all the in vivo carcinogenesis experiments, reduction of AIMP2 level in heterozygous AIMP2 mice provided higher susceptibility to tumor formation. Thus, this work proves the functional significance of AIMP2 in determination of cell proliferation and death, and as a haploinsufficient tumor suppressor.

    Topics: Amino Acyl-tRNA Synthetases; Animals; Apoptosis; Cells, Cultured; Colonic Neoplasms; Disease Susceptibility; DNA Damage; Heterozygote; Lung Neoplasms; Mice; Mice, Inbred C57BL; Neoplasms; NF-kappa B; Papilloma; Skin Neoplasms; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Tumor Suppressor Proteins

2009
Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity.
    Cancer research, 2009, Oct-01, Volume: 69, Issue:19

    Natural killer (NK) cells play an important role in cancer immunosurveillance and may prevent tumor progression and metastasis due to their ability to mediate direct cellular cytotoxicity and by releasing immunoregulatory cytokines, which shape adaptive immune responses. Their reactivity is governed by various activating and inhibitory molecules expressed on target cells and reciprocal interactions with other hematopoietic cells such as dendritic cells. In mice, thrombocytopenia inhibits metastasis, and this is reversed by NK cell depletion, suggesting that platelets are an important additional player in NK cell-tumor interaction. Moreover, it has been shown that metastasizing tumor cells do not travel through the blood alone but are rapidly coated by platelets. However, the knowledge about the molecular mechanisms by which platelets influence NK cells is fragmentary at best. Here we show that platelet-derived soluble factors, secreted on coating of tumor cells or after stimulation with classic platelet agonists, impair NK cell antitumor reactivity resulting in diminished granule mobilization, cytotoxicity, and IFN-gamma production. The impaired NK cell reactivity was not due to induction of apoptosis but mediated by down-regulation of the activating immunoreceptor natural killer group 2, member D (NKG2D) on NK cells by platelet-derived transforming growth factor beta (TGF-beta). Neutralization of TGF-beta in platelet releasate not only prevented NKG2D down-regulation but also restored NK cell antitumor reactivity. Thus, our data elucidate the molecular basis of the previously described influence of platelets on NK cell antitumor reactivity and suggest that therapeutic intervention in tumor cell-platelet interaction and the resulting TGF-beta release by platelets may serve to enhance antitumor immunity.

    Topics: Blood Platelets; Cell Line, Tumor; Down-Regulation; HCT116 Cells; Humans; Interferon-gamma; Killer Cells, Natural; Neoplasms; NK Cell Lectin-Like Receptor Subfamily K; Platelet Activation; Transforming Growth Factor beta

2009
TGF-beta induces fibroblast activation protein expression; fibroblast activation protein expression increases the proliferation, adhesion, and migration of HO-8910PM [corrected].
    Experimental and molecular pathology, 2009, Volume: 87, Issue:3

    Several studies recognize cancer-stromal fibroblasts' role in cancer-cell invasion and metastasis. Through paracrine signaling molecules, TGF-beta and IL-1beta, cancer cells activate stromal fibroblasts and induce the expression of fibroblast activation protein (FAP). FAP, in turn, affects the proliferation, invasion and migration of the cancer cells. We report that TGF-beta and IL-1beta are important factors in inducing differentiation of myofibroblasts and expression of functional markers, notably alpha-SMA. We discover that TGF-beta is the dominant factor in promoting FAPalpha protein expression. This study also examines FAP's function in vitro by assaying the proliferation, migration and invasion of ovarian cancer cell line HO-8910PM.

    Topics: Animals; Cell Adhesion; Cell Differentiation; Cell Line, Tumor; Cell Movement; Cell Proliferation; Endopeptidases; Enzyme Activation; Fibroblasts; Gelatinases; Humans; Interleukin-1beta; Membrane Proteins; Mice; Neoplasm Invasiveness; Neoplasms; NIH 3T3 Cells; Serine Endopeptidases; Transforming Growth Factor beta

2009
TGF-beta helps cells fly solo.
    Nature cell biology, 2009, Volume: 11, Issue:11

    Topics: Animals; Humans; Lymphatic Metastasis; Neoplasm Metastasis; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2009
Enhancing cancer vaccine efficacy via modulation of the tumor microenvironment.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2009, Nov-01, Volume: 15, Issue:21

    The development of therapeutic cancer vaccines is impacted by immunosuppressive elements in the tumor microenvironment. Most immunogenic cancer proteins are "self," therefore, peripheral tolerance contributes substantially to tumor immune escape. Transforming growth factor beta (TGFbeta) actively modulates both inflammation and tolerance induction. Combining vaccination with agents that disarm TGFbeta will enhance vaccine efficacy.

    Topics: Animals; Autoantigens; Cancer Vaccines; Female; Immune Tolerance; Immunomodulation; Male; Mice; Models, Immunological; Neoplasms; Transforming Growth Factor beta; Tumor Escape

2009
Companies waver in efforts to target transforming growth factor beta in cancer.
    Journal of the National Cancer Institute, 2009, Dec-16, Volume: 101, Issue:24

    Topics: Animals; Antibodies, Monoclonal; Antineoplastic Agents; Cell Proliferation; Clinical Trials as Topic; Combined Modality Therapy; Drug Industry; Humans; Immunotherapy; Mice; Mice, Transgenic; Neoplasms; Oligonucleotides, Antisense; Precision Medicine; Protein Kinase Inhibitors; Transforming Growth Factor beta

2009
Discussion. A comparative analysis and systematic review of the wound-healing milieu: implications for body contouring after massive weight loss.
    Plastic and reconstructive surgery, 2009, Volume: 124, Issue:5

    Topics: Bariatric Surgery; Biomarkers; Burns; Humans; Matrix Metalloproteinase 2; Matrix Metalloproteinase 3; Matrix Metalloproteinase 9; Neoplasms; Obesity; Organ Transplantation; Plastic Surgery Procedures; Tissue Inhibitor of Metalloproteinase-1; Tissue Inhibitor of Metalloproteinase-2; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A; Weight Loss; Wound Healing

2009
Lefty peptides, derived by MMP2 cleavage, act as a new class of gelatinase A inhibitor.
    Frontiers in bioscience : a journal and virtual library, 2008, May-01, Volume: 13

    MMPs are zinc-dependent endopeptidases that are involved in proteolysis of extracellular matrix in both physiological and pathological processes including cancer. MMPs are involved at all stages of tumor progression, including tumor growth, angiogenesis, and metastasis. We recently showed that overexpression of Lefty in cancer cells restrains tumor growth. Here, we show that small forms of Lefty are generated by MMP2 (gelatinase A) mediated cleavage. In turn, these forms of Lefty strongly inhibit the autocatalytic, gelatinolytic and caseinolytic activities of MMP2 in vitro. We show that a short synthesized form of Lefty peptide (CASDGALVP) inhibits gelatinolytic and caseinolytic activities of MMP2 in vitro and inhibits tumor growth in vivo. Together, these findings show that lefty peptides are a new class of gelatinase A inhibitors that restrain tumor growth.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Electrophoresis, Polyacrylamide Gel; Extracellular Matrix; Female; Gelatinases; Humans; Left-Right Determination Factors; Matrix Metalloproteinase 2; Mice; Neoplasms; Tissue Inhibitor of Metalloproteinase-2; Transforming Growth Factor beta

2008
SUMO amplifies TGF-beta signalling.
    Nature cell biology, 2008, Volume: 10, Issue:6

    Transforming growth factor-beta (TGF-beta) stimulates phosphorylation of TGF-beta type I receptor. This receptor is now shown to be sumoylated, leading to enhanced activation and modulation of the downstream Smad signalling pathway.

    Topics: Animals; Gene Expression Regulation; Humans; Models, Biological; Mutation; Neoplasm Metastasis; Neoplasms; Phosphorylation; Signal Transduction; SUMO-1 Protein; Transforming Growth Factor beta

2008
Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells.
    Proceedings of the National Academy of Sciences of the United States of America, 2008, Jul-08, Volume: 105, Issue:27

    Although mature dendritic cells (DCs) are potent initiators of adaptive immune response, immature steady-state DCs contribute to immune tolerance. In this study, we show that ex vivo splenic DCs are capable of inducing conversion of naïve CD4(+) T cells to adaptive Foxp3(+)CD4(+) regulatory T cells (aTreg) in the presence of TGF-beta. In particular, when compared with splenic CD8alpha(-) DCs, the CD8alpha(+) DC subset were superior in inducing higher frequencies of conversion. This was not attributable to the difference in basal level of costimulation, because deficiency of CD40 or CD80/86 signaling did not diminish the differential induction of Foxp3. Conversion was regulated by DC maturation status. Further insights into the molecular mechanisms of conversion were gained by analyzing the contribution of several costimulatory and coinhibitory receptors. Costimulatory signals through GITR suppressed conversion, whereas coinhibitory signaling via programmed death 1 ligand (PD-L1) but not PD-L2 was required for conversion. Ex vivo PD-L1(-/-) DCs failed to support Foxp3 induction in the presence of TGF-beta. In vivo blocking PD-L1 signaling abolished conversion in a tumor-induced aTreg conversion model. Collectively, this study highlights the cellular and molecular parameters that might be exploited to control the de novo generation of aTregs and peripheral tolerance.

    Topics: Animals; Antigens, Surface; Apoptosis Regulatory Proteins; CD8 Antigens; Cell Differentiation; Cell Proliferation; Dendritic Cells; Epitopes; Forkhead Transcription Factors; Kinetics; Mice; Mice, Inbred C57BL; Neoplasm Transplantation; Neoplasms; Programmed Cell Death 1 Receptor; Signal Transduction; Spleen; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2008
A novel gene expression profile in lymphatics associated with tumor growth and nodal metastasis.
    Cancer research, 2008, Sep-15, Volume: 68, Issue:18

    Invasion of lymphatic vessels is a key step in the metastasis of primary tumors to draining lymph nodes. Although the process is enhanced by tumor lymphangiogenesis, it is unclear whether this is a consequence of increased lymphatic vessel number, altered lymphatic vessel properties, or both. Here we have addressed the question by comparing the RNA profiles of primary lymphatic endothelial cells (LEC) isolated from the vasculature of normal tissue and from highly metastatic T-241/vascular endothelial growth factor (VEGF)-C fibrosarcomas implanted in C57BL/6 mice. Our findings reveal significant differences in expression of some 792 genes (i.e., >or=2-fold up- or down-regulated, P

    Topics: Animals; Cell Adhesion Molecules; Cell Growth Processes; Endoglin; Endothelial Cells; Fibrosarcoma; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Intracellular Signaling Peptides and Proteins; Junctional Adhesion Molecules; Lymph Nodes; Lymphatic Metastasis; Mice; Mice, Inbred C57BL; Neoplasms; Neovascularization, Pathologic; Oligonucleotide Array Sequence Analysis; Receptors, Leptin; Transforming Growth Factor beta; Vascular Endothelial Growth Factor C

2008
Stem cell antigen 2: a new gene involved in the self-renewal of erythroid progenitors.
    Cell proliferation, 2008, Volume: 41, Issue:5

    Stem cell antigen 2 (SCA2), also known as TSA1 and LY6E, is a glycosylphosphatidylinositol-anchored molecule that belongs to the Ly-6 family and whose function remains largely unknown. We have previously shown that SCA2 is overexpressed in self-renewing avian erythroid progenitors (T2ECs) as opposed to differentiating T2ECs. The aim of this study was to define the role of SCA2 in the switch between self-renewal and differentiation of erythroid progenitors.. We have investigated the cellular processes controlled by SCA2 in T2ECs by RNA interference and overexpression approaches. Moreover, we have used a SAGE Querying and analysis tools developed in our laboratory, to investigate the expression level of SCA2 gene in different human cell types.. We demonstrate the regulation of SCA2 expression by TGF-beta, a growth factor essential for self-renewal of T2ECs. We establish that SCA2 knockdown by RNA interference reduced the proliferation and promoted the differentiation of T2ECs. In contrast, SCA2 overexpression inhibited differentiation of T2ECs only. Furthermore, by using a bioinformatic approach, we found that SCA2 is highly expressed in a variety of human cancer cells. We confirmed this result by quantitative PCR on human colon and kidney tissues.. Altogether, these findings imply that SCA2 may function in a dose-dependent manner to support the self-renewal state and that its deregulation might contribute to the development of some human cancers.

    Topics: Animals; Cell Differentiation; Cell Proliferation; Chickens; Down-Regulation; Erythroid Precursor Cells; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Membrane Proteins; Neoplasms; Receptors, Transforming Growth Factor beta; RNA Interference; Transforming Growth Factor beta

2008
HMGA2 and Smads co-regulate SNAIL1 expression during induction of epithelial-to-mesenchymal transition.
    The Journal of biological chemistry, 2008, Nov-28, Volume: 283, Issue:48

    Epithelial-mesenchymal transition (EMT) is important during embryonic cell layer movement and tumor cell invasiveness. EMT converts adherent epithelial cells to motile mesenchymal cells, favoring metastasis in the context of cancer progression. Transforming growth factor-beta (TGF-beta) triggers EMT via intracellular Smad transducers and other signaling proteins. We previously reported that the high mobility group A2 (HMGA2) gene is required for TGF-beta to elicit EMT in mammary epithelial cells. In the present study we investigated the molecular mechanisms by which HMGA2 induces EMT. We found that HMGA2 regulates expression of many important repressors of E-cadherin. Among these, we analyzed in detail the zinc-finger transcription factor SNAIL1, which plays key roles in tumor progression and EMT. We demonstrate that HMGA2 directly binds to the SNAIL1 promoter and acts as a transcriptional regulator of SNAIL1 expression. Furthermore, we observed that HMGA2 cooperates with the TGF-beta/Smad pathway in regulating SNAIL1 gene expression. The mechanism behind this cooperation involves physical interaction between these factors, leading to an increased binding of Smads to the SNAIL1 promoter. SNAIL1 seems to play the role of a master effector downstream of HMGA2 for induction of EMT, as SNAIL1 knock-down partially reverts HMGA2-induced loss of epithelial differentiation. The data propose that HMGA2 acts in a gene-specific manner to orchestrate the transcriptional network necessary for the EMT program.

    Topics: Animals; Cell Differentiation; Cell Movement; Chlorocebus aethiops; COS Cells; Epithelium; Gene Expression Regulation, Developmental; Gene Knockdown Techniques; HMGA2 Protein; Humans; Mammary Glands, Animal; Mammary Glands, Human; Mesoderm; Mice; Neoplasm Metastasis; Neoplasms; Promoter Regions, Genetic; Signal Transduction; Smad Proteins; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2008
[Rationale for the use of systemic enzyme therapy in malignant diseases].
    Deutsche medizinische Wochenschrift (1946), 2008, Volume: 133 Suppl 9

    Topics: Administration, Oral; alpha-Macroglobulins; Animals; Female; Humans; Male; Neoplasms; Peptide Hydrolases; Quality of Life; Survival Rate; Transforming Growth Factor beta

2008
Enzymes and cancer: a look toward the past as we move forward.
    Integrative cancer therapies, 2008, Volume: 7, Issue:4

    Topics: Clinical Trials as Topic; Complementary Therapies; Humans; Neoplasms; Peptide Hydrolases; Transforming Growth Factor beta; Treatment Outcome

2008
Choline kinase as a link connecting phospholipid metabolism and cell cycle regulation: implications in cancer therapy.
    The international journal of biochemistry & cell biology, 2008, Volume: 40, Issue:9

    Choline kinase alpha (ChoKalpha) is an enzyme involved in the metabolism of phospholipids recently found to play a relevant role in the regulation of cell proliferation, oncogenic transformation and human carcinogenesis. In addition, this novel oncogene has been recently defined as a prognostic factor in human cancer, and as a promising target for therapy since its specific inhibitors display efficient antitumoral activity in vivo. However, the mechanism by which this enzyme is involved in the regulation of these processes is not yet understood. Using differential microarray analysis, we identify target genes that provide the basis for the understanding of the molecular mechanism for the regulation of cell proliferation and transformation mediated by over-expression of the human ChoKalpha. These results fully support a critical role of this enzyme in the regulation of the G1-->S transition at different levels, and its relevant role in human carcinogenesis. The molecular basis to understand the connection between phospholipids metabolism and cell cycle regulation through choline kinase is reported.

    Topics: Animals; Apoptosis; Cattle; Cell Cycle; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Choline Kinase; Enzyme Inhibitors; Gene Expression Regulation, Neoplastic; Humans; Mice; Neoplasms; Oligonucleotide Array Sequence Analysis; Phospholipids; Receptors, Transforming Growth Factor beta; Reproducibility of Results; Signal Transduction; Substrate Specificity; Transforming Growth Factor beta

2008
[The "Snail" genes and renal diseases: what we learn from organogenesis].
    Medecine sciences : M/S, 2008, Volume: 24, Issue:3

    Topics: Animals; Cadherins; Cell Transdifferentiation; Embryonic Development; Epithelial Cells; Fibroblasts; Fibrosis; Humans; Kidney; Kidney Diseases; Mesoderm; Neoplasms; Neural Crest; Organogenesis; Snail Family Transcription Factors; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta

2008
Human embryonic stem cell microenvironment suppresses the tumorigenic phenotype of aggressive cancer cells.
    Proceedings of the National Academy of Sciences of the United States of America, 2008, Mar-18, Volume: 105, Issue:11

    Embryonic stem cells sustain a microenvironment that facilitates a balance of self-renewal and differentiation. Aggressive cancer cells, expressing a multipotent, embryonic cell-like phenotype, engage in a dynamic reciprocity with a microenvironment that promotes plasticity and tumorigenicity. However, the cancer-associated milieu lacks the appropriate regulatory mechanisms to maintain a normal cellular phenotype. Previous work from our laboratory reported that aggressive melanoma and breast carcinoma express the embryonic morphogen Nodal, which is essential for human embryonic stem cell (hESC) pluripotency. Based on the aberrant expression of this embryonic plasticity gene by tumor cells, this current study tested whether these cells could respond to regulatory cues controlling the Nodal signaling pathway, which might be sequestered within the microenvironment of hESCs, resulting in the suppression of the tumorigenic phenotype. Specifically, we discovered that metastatic tumor cells do not express the inhibitor to Nodal, Lefty, allowing them to overexpress this embryonic morphogen in an unregulated manner. However, exposure of the tumor cells to a hESC microenvironment (containing Lefty) leads to a dramatic down-regulation in their Nodal expression concomitant with a reduction in clonogenicity and tumorigenesis accompanied by an increase in apoptosis. Furthermore, this ability to suppress the tumorigenic phenotype is directly associated with the secretion of Lefty, exclusive to hESCs, because it is not detected in other stem cell types, normal cell types, or trophoblasts. The tumor-suppressive effects of the hESC microenvironment, by neutralizing the expression of Nodal in aggressive tumor cells, provide previously unexplored therapeutic modalities for cancer treatment.

    Topics: Cell Culture Techniques; Cells, Cultured; Embryonic Stem Cells; Humans; Neoplasms; Nodal Protein; Phenotype; Signal Transduction; Transforming Growth Factor beta; Tumor Stem Cell Assay

2008
Two novel VHL targets, TGFBI (BIGH3) and its transactivator KLF10, are up-regulated in renal clear cell carcinoma and other tumors.
    Biochemical and biophysical research communications, 2008, Jun-13, Volume: 370, Issue:4

    Mutations in the VHL gene are associated with highly vascular tumors of kidney, brain, retina, and adrenal gland. The inability of the mutant VHL protein to destabilize HIF-1 plays a crucial role in malignant angiogenesis. VHL is also associated with ECM assembly but the molecular mechanisms of this activity remain unclear. We used expression arrays and cell lines with different VHL status to identify ECM-associated genes controlled by VHL. One of them, adhesion-associated TGFBI, was repressed by VHL and overexpressed in renal, gastrointestinal, brain, and other tumors. Analyzing the mechanism of TGFBI up-regulation in clear cell carcinoma, we identified a novel VHL target, a Kruppel-like transcriptional factor 10 (KLF10). The TGFBI promoter, which we isolated and studied in Luc-reporter assay, was induced by KLF10 but not hypoxia. These data provide the molecular basis for the observed VHL effect on TGFBI and stimulate further research into the KLF10 and TGFBI roles in cancer.

    Topics: Antineoplastic Agents; Carcinoma, Renal Cell; Cell Adhesion; Early Growth Response Transcription Factors; Extracellular Matrix Proteins; Gene Expression Regulation, Neoplastic; Humans; Kidney Neoplasms; Kruppel-Like Transcription Factors; Neoplasms; Oligonucleotide Array Sequence Analysis; Promoter Regions, Genetic; Transcription, Genetic; Transforming Growth Factor beta; Up-Regulation; Von Hippel-Lindau Tumor Suppressor Protein

2008
Regulation of Id1 expression by SRC: implications for targeting of the bone morphogenetic protein pathway in cancer.
    Cancer research, 2008, Apr-01, Volume: 68, Issue:7

    Deregulated activation of the Src tyrosine kinase and heightened Id1 expression are independent mediators of aggressive tumor biology. The present report implicates Src signaling as a critical regulator of Id1 gene expression. Microarray analyses showed that Id family genes were among the most highly down-regulated by incubation of A549 lung carcinoma cells with the small-molecule Src inhibitor AZD0530. Id1 transcript and protein levels were potently reduced in a dose-dependent manner concomitantly with the reduction of activated Src levels. These effects were conserved across a panel of lung, breast, prostate, and colon cancer cell lines and confirmed by the ability of PP2, Src siRNA, and Src-blocking peptides to suppress Id1 expression. PP2, AZD0530, and dominant-negative Src abrogated Id1 promoter activity, which was induced by constitutively active Src. The Src-responsive region of the Id1 promoter was mapped to a region 1,199 to 1,360 bps upstream of the translation start site and contained a Smad-binding element. Src was also required for bone morphogenetic protein-2 (BMP-2)-induced Id1 expression and promoter activity, was moderately activated by BMP-2, and complexed with Smad1/5. Conversely, Src inhibitors blocked Smad1/5 nuclear translocation and binding to the Src-responsive region of the Id1 promoter. Consistent with a role for Src and Id1 in cancer cell invasion, Src inhibitors and Id1 siRNA decreased cancer cell invasion, which was increased by Id1 overexpression. Taken together, these results reveal that Src positively interacts with the BMP-Smad-Id pathway and provide new ways for targeted inhibition of Id1.

    Topics: Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Cell Line, Tumor; Down-Regulation; Humans; Inhibitor of Differentiation Protein 1; Neoplasm Invasiveness; Neoplasms; Promoter Regions, Genetic; RNA, Messenger; RNA, Small Interfering; Signal Transduction; Smad Proteins; src-Family Kinases; Transfection; Transforming Growth Factor beta

2008
Cancer biology. All in the stroma: cancer's Cosa Nostra.
    Science (New York, N.Y.), 2008, Apr-04, Volume: 320, Issue:5872

    Topics: Animals; Cell Communication; Extracellular Matrix; Fibroblasts; Humans; Integrins; Intercellular Signaling Peptides and Proteins; Macrophages; Matrix Metalloproteinases; Mesenchymal Stem Cells; Myeloid Cells; Neoplasm Metastasis; Neoplasms; Transforming Growth Factor beta

2008
The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2.
    The Journal of biological chemistry, 2008, May-30, Volume: 283, Issue:22

    MicroRNAs are small non-coding RNA molecules that can regulate gene expression by interacting with multiple mRNAs and inducing either translation suppression or degradation of mRNA. Recently, several miRNAs were identified as either promoters or suppressors of metastasis. However, it is unclear in which step(s) of the multistep metastatic cascade these miRNAs play a defined functional role. To study the functional importance of miRNAs in epithelial-mesenchymal transition (EMT), a process thought to initiate metastasis by enhancing the motility of tumor cells, we used a well established in vitro EMT assay: transforming growth factor-beta-induced EMT in NMuMG murine mammary epithelial cells. We found that members of the miR-200 family, organized as two clusters in the genome, were repressed during EMT. Overexpression of each miRNA individually or as clusters in NMuMG cells hindered EMT by enhancing E-cadherin expression through direct targeting of ZEB1 and ZEB2, which encode transcriptional repressors of E-cadherin. In the 4TO7 mouse carcinoma cell line, which expresses low levels of endogenous E-cadherin and displays a mesenchymal phenotype, ectopic expression of the miR-200 family miRNAs significantly increased E-cadherin expression and altered cell morphology to an epithelial phenotype. Furthermore, ectopic expression of each miR-200 miRNA cluster significantly reduced the in vitro motility of 4TO7 cells in migration assays. These results suggested that loss of expression of the miR-200 family members may play a critical role in the repression of E-cadherin by ZEB1 and ZEB2 during EMT, thereby enhancing migration and invasion during cancer progression.

    Topics: Animals; Cadherins; Cell Movement; Epithelial Cells; Gene Expression Regulation, Neoplastic; Genome, Human; HeLa Cells; Homeodomain Proteins; Humans; Kruppel-Like Transcription Factors; Mice; MicroRNAs; Multigene Family; Neoplasm Metastasis; Neoplasms; Repressor Proteins; Transcription Factors; Transforming Growth Factor beta; Zinc Finger E-box Binding Homeobox 2; Zinc Finger E-box-Binding Homeobox 1

2008
Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-beta receptor.
    Journal of immunotherapy (Hagerstown, Md. : 1997), 2008, Volume: 31, Issue:5

    Transforming growth factor (TGF)-beta is produced in most human tumors and markedly inhibits tumor antigen-specific cellular immunity, representing a major obstacle to the success of tumor immunotherapy. TGF-beta is produced in Epstein-Barr virus (EBV)-positive Hodgkin disease and non-Hodgkin lymphoma both by the tumor cells and by infiltrating T-regulatory cells and may contribute the escape of these tumors from infused EBV-specific T cells. To determine whether tumor antigen-specific cytotoxic T lymphocytes (CTLs) can be shielded from the inhibitory effects of tumor-derived TGF-beta, we previously used a hemagglutinin-tagged dominant negative TGF-betaRII expressed from a retrovirus vector to provide CTLs with resistance to the inhibitory effects of TGF-beta in vitro. We now show that human tumor antigen-specific CTLs can be engineered to resist the inhibitory effects of tumor-derived TGF-beta both in vitro and in vivo using a clinical grade retrovirus vector in which the dominant negative TGF-beta type II receptor (DNRII) was modified to remove the immunogenic hemagglutinin tag. TGF-beta-resistant CTL had a functional advantage over unmodified CTL in the presence of TGF-beta-secreting EBV-positive lymphoma, and had enhanced antitumor activity, supporting the potential value of this countermeasure.

    Topics: Animals; Cell Line, Tumor; Cell Proliferation; Genes, Dominant; Herpesvirus 4, Human; Humans; Mice; Neoplasms; Receptors, Transforming Growth Factor beta; T-Lymphocytes; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2008
Transforming growth factor beta subverts the immune system into directly promoting tumor growth through interleukin-17.
    Cancer research, 2008, May-15, Volume: 68, Issue:10

    Overexpression of the immunosuppressive cytokine transforming growth factor beta (TGF-beta) is one strategy that tumors have developed to evade effective immunesurveillance. Using transplantable models of breast and colon cancer, we made the unexpected finding that CD8+ cells in tumor-bearing animals can directly promote tumorigenesis, by a mechanism that is dependent on TGF-beta. We showed that CD8+ splenocytes from tumor-bearing mice expressed elevated interleukin (IL)-17 when compared with naive mice, and that CD8+ T cells could be induced to make IL-17 on addition of TGF-beta and IL-6 in vitro. Treatment of mice with anti-TGF-beta antibodies in vivo reduced IL-17 expression both in the tumor and the locoregional lymph nodes. Although IL-17 has not previously been shown to act as a survival factor for epithelial cells, we found that IL-17 suppressed apoptosis of several tumor cell lines in vitro, suggesting that this altered T-cell polarization has the potential to promote tumorigenesis directly, rather than indirectly through inflammatory sequelae. Consistent with this hypothesis, knockdown of the IL-17 receptor in 4T1 mouse mammary cancer cells enhanced apoptosis and decreased tumor growth in vivo. Thus, in addition to suppressing immune surveillance, tumor-induced TGF-beta may actively subvert the CD8+ arm of the immune system into directly promoting tumor growth by an IL-17-dependent mechanism.

    Topics: Animals; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cell Line, Tumor; Female; Gene Expression Regulation, Neoplastic; Humans; Interleukin-17; Lymph Nodes; Mice; Mice, Inbred BALB C; Neoplasm Transplantation; Neoplasms; Rats; Transforming Growth Factor beta

2008
Circulating myeloid dendritic cells of advanced cancer patients result in reduced activation and a biased cytokine profile in invariant NKT cells.
    Journal of immunology (Baltimore, Md. : 1950), 2008, Jun-01, Volume: 180, Issue:11

    CD1d-restricted invariant NKT (iNKT) cells play important regulatory roles in various immune responses, including antitumor immune responses. Previous studies have demonstrated quantitative and qualitative defects in iNKT cells of cancer patients, and these defects are clinically relevant as they are associated with poor prognosis. In this study we demonstrate that defects in the iNKT cell population can, at least in part, be attributed to defective interactions between iNKT cells and CD1d-expressing circulating myeloid dendritic cells (mDC), as mDC of patients with advanced melanoma and renal cell cancer reduced the activation and Th1 cytokine production of healthy donor-derived iNKT cells. Interestingly, this reduced activation of iNKT cells was restricted to patients with low circulating iNKT cell numbers and could be reversed by IL-12 and in part by the neutralization of TGF-beta, but it was further reduced by the neutralization of IL-10 in vitro. Additional experiments revealed discordant roles for TGF-beta and IL-10 on human iNKT cells, because TGF-beta suppressed iNKT cell activation and proliferation and IFN-gamma production while IL-10 was identified as a cytokine involved in stimulating the activation and expansion of iNKT cells that could subsequently suppress NK cell and T cell responses.

    Topics: Adult; Aged; Antigens, CD1; Cell Line; Cytokines; Dendritic Cells; Female; Humans; Interferon-gamma; Interleukin-10; Killer Cells, Natural; Lymphocyte Activation; Male; Middle Aged; Myeloid Cells; Neoplasms; T-Lymphocyte Subsets; Transforming Growth Factor beta

2008
The human Cyr61 gene is a transcriptional target of transforming growth factor beta in cancer cells.
    Cancer letters, 2007, Feb-08, Volume: 246, Issue:1-2

    Cyr61 is a multifunctional protein that can stimulate angiogenesis and tumor growth. Its expression by many cancers and breast cancers increases with tumor grade. Cyr61 is closely related to connective tissue growth factor, CTGF. Both proteins regulate skeletal development, suggesting that they could contribute to breast cancer metastases to bone, a process regulated by TGFbeta. We show that Cyr61 transcription is activated by TGFbeta and that the human Cyr61 promoter contains consensus sequences that bind Smad proteins. TGFbeta in the tumor microenvironment may stimulate cancer metastases to sites such as bone by increasing Cyr61 expression and secretion.

    Topics: Cell Line, Tumor; Connective Tissue Growth Factor; Cysteine-Rich Protein 61; Electrophoretic Mobility Shift Assay; Gene Expression Regulation, Neoplastic; Humans; Immediate-Early Proteins; Intercellular Signaling Peptides and Proteins; Luciferases; Mutation; Neoplasms; Promoter Regions, Genetic; Protein Binding; Recombinant Fusion Proteins; RNA, Messenger; Smad Proteins; Smad3 Protein; Transcription, Genetic; Transfection; Transforming Growth Factor beta

2007
Inactivation of myocardin and p16 during malignant transformation contributes to a differentiation defect.
    Cancer cell, 2007, Volume: 11, Issue:2

    Myocardin is known as an important transcriptional regulator in smooth and cardiac muscle development. Here we found that myocardin is frequently repressed during human malignant transformation, contributing to a differentiation defect. We demonstrate that myocardin is a transcriptional target of TGFbeta required for TGFbeta-mediated differentiation of human fibroblasts. Serum deprivation, intact contact inhibition response, and the p16ink4a/Rb pathway contribute to myocardin induction and differentiation. Restoration of myocardin expression in sarcoma cells results in differentiation and inhibition of malignant growth, whereas inactivation of myocardin in normal fibroblasts increases their proliferative potential. Myocardin expression is reduced in multiple types of human tumors. Collectively, our results demonstrate that myocardin is an important suppressive modifier of the malignant transformation process.

    Topics: Blotting, Western; Cell Adhesion; Cell Differentiation; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Colony-Forming Units Assay; Cyclin-Dependent Kinase Inhibitor p16; DNA Methylation; Fibroblasts; Fluorescent Antibody Technique; Gene Expression Regulation; Humans; Lung; Mesoderm; Neoplasms; Nuclear Proteins; Plasmids; Promoter Regions, Genetic; RNA, Small Interfering; Trans-Activators; Transforming Growth Factor beta

2007
Improvement of cancer-targeting therapy, using nanocarriers for intractable solid tumors by inhibition of TGF-beta signaling.
    Proceedings of the National Academy of Sciences of the United States of America, 2007, Feb-27, Volume: 104, Issue:9

    Transforming growth factor (TGF)-beta plays a pivotal role in regulation of progression of cancer through effects on tumor microenvironment as well as on cancer cells. TGF-beta inhibitors have recently been shown to prevent the growth and metastasis of certain cancers. However, there may be adverse effects caused by TGF-beta signaling inhibition, including the induction of cancers by the repression of TGF-beta-mediated growth inhibition. Here, we present an application of a short-acting, small-molecule TGF-beta type I receptor (TbetaR-I) inhibitor at a low dose in treating several experimental intractable solid tumors, including pancreatic adenocarcinoma and diffuse-type gastric cancer, characterized by hypovascularity and thick fibrosis in tumor microenvironments. Low-dose TbetaR-I inhibitor altered neither TGF-beta signaling in cancer cells nor the amount of fibrotic components. However, it decreased pericyte coverage of the endothelium without reducing endothelial area specifically in tumor neovasculature and promoted accumulation of macromolecules, including anticancer nanocarriers, in the tumors. Compared with the absence of TbetaR-I inhibitor, anticancer nanocarriers exhibited potent growth-inhibitory effects on these cancers in the presence of TbetaR-I inhibitor. The use of TbetaR-I inhibitor combined with nanocarriers may thus be of significant clinical and practical importance in treating intractable solid cancers.

    Topics: Animals; Cell Line, Tumor; Drug Delivery Systems; Endothelium, Vascular; Humans; Immunohistochemistry; Mice; Mice, Inbred BALB C; Nanoparticles; Neoplasms; Protein Serine-Threonine Kinases; Pyrazoles; Pyrroles; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2007
Inflammation and cancer: a failure of resolution?
    Trends in pharmacological sciences, 2007, Volume: 28, Issue:4

    There are clear links between chronic inflammation and cancer; strong epidemiological and genetic evidence indicates that inflammation can drive tumour progression, and more-recent evidence indicates that the disruption of endogenous anti-inflammatory mechanisms in mice can lead to tumour development. The resolution of inflammation is an active coordinated process that requires the production of anti-inflammatory mediators, the termination of proinflammatory signalling pathways and the appropriate clearance or migration of inflammatory cells. Disruption of any of these processes can lead to chronic persistent inflammation and tumour growth. Although the mediators and mechanisms that drive inflammation have become increasingly well characterized, the endogenous mechanisms that limit the inflammatory response, and particularly their role in cancer, are unclear. There are clear opportunities for drug discovery and the development of new therapeutic approaches that target tumour-associated inflammation and the mechanisms of chronic inflammation.

    Topics: Animals; Humans; Inflammation; Interleukin-10; Macrophages; Neoplasms; Transforming Growth Factor beta

2007
CD28 costimulation overcomes transforming growth factor-beta-mediated repression of proliferation of redirected human CD4+ and CD8+ T cells in an antitumor cell attack.
    Cancer research, 2007, Mar-01, Volume: 67, Issue:5

    The T-cell-mediated antitumor immune response is frequently repressed in the tumor environment by an immunologic barrier, the predominant mediators of which are thought to be interleukin-10 (IL-10) and transforming growth factor-beta (TGF-beta). We explored the effect of these cytokines on the individual T-cell effector functions on antigen engagement during an antitumor cell attack. Isolated CD4+ and CD8+ T cells were antigen-specifically redirected toward carcinoembryonic antigen (CEA)-positive tumor cells by expression of a recombinant T-cell receptor (immunoreceptor), which triggers T-cell activation via CD3zeta on binding to CEA. Immunoreceptor-activated T cells secrete IFN-gamma, proliferate, and lyse CEA+ but not CEA- tumor cells. Whereas IL-10 has no direct effect on immunoreceptor-triggered effector functions, TGF-beta represses proliferation of both CD4+ and CD8+ T cells but neither IFN-gamma secretion nor specific cytolytic activities. CD28 costimulation, however, overcomes TGF-beta-mediated repression in T-cell proliferation. Consequently, T cells redirected by a combined CD28-CD3zeta signaling immunoreceptor are largely resistant to TGF-beta-mediated repression. This is reflected in vivo by a more pronounced antitumor activity of T cells against TGF-beta-secreting tumors when redirected by a costimulatory CD28-CD3zeta than by a CD3zeta signaling immunoreceptor.

    Topics: Animals; CD28 Antigens; CD3 Complex; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cell Proliferation; Cell Survival; Humans; Immunity, Cellular; Mice; Mice, Nude; Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2007
Expression profiling of genes regulated by TGF-beta: differential regulation in normal and tumour cells.
    BMC genomics, 2007, Apr-11, Volume: 8

    TGF-beta is one of the key cytokines implicated in various disease processes including cancer. TGF-beta inhibits growth and promotes apoptosis in normal epithelial cells and in contrast, acts as a pro-tumour cytokine by promoting tumour angiogenesis, immune-escape and metastasis. It is not clear if various actions of TGF-beta on normal and tumour cells are due to differential gene regulations. Hence we studied the regulation of gene expression by TGF-beta in normal and cancer cells.. Using human 19 K cDNA microarrays, we show that 1757 genes are exclusively regulated by TGF-beta in A549 cells in contrast to 733 genes exclusively regulated in HPL1D cells. In addition, 267 genes are commonly regulated in both the cell-lines. Semi-quantitative and real-time qRT-PCR analysis of some genes agrees with the microarray data. In order to identify the signalling pathways that influence TGF-beta mediated gene regulation, we used specific inhibitors of p38 MAP kinase, ERK kinase, JNK kinase and integrin signalling pathways. The data suggest that regulation of majority of the selected genes is dependent on at least one of these pathways and this dependence is cell-type specific. Interestingly, an integrin pathway inhibitor, RGD peptide, significantly affected TGF-beta regulation of Thrombospondin 1 in A549 cells.. These data suggest major differences with respect to TGF-beta mediated gene regulation in normal and transformed cells and significant role of non-canonical TGF-beta pathways in the regulation of many genes by TGF-beta.

    Topics: Cluster Analysis; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Integrins; Mitogen-Activated Protein Kinase Kinases; Neoplasms; Oligonucleotide Array Sequence Analysis; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured

2007
Induction of Smad1 by MT1-MMP contributes to tumor growth.
    International journal of cancer, 2007, Sep-01, Volume: 121, Issue:5

    MT1-MMP is a key integral membrane protease, which regulates tumor growth by cleaving extracellular matrix components, activating growth factors and receptors, and consequently, triggering downstream signals. To study what genes or pathways are mediated by endogenous MT1-MMP during tumor growth in vivo, we stably suppressed endogenous MT1-MMP in human tumor cells using RNA interference (RNAi). Tumor growth was significantly reduced in tumors derived from MT1-MMP-suppressed cells relative to control cells; the effect was rescued in cells engineered to re-express MT1-MMP expression. Gene expression profiling of cultured and tumor-derived cells by DNA microarray and real-time RT-PCR revealed that Smad1 expression was upregulated in MT1-MMP-expressing cells and rapidly growing tumors; this was confirmed in 4 additional tumor cell lines. Furthermore, tumor growth of MT1-MMP-expressing cells was reduced when Smad1 was suppressed by RNAi. We also found that the active form, but not the latent form, of TGF-beta was capable in promoting Smad1 expression and 3D cell proliferation in MT1-MMP-suppressed cells. In addition, a dominant-negative form of the TGF-beta Type II receptor reduced Smad1 expression in MT1-MMP-expressing cells. Thus, we propose that MT1-MMP functions, in part, to promote tumor growth by inducing the expression of Smad1 via TGF-beta signaling.

    Topics: Animals; Base Sequence; Cell Line, Tumor; Cell Proliferation; DNA Primers; Gene Expression Profiling; Humans; Matrix Metalloproteinase 14; Mice; Mice, Nude; Neoplasms; Oligonucleotide Array Sequence Analysis; Reverse Transcriptase Polymerase Chain Reaction; RNA Interference; RNA, Messenger; Smad1 Protein; Transforming Growth Factor beta

2007
Targeted TGF-beta chemotherapies: friend or foe in treating human malignancies?
    Expert review of anticancer therapy, 2007, Volume: 7, Issue:5

    Topics: Antineoplastic Agents; Humans; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2007
[Extracellular and cell-matrix interaction in pathology].
    Rossiiskii fiziologicheskii zhurnal imeni I.M. Sechenova, 2007, Volume: 93, Issue:5

    A lot of physiological processes including cell proliferation, cell migration, and differentiation are regulated by intercellular and cell-matrix interaction. The misbalance of intercellular and stroma-epithelial interaction is one of main factor of initiation of different pathological processes. Nonhealing wounds (chronic inflammation) and adenocarcinomas, despite the different external features, have many general inner features: cell proliferation, survival, cell migration, differentiation induced by cocktail of different growth factors and cytokines which promote inflammation and angiogenesis. Various stroma components including extracellular matrix are active participants of wound healing and cancer growth. The changes of pl-integrins distribution and cytokine expression, TGFbeta in particular, influence the development of pathological processes. It is possible to consider these factors as potential pharmacological targets.

    Topics: Epithelial Cells; Extracellular Matrix; Humans; Neoplasms; Neovascularization, Pathologic; Stromal Cells; Transforming Growth Factor beta; Wound Healing; Wounds and Injuries

2007
Effect of RNA oligonucleotide targeting Foxo-1 on muscle growth in normal and cancer cachexia mice.
    Cancer gene therapy, 2007, Volume: 14, Issue:12

    Foxo-1, a member of the Foxo forkhead type transcription factors, is markedly upregulated in skeletal muscle in energy-deprived states such as fasting, cancer and severe diabetes. In this study, we target the Foxo-1 mRNA in a mouse skeletal myoblast cell line C2C12 and in vivo models of normal and cancer cachexia mice by a Foxo-1 specific RNA oligonucleotide. Our results demonstrate that the RNA oligonucleotide can reduce the expression of Foxo-1 in cells and in normal and cachectic mice, leading to an increase in skeletal muscle mass of the mice. In search for the possible downstream target genes of Foxo-1, we show that when Foxo-1 expression is blocked both in cells and in mice, the level of MyoD, a myogenic factor, is increased while a muscle negative regulator GDF-8 or myostatin is suppressed. Taken together, these results show that Foxo-1 pays a critical role in development of muscle atrophy, and suggest that Foxo-1 is a potential molecular target for treatment of muscle wasting conditions.

    Topics: Animals; Cachexia; Cell Line, Tumor; Female; Forkhead Box Protein O1; Forkhead Transcription Factors; Gene Expression Regulation; Mice; Mice, Inbred BALB C; Muscle, Skeletal; Muscular Atrophy; Myoblasts; MyoD Protein; Myostatin; Neoplasms; Oligoribonucleotides, Antisense; Transforming Growth Factor beta

2007
CD16+ monocytes in human cutaneous leishmaniasis: increased ex vivo levels and correlation with clinical data.
    Journal of leukocyte biology, 2006, Volume: 79, Issue:1

    Peripheral blood CD16 (Fc receptor for immunoglobulin G III)-positive monocytes have been shown to expand in different pathological conditions, such as cancer, asthma, sepsis, human immunodeficiency virus infection, and AIDS progression, but data in leishmaniasis are lacking. We found that cutaneous leishmaniasis patients (n = 15) displayed a significant increase in the percentage (3.5 vs. 10.1) as well as mean fluorescent intensity (13.5 vs. 29.2) of ex vivo CD16 expression in monocytes as compared with healthy controls. We observed a significant positive correlation between the percentage of ex vivo CD16+ monocytes and lesion size (P = 0.0052, r = 0.75) or active transforming growth factor-beta plasma levels (P = 0.0017, r = 0.78). In addition, two patients with nonhealing lesions during a 3-year follow-up had high (9.1-19.4%) CD16 levels at diagnosis. Our data suggest a deleterious role for CD16 in human leishmaniasis, as well as its possible use as a marker for disease severity and/or adverse disease outcome.

    Topics: Acquired Immunodeficiency Syndrome; Adult; Antigens, CD; Biomarkers; Follow-Up Studies; Gene Expression Regulation; GPI-Linked Proteins; HIV; Humans; Leishmaniasis, Cutaneous; Male; Monocytes; Neoplasms; Receptors, IgG; Sepsis; Transforming Growth Factor beta

2006
CUTL1: a key mediator of TGFbeta-induced tumor invasion.
    Cell cycle (Georgetown, Tex.), 2006, Volume: 5, Issue:2

    The TGFbeta pathway plays a dual role in human carcinogenesis. On one hand, TGFbeta is well known for its ability to inhibit epithelial cell proliferation and promote apoptosis. However, many advanced cancers acquire resistance to the growth-inhibitory effects of TGFbeta and respond to it instead with promotion of proliferation, invasion and tumor progression. The homeobox transcription factor CUTL1, also known as CCAAT displacement protein, CDP or Cux-1, is involved in the control of normal embryonic development and differentiation. Recently, we found that CUTL1 is a transcriptional target of TGFbeta and is an important mediator of the TGFbeta-induced cell migration and invasion. In addition, CUTL1 is highly expressed in various epithelial cancers and seems to negatively correlate with tumor differentiation and patient survival. Therefore we postulate that CUTL1 might be a key mediator of the tumor-promoting effects of TGFbeta in advanced cancers.

    Topics: Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Neoplasm Invasiveness; Neoplasms; Nuclear Proteins; Repressor Proteins; Transcription Factors; Transforming Growth Factor beta

2006
Signal processing in the TGF-beta superfamily ligand-receptor network.
    PLoS computational biology, 2006, Volume: 2, Issue:1

    The TGF-beta pathway plays a central role in tissue homeostasis and morphogenesis. It transduces a variety of extracellular signals into intracellular transcriptional responses that control a plethora of cellular processes, including cell growth, apoptosis, and differentiation. We use computational modeling to show that coupling of signaling with receptor trafficking results in a highly versatile signal-processing unit, able to sense by itself absolute levels of ligand, temporal changes in ligand concentration, and ratios of multiple ligands. This coupling controls whether the response of the receptor module is transient or permanent and whether or not different signaling channels behave independently of each other. Our computational approach unifies seemingly disparate experimental observations and suggests specific changes in receptor trafficking patterns that can lead to phenotypes that favor tumor progression.

    Topics: Computer Simulation; Kinetics; Ligands; Models, Biological; Neoplasms; Protein Binding; Protein Transport; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2006
Lipoprotein(a), lipids and proinflammatory cytokines in patients undergoing major abdominal surgery.
    The British journal of surgery, 2006, Volume: 93, Issue:3

    The aims were to investigate whether surgical stress can induce a positive or negative lipoprotein(a) acute response, to determine any association with apolipoprotein(a) phenotypes, and to establish whether any such response is dependent on changes in lipids and proinflammatory cytokines. In addition, the impact of interleukin (IL) 6 genetic variability on the cytokine response to surgery was examined.. This prospective, observational study included 41 patients with cancer referred for abdominal surgery. Preoperative (T0) plasma concentrations of lipoprotein(a), IL-6, tumour necrosis factor alpha, and serum concentrations of transforming growth factor beta1 and lipids, were compared with values obtained 5 h (T1), 24 h (T2) and 5 days (T3) after surgery. Apolipoprotein(a) Kringle IV (KIV)-VNTR (variable-number tandem repeat) and IL-6 - 174 G/C polymorphisms were analysed.. Lipoprotein(a) was found to act as a negative acute-phase reactant (30.0 per cent reduction at T2) (P = 0.009). Surgery had a more profound impact on subjects with low KIV-VNTR. After surgery, lipoprotein(a) correlated significantly with corrected low-density lipoprotein (LDL)-cholesterol (r = 0.408 at T2). IL-6 inversely correlated with lipoprotein(a) (r = -0.321 at T1) and LDL-cholesterol (r = -0.418 at T1). The IL-6 response could be predicted from a combination of the surgical severity and -174 G/C genotype.. Although temporal associations did not indicate causality, these data provide a hypothesis to explain the inverse relationship between lipoprotein(a) and IL-6.

    Topics: Abdomen; Acute-Phase Reaction; Adult; Aged; Aged, 80 and over; Analysis of Variance; Apolipoproteins; Apoprotein(a); Enzyme-Linked Immunosorbent Assay; Female; Humans; Interleukin-6; Lipids; Lipoprotein(a); Male; Middle Aged; Neoplasms; Postoperative Complications; Prospective Studies; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Necrosis Factor-alpha

2006
TGFbeta in cancer and other disease - AACR special conference in cancer research.
    IDrugs : the investigational drugs journal, 2006, Volume: 9, Issue:4

    Topics: Animals; Antibodies, Monoclonal; Benzamides; Humans; Imatinib Mesylate; Neoplasms; Oligonucleotides, Antisense; Piperazines; Protein Kinase Inhibitors; Pyrimidines; Transforming Growth Factor beta; Transforming Growth Factor beta2

2006
Epigenetic inactivation of Betaig-h3 gene in human cancer cells.
    Cancer research, 2006, May-01, Volume: 66, Issue:9

    Gene silencing by CpG island methylation in the promoter region is one of the mechanisms by which tumor suppressor genes are inactivated in human cancers. It has been shown previously that Betaig-h3 gene, which encodes an extracellular matrix protein involved in cell adhesion and tumorigenesis, is down-regulated or silenced in a variety of human cancer cell lines. To unravel the underlying molecular mechanism(s) for this phenomenon, DNA methylation patterns of Betaig-h3 CpG island were examined in normal, immortalized, and cancer cell lines derived from lung, prostate, mammary, and kidney. A good correlation was observed between promoter hypermethylation and lost expression of Betaig-h3 gene, which was supported by the data that demethylation of promoter by 5-aza-2'-deoxycytidine reactivated Betaig-h3 and restored its expression in Betaig-h3-silenced tumor cell lines. This result was further substantiated by a luciferase reporter assay, showing the restoration of promoter activities and increased response to transforming growth factor-beta treatment in Betaig-h3-negative 293T cells when transfected with unmethylated Betaig-h3 promoter. In contrast, activity of Betaig-h3 promoter was completely inactivated by in vitro methylation. Furthermore, CpG methylation of Betaig-h3 promoter was also shown in primary lung tumors that expressed decreased level of Betaig-h3 protein. These results suggest that promoter methylation plays a critical role in promoter silencing of the Betaig-h3 gene in human tumor cells.

    Topics: Azacitidine; Breast Neoplasms; Carcinoma, Non-Small-Cell Lung; Cell Line, Tumor; CpG Islands; Decitabine; DNA Methylation; Epigenesis, Genetic; Extracellular Matrix Proteins; Female; Gene Expression Regulation, Neoplastic; Gene Silencing; Genes, Reporter; Humans; Luciferases; Lung Neoplasms; Male; Neoplasms; Promoter Regions, Genetic; Prostatic Neoplasms; Transforming Growth Factor beta

2006
Genetic predictors of adverse radiotherapy effects: the Gene-PARE project.
    International journal of radiation oncology, biology, physics, 2006, Jul-01, Volume: 65, Issue:3

    The development of adverse effects resulting from the radiotherapy of cancer limits the use of this treatment modality. The validation of a test capable of predicting which patients would be most likely to develop adverse responses to radiation treatment, based on the possession of specific genetic variants, would therefore be of value. The purpose of the Genetic Predictors of Adverse Radiotherapy Effects (Gene-PARE) project is to help achieve this goal.. A continuously expanding biorepository has been created consisting of frozen lymphocytes and DNA isolated from patients treated with radiotherapy. In conjunction with this biorepository, a database is maintained with detailed clinical information pertaining to diagnosis, treatment, and outcome. The DNA samples are screened using denaturing high performance liquid chromatography (DHPLC) and the Surveyor nuclease assay for variants in ATM, TGFB1, XRCC1, XRCC3, SOD2, and hHR21. It is anticipated that additional genes that control the biologic response to radiation will be screened in future work.. Evidence has been obtained that possession of variants in genes, the products of which play a role in radiation response, is predictive for the development of adverse effects after radiotherapy.. It is anticipated that the Gene-PARE project will yield information that will allow radiation oncologists to use genetic data to optimize treatment on an individual basis.

    Topics: Ataxia Telangiectasia Mutated Proteins; Cell Cycle Proteins; Chromatography, High Pressure Liquid; Deoxyribonuclease I; DNA-Binding Proteins; Humans; Mutation; Neoplasms; Nuclear Proteins; Phosphoproteins; Polymorphism, Single Nucleotide; Predictive Value of Tests; Protein Serine-Threonine Kinases; Radiation Injuries; Radiation Tolerance; Radiotherapy; Superoxide Dismutase; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Suppressor Proteins; X-ray Repair Cross Complementing Protein 1

2006
A mathematical model for the regulation of tumor dormancy based on enzyme kinetics.
    Bulletin of mathematical biology, 2006, Volume: 68, Issue:7

    In this paper we present a two-compartment model for tumor dormancy based on an idea of Zetter [1998, Ann. Rev. Med. 49, 407-422] to wit: The vascularization of a secondary (daughter) tumor can be suppressed by an inhibitor originating from a larger primary (mother) tumor. We apply this idea at the avascular level to develop a model for the remote suppression of secondary avascular tumors via the secretion of primary avascular tumor inhibitors. The model gives good agreement with the observations of [De Giorgi et al., 2003, Derm. Surgery 29, 664-667]. These authors reported on the emergence of a polypoid melanoma at a site remote from a primary polypoid melanoma after excision of the latter. The authors observed no recurrence of the melanoma at the primary site, but did observe secondary tumors at secondary sites 5-7 cm from the primary site within a period of 1 month after the excision of the primary site. We attempt to provide a reasonable biochemical/cell biological model for this phenomenon. We show that when the tumors are sufficiently remote, the primary tumor will not influence the secondary tumor while, if they are too close together, the primary tumor can effectively prevent the growth of the secondary tumor, even after it is removed. It should be possible to use the model as the basis for a testable hypothesis.

    Topics: Algorithms; Cell Movement; Cell Proliferation; Computer Simulation; Enzymes; Fibrinolysin; Fibroblast Growth Factor 2; Humans; Kinetics; Melanoma; Models, Biological; Neoplasm Metastasis; Neoplasms; Plasminogen; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator

2006
ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells.
    Cancer cell, 2006, Volume: 10, Issue:3

    Erk/MAPK and TGFbeta signaling cause epithelial to mesenchymal transition (EMT) and metastasis in mouse mammary epithelial cells (EpH4) transformed with oncogenic Ras (EpRas). In trials to unravel underlying mechanisms, expression profiling for EMT-specific genes identified a secreted interleukin-related protein (ILEI), upregulated exclusively at the translational level. Stable overexpression of ILEI in EpH4 and EpRas cells caused EMT, tumor growth, and metastasis, independent of TGFbeta-R signaling and enhanced by Bcl2. RNAi-mediated knockdown of ILEI in EpRas cells before and after EMT (EpRasXT) prevented and reverted TGFbeta-dependent EMT, also abrogating metastasis formation. ILEI is overexpressed and/or altered in intracellular localization in multiple human tumors, an event strongly correlated to invasion/EMT, metastasis formation, and survival in human colon and breast cancer.

    Topics: Animals; Cell Differentiation; Cell Line; Cell Transformation, Neoplastic; Cytokines; Epithelial Cells; Gene Expression Regulation, Neoplastic; Humans; Mesenchymal Stem Cells; Mice; Mice, Inbred BALB C; Neoplasm Metastasis; Neoplasm Proteins; Neoplasm Transplantation; Neoplasms; Prognosis; Protein Biosynthesis; RNA Interference; RNA, Messenger; Signal Transduction; Survival Rate; Time Factors; Transforming Growth Factor beta

2006
Epistatic interactions between modifier genes confer strain-specific redundancy for Tgfb1 in developmental angiogenesis.
    Genomics, 2005, Volume: 85, Issue:1

    Tgfbm1 (chromosome 5, P = 8 x 10(-5)) and Tgfbm3 (chromosome 12, P = 6 x 10(-11)) were identified as loci that modify developmental angiogenesis of Tgfb1 -/- mice. Congenic mice validated these loci and demonstrated epistatic interaction between them. The novel locus, Tgfbm3, encompasses approximately 22 genes, colocalizes with both tumor susceptibility and atherosclerosis susceptibility loci, and is enriched in genes regulating cell growth and morphogenesis. The use of gene knockout and/or transgenic mice that predispose to a complex trait, such as vascular development/angiogenesis, facilitates the identification of modifiers by simplifying genetic analysis. Identification of genes that modify response to lack of transforming growth factor beta1 (TGFbeta1) will enhance the understanding of TGFbeta1 action in vivo and may help predict which patients would respond well to anti-TGFbeta therapy. Identification of angiogenesis-modifying genes may provide new targets for angiogenesis therapies and analysis of polymorphisms therein may contribute to assessment of risk for diseases involving angiogenesis.

    Topics: Animals; Arteriosclerosis; Chromosomes; Epistasis, Genetic; Gene Expression Regulation, Developmental; Genetic Predisposition to Disease; Lod Score; Mice; Mice, Knockout; Neoplasms; Neovascularization, Physiologic; Quantitative Trait Loci; Species Specificity; Transforming Growth Factor beta; Transforming Growth Factor beta1; Transforming Growth Factor beta3

2005
Interleukin-7 deficiency in rheumatoid arthritis: consequences for therapy-induced lymphopenia.
    Arthritis research & therapy, 2005, Volume: 7, Issue:1

    We previously demonstrated prolonged, profound CD4+ T-lymphopenia in rheumatoid arthritis (RA) patients following lymphocyte-depleting therapy. Poor reconstitution could result either from reduced de novo T-cell production through the thymus or from poor peripheral expansion of residual T-cells. Interleukin-7 (IL-7) is known to stimulate the thymus to produce new T-cells and to allow circulating mature T-cells to expand, thereby playing a critical role in T-cell homeostasis. In the present study we demonstrated reduced levels of circulating IL-7 in a cross-section of RA patients. IL-7 production by bone marrow stromal cell cultures was also compromised in RA. To investigate whether such an IL-7 deficiency could account for the prolonged lymphopenia observed in RA following therapeutic lymphodepletion, we compared RA patients and patients with solid cancers treated with high-dose chemotherapy and autologous progenitor cell rescue. Chemotherapy rendered all patients similarly lymphopenic, but this was sustained in RA patients at 12 months, as compared with the reconstitution that occurred in cancer patients by 3-4 months. Both cohorts produced naive T-cells containing T-cell receptor excision circles. The main distinguishing feature between the groups was a failure to expand peripheral T-cells in RA, particularly memory cells during the first 3 months after treatment. Most importantly, there was no increase in serum IL-7 levels in RA, as compared with a fourfold rise in non-RA control individuals at the time of lymphopenia. Our data therefore suggest that RA patients are relatively IL-7 deficient and that this deficiency is likely to be an important contributing factor to poor early T-cell reconstitution in RA following therapeutic lymphodepletion. Furthermore, in RA patients with stable, well controlled disease, IL-7 levels were positively correlated with the T-cell receptor excision circle content of CD4+ T-cells, demonstrating a direct effect of IL-7 on thymic activity in this cohort.

    Topics: Alemtuzumab; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Antibodies, Neoplasm; Arthritis, Rheumatoid; Autoimmune Diseases; Blood Specimen Collection; Bone Marrow; CD4-Positive T-Lymphocytes; Cells, Cultured; Cohort Studies; Combined Modality Therapy; Cytokines; Gene Rearrangement, T-Lymphocyte; Humans; Interleukin-6; Interleukin-7; Lymphocyte Depletion; Lymphopenia; Lymphopoiesis; Neoplasms; Oncostatin M; Peripheral Blood Stem Cell Transplantation; Stromal Cells; Thymus Gland; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2005
Intratumor depletion of CD4+ cells unmasks tumor immunogenicity leading to the rejection of late-stage tumors.
    The Journal of experimental medicine, 2005, Mar-07, Volume: 201, Issue:5

    Tumor environment can be critical for preventing the immunological destruction of antigenic tumors. We have observed a selective accumulation of CD4(+)CD25(+) T cells inside tumors. In a murine fibrosarcoma L(d)-expressing Ag104, these cells made up the majority of tumor-infiltrating lymphocytes at the late stage of tumor progression, and their depletion during the effector phase, rather than priming phase, successfully enhanced antitumor immunity. We show here that CD4(+)CD25(+) T cells suppressed the proliferation and interferon-gamma production of CD8(+) T cells in vivo at the local tumor site. Blockade of the effects of IL-10 and TGF-beta partially reversed the suppression imposed by the CD4(+) cells. Furthermore, local depletion of CD4(+) cells inside the tumor resulted in a change of cytokine milieu and led to the eradication of well-established highly aggressive tumors and the development of long-term antitumor memory. Therefore, CD4(+)CD25(+) T cells maintained an environment in the tumor that concealed the immunogenicity of tumor cells to permit progressive growth of antigenic tumors. Our study illustrates that the suppression of antitumor immunity by regulatory T cells occurs predominantly at the tumor site, and that local reversal of suppression, even at a late stage of tumor development, can be an effective treatment for well-established cancers.

    Topics: Adoptive Transfer; Animals; Antigens, Neoplasm; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cell Proliferation; Cells, Cultured; Immune Tolerance; Interferon-gamma; Interleukin-10; Male; Mice; Neoplasm Staging; Neoplasms; Receptors, Interleukin-2; Transforming Growth Factor beta

2005
Transforming growth factor beta facilitates beta-TrCP-mediated degradation of Cdc25A in a Smad3-dependent manner.
    Molecular and cellular biology, 2005, Volume: 25, Issue:8

    Ubiquitin-dependent degradation of Cdc25A is a major mechanism for damage-induced S-phase checkpoint. Two ubiquitin ligases, the Skp1-cullin-beta-TrCP (SCFbeta-TrCP) complex and the anaphase-promoting complex (APCCdh1), are involved in Cdc25A degradation. Here we demonstrate that the transforming growth factor beta (TGF-beta)-Smad3 pathway promotes SCF(beta-TrCP)-mediated Cdc25A ubiquitination. Cells treated with TGF-beta, as well as cells transfected with Smad3 or a constitutively active type I TGF-beta receptor, exhibit increased ubiquitination and markedly shortened half-lives of Cdc25A. Furthermore, Cdc25A is stabilized in cells transfected with Smad3 small interfering RNA (siRNA) and cells from Smad3-null mice. TGF-beta-induced ubiquitination is associated with Cdc25A phosphorylation at the beta-TrCP docking site (DS82G motif) and physical association of Cdc25A with Smad3 and beta-TrCP. Cdc25A mutant proteins deficient in DS82G phosphorylation are resistant to TGF-beta-Smad3-induced degradation, whereas a Cdc25A mutant protein defective in APCCdh1 recognition undergoes efficient degradation. Smad3 siRNA inhibits beta-TrCP-Cdc25A interaction and Cdc25A degradation in response to TGF-beta. beta-TrCP2 siRNA also inhibits Smad3-induced Cdc25A degradation. In contrast, Cdh1 siRNA had no effect on Cdc25A down-regulation by Smad3. These data suggest that Smad3 plays a key role in the regulation of Cdc25A ubiquitination by SCFbeta-TrCP and that Cdc25A stabilization observed in various cancers could be associated with defects in the TGF-beta-Smad3 pathway.

    Topics: Animals; beta-Transducin Repeat-Containing Proteins; cdc25 Phosphatases; Cell Line, Tumor; DNA-Binding Proteins; Half-Life; Humans; Immunoprecipitation; Mice; Mutation; Neoplasms; Phosphorylation; Proteasome Endopeptidase Complex; Receptors, Transforming Growth Factor beta; RNA, Small Interfering; Smad3 Protein; Trans-Activators; Transfection; Transforming Growth Factor beta; Ubiquitin

2005
Differential expression and cytoplasm/membrane distribution of endoglin (CD105) in human tumour cell lines: Implications in the modulation of cell proliferation.
    International journal of oncology, 2005, Volume: 26, Issue:5

    Endoglin (CD105, an accessory component of the TGF-beta receptor complex) expression and distribution on different human tumour cells and its role in cellular proliferation were evaluated. We examined: 1) sixteen human carcinoma cell lines, 2) eight human sarcoma cell lines, 3) five miscellaneous tumour cell lines. HECV (endothelial cells) were employed as a positive control for endoglin expression. Normal Human Dermal Fibroblasts (NHDF) and 293 cells (epithelial kidney cells) were used as normal controls for connective and epithelial tissues, respectively. The results showed that CD105 was poorly expressed in the majority of human carcinoma cells (10/16), whereas it was highly expressed in most human sarcoma cells (7/8), and differently expressed by miscellaneous tumour cell lines. These data reflect endoglin expression by the normal counterparts of tumour cell lines, i.e. NHDF and 293 cells. However, CD105 levels in sarcoma cell lines, even though consistently lower than in NHDF, were significantly higher than those observed in carcinoma cells. Interestingly, CD105 presented a strong expression in the cytoplasm of MDA-MB-453 (breast carcinoma), NPA (papillary thyroid carcinoma), COLO-853 (melanoma) and SaOS-2 (osteosarcoma), but was weakly expressed on their cell membrane. This differential expression in the cytoplasm and on the membrane of some tumour cells, suggests a complex mechanism of translocation for this protein. The analysis of clonal growth in soft agar of some cell lines, characterized by high CD105 expression, showed an increased colony formation potential that was antagonized by the addition of anti-CD105 blocking mAb. The results indicated that endoglin is differentially expressed in human carcinoma and sarcoma cells and its overexpression modulates the proliferative rate of human solid tumour cells. Moreover, these data suggest that CD105 is involved in the regulation of TGF-beta effects in human solid malignancies, and therefore it could play an important role in tumour diagnosis and treatment.

    Topics: Antigens, CD; Carcinoma; Cell Membrane; Cell Proliferation; Cytoplasm; Endoglin; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Receptors, Cell Surface; Sarcoma; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation; Vascular Cell Adhesion Molecule-1

2005
Recent progress in defining mechanisms and potential targets for prevention of normal tissue injury after radiation therapy.
    International journal of radiation oncology, biology, physics, 2005, May-01, Volume: 62, Issue:1

    The ability to optimize treatments for cancer on the basis of relative risks for normal tissue injury has important implications in oncology, because higher doses of radiation might, in some diseases, improve both local control and survival. To achieve this goal, a thorough understanding of the molecular mechanisms responsible for radiation-induced toxicity will be essential. Recent research has demonstrated that ionizing radiation triggers a series of genetic and molecular events, which might lead to chronic persistent alterations in the microenvironment and an aberrant wound-healing response. Disrupted epithelial-stromal cell communication might also be important. With the application of a better understanding of fundamental biology to clinical practice, new approaches to treating and preventing normal tissue injury can focus on correcting these disturbed molecular processes.

    Topics: Cell Hypoxia; Cytokines; Endothelium, Vascular; Humans; Neoplasms; Radiation Injuries; Radiation Tolerance; Reactive Oxygen Species; Superoxide Dismutase; Transforming Growth Factor beta

2005
[Effect of transforming growth factor beta1 on cellular biological behavior in primary and recurrence ovarian cancer].
    Zhonghua yi xue za zhi, 2005, Feb-16, Volume: 85, Issue:6

    Topics: Cell Adhesion; Cell Proliferation; Female; Humans; Neoplasm Recurrence, Local; Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured

2005
Stability of frozen serum levels of insulin-like growth factor-I, insulin-like growth factor-II, insulin-like growth factor binding protein-3, transforming growth factor beta, soluble Fas, and superoxide dismutase activity for the JACC study.
    Journal of epidemiology, 2005, Volume: 15 Suppl 1

    Subjects of the Japan Collaborate Cohort Study (JACC Study) gave peripheral blood samples collected between 1988 and 1990. We conducted to investigate whether levels of serum components measured after 9 years of frozen storage are stable or not.. To assess the degradation of frozen serum components in the JACC Study, we compared levels of various components (IGF-I, IGF-II, IGFBP-3, TGF-beta 1, sFas, and total SOD activity) between fresh and stored sera collected from other inhabitants. Serum levels of constituents were measured by immunoradiometric assay (IGF-I, IGF-II and IGFBP-3), quantitative enzyme immunoassay (TGF-beta1), enzyme-linked immuno-adsorbent assay (sFas), and an improved nitrite method (SOD activity).. The coefficients of variation for intra- and inter-assay precisions of the measurements were less than 9%. Levels of IGF-I, IGF-II, IGFBP-3, TGF-beta 1 and sFas in sera after storage for 9 years at--80 degrees C were similar to those of fresh sera newly collected from inhabitants. The distributions of serum IGF-I, IGF-II, IGFBP-3, TGF-beta 1, sFas and SOD activity for specimens collected from different individuals tended to be similar to those of serum levels for frozen specimens collected from different individuals and stored for 9 years.. There was no statistically significant difference in distribution of measured values of IGF-I, IGF-II, IGFBP-3, TGF-beta 1, and sFas between newly collected sera and frozen specimens stored for 9 years. Thus, measurements of these serum constituents of specimens stored for the JACC Study can be reliably used in nested case-control study.

    Topics: Adult; Aged; Biomarkers; Cohort Studies; Enzyme-Linked Immunosorbent Assay; fas Receptor; Female; Freezing; Humans; Insulin-Like Growth Factor Binding Protein 3; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Japan; Male; Middle Aged; Neoplasms; Risk Factors; Specimen Handling; Superoxide Dismutase; Transforming Growth Factor beta; Transforming Growth Factor beta1

2005
Regulation of tumor angiogenesis by fastatin, the fourth FAS1 domain of betaig-h3, via alphavbeta3 integrin.
    Cancer research, 2005, May-15, Volume: 65, Issue:10

    We previously reported that the FAS1 domains of betaig-h3 bear motifs that mediate endothelial cell adhesion and migration via interactions with alphavbeta3 integrin and regulate angiogenesis. In the present study, we show that the fourth FAS1 domain, designated fastatin, inhibits endothelial adhesion and migration, not only to betaig-h3, but also fibronectin and vitronectin, in a RGD-dependent manner. Fastatin and other FAS1 domains suppress endothelial cell tube formation and in vivo neovascularization in a Matrigel plug assay. The antiangiogenic activity of fastatin is associated with antitumor activity in mouse tumor models. Fastatin additionally induces apoptosis in several cells expressing alphavbeta3 integrin, including endothelial cells. Binding of fastatin to alphavbeta3 integrin inhibits phosphorylation of focal adhesion kinase, Raf, extracellular signal-regulated kinase, Akt, and mammalian target of rapamycin. Fastatin is thus the first endogenous angiogenesis regulator identified that inhibits both endothelial cell migration and growth by binding to alphavbeta3 integrin. Our data suggest that FAS1 domains from all possible forms of the four human FAS1 family proteins are potential endogenous regulators for pathologic angiogenesis. Moreover, FAS1 domains such as fastatin may be developed into drugs for blocking tumor angiogenesis.

    Topics: Animals; Apoptosis; Cell Adhesion; Cell Movement; Endothelial Cells; Extracellular Matrix Proteins; fas Receptor; Humans; Integrin alphaVbeta3; Male; Melanoma, Experimental; Mice; Mice, Inbred C57BL; Neoplasms; Neovascularization, Pathologic; Peptide Fragments; Protein Structure, Tertiary; Receptors, Tumor Necrosis Factor; Transfection; Transforming Growth Factor beta

2005
Smad7 induces tumorigenicity by blocking TGF-beta-induced growth inhibition and apoptosis.
    Experimental cell research, 2005, Jul-01, Volume: 307, Issue:1

    Smad proteins play a key role in the intracellular signaling of the transforming growth factor beta (TGF-beta) superfamily of extracellular polypeptides that initiate signaling to regulate a wide variety of biological processes. The inhibitory Smad, Smad7, has been shown to function as intracellular antagonists of TGF-beta family signaling and is upregulated in several cancers. To determine the effect of Smad7-mediated blockade of TGF-beta signaling, we have stably expressed Smad7 in a TGF-beta-sensitive, well-differentiated, and non-tumorigenic cell line, FET, that was derived from human colon adenocarcinoma. Smad7 inhibits TGF-beta-induced transcriptional responses by blocking complex formation between Smad 2/3 and Smad4. While Smad7 has no effect on TGF-beta-induced activation of p38 MAPK and ERK, it blocks the phosphorylation of Akt by TGF-beta and enhances TGF-beta-induced phosphorylation of c-Jun. FET cells expressing Smad7 show anchorage-independent growth and enhance tumorigenicity in athymic nude mice. Smad7 blocks TGF-beta-induced growth inhibition by preventing TGF-beta-induced G1 arrest. Smad7 inhibits TGF-beta-mediated downregulation of c-Myc, CDK4, and Cyclin D1, and suppresses the expression of p21(Cip1). As a result, Smad7 inhibits TGF-beta-mediated downregulation of Rb phosphorylation. Furthermore, Smad7 inhibits the apoptosis of these cells. Together, Smad7 may increase the tumorigenicity of FET cells by blocking TGF-beta-induced growth inhibition and by inhibiting apoptosis. Thus, this study provides a mechanism by which a portion of human colorectal tumors may become refractory to tumor-suppressive actions of TGF-beta that might result in increased tumorigenicity.

    Topics: Animals; Apoptosis; Blotting, Western; Cell Line; Chlorocebus aethiops; COS Cells; DNA-Binding Proteins; Dose-Response Relationship, Drug; Gene Expression Regulation, Neoplastic; Growth Inhibitors; Humans; Mice; Mice, Nude; Neoplasm Transplantation; Neoplasms; Precipitin Tests; Retroviridae; Signal Transduction; Smad7 Protein; Trans-Activators; Transcription, Genetic; Transfection; Transforming Growth Factor beta; Transplantation, Heterologous

2005
Coevolution of cancer and stromal cellular responses.
    Cancer cell, 2005, Volume: 7, Issue:6

    It is now becoming apparent that multiple types of stromal cells, including macrophages, mast cells, adipocytes, and fibroblasts make pivotal contributions to carcinogenesis. In the May 6 issue of Cell, Orimo and colleagues (Orimo et al., 2005) show that carcinoma-associated fibroblasts can promote epithelial tumorigenesis by secreting the chemokine SDF-1alpha (CXCL12). SDF-1alpha stimulates carcinoma cell proliferation and recruitment of endothelial precursor cells.

    Topics: Animals; Calcium-Binding Proteins; Cell Movement; Chemokine CXCL12; Chemokines, CXC; Endothelial Cells; Fibroblasts; Humans; Leukocytes; Matrix Metalloproteinase 9; Mesenchymal Stem Cells; Models, Biological; Neoplasms; Neovascularization, Pathologic; S100 Calcium-Binding Protein A4; S100 Proteins; Stromal Cells; Transforming Growth Factor beta

2005
CUTL1 is a target of TGF(beta) signaling that enhances cancer cell motility and invasiveness.
    Cancer cell, 2005, Volume: 7, Issue:6

    CUTL1, also known as CDP, Cut, or Cux-1, is a homeodomain transcriptional regulator known to be involved in development and cell cycle progression. Here we report that CUTL1 activity is associated with increased migration and invasiveness in numerous tumor cell lines, both in vitro and in vivo. Furthermore, we identify CUTL1 as a transcriptional target of transforming growth factor beta and a mediator of its promigratory effects. CUTL1 activates a transcriptional program regulating genes involved in cell motility, invasion, and extracellular matrix composition. CUTL1 expression is significantly increased in high-grade carcinomas and is inversely correlated with survival in breast cancer. This suggests that CUTL1 plays a central role in coordinating a gene expression program associated with cell motility and tumor progression.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Movement; Disease-Free Survival; DNA-Binding Proteins; Down-Regulation; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasms; NIH 3T3 Cells; Nuclear Proteins; Oligonucleotide Array Sequence Analysis; p38 Mitogen-Activated Protein Kinases; Pancreatic Neoplasms; Repressor Proteins; RNA, Double-Stranded; Signal Transduction; Smad4 Protein; Trans-Activators; Transcription Factors; Transcription, Genetic; Transfection; Transforming Growth Factor beta; Up-Regulation

2005
[Expression of Tim-3 in tumor tissue and its role in the induction of tumor immune tolerance].
    Xi bao yu fen zi mian yi xue za zhi = Chinese journal of cellular and molecular immunology, 2005, Volume: 21, Issue:4

    To explore the role of T cell immunoglobulin- and mucin- domain containing molecule-3 (Tim-3) in the induction of tumor immune tolerance by investigating the expression of two types of Tim-3 at the early stage of tumorigenesis and the temporal-spatial relationship between the expression of Tim-3 and other immuno-inhibitory genes in tumor.. By using semi-quantitative RT-PCR method, we investigated the expression of Tim-3, forkhead box P3 (Foxp3), cytotoxic T lymphocyte antigen 4 (CTLA-4), glucocorticoid-induced tumor necrosis factor receptor family-related receptor (GITR), TGF-beta, IL-10 and IFN-gamma at different time in tumor of tumor-bearing mice. Meanwhile, the growth of tumor was determined.. At the early stage of tumorigenesis, sTim-3 was expressed before flTim-3 in tumor. When flTim-3 was expressed, the expression of sTim-3 was down-regulated. The expression of Foxp3, and GITR was simultaneous with that of flTim-3 and was up-regulated gradually with tumor growth. The expression of CTLA-4 was earlier than that of flTim-3 and also up-regulated gradually. The expression of flTim-3 and Foxp3 was also consistent with each other in tissue distribution. The growth of tumor was postively correlated with the expression of flTim-3, Foxp3, and CTLA-4 and negatively with the expression of sTim-3.. Along with the growth of tumor, immune tolerance was gradually established in tumor. flTim-3 but not sTim-3 may induce tumor immunological tolerance.

    Topics: Animals; Antigens, CD; Cell Line, Tumor; CTLA-4 Antigen; Female; Forkhead Transcription Factors; Gene Expression Regulation, Neoplastic; Hepatitis A Virus Cellular Receptor 2; Immune Tolerance; Immunization; Interferon-gamma; Interleukin-10; Membrane Proteins; Mice; Mice, Inbred BALB C; Neoplasms; Receptors, Virus; RNA, Messenger; Solubility; Transforming Growth Factor beta

2005
Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance.
    Nature, 2005, Sep-01, Volume: 437, Issue:7055

    The recognition and elimination of tumours by T cells, a process termed cancer immunosurveillance, is effective against certain virus-associated cancers. Spontaneous tumours often induce a specific immune response and are therefore also immunogenic. However, it is not clear whether they can be controlled by T cells. The immunosurveillance hypothesis postulates that tumours, if they eventually grow, escaped T-cell recognition by losing immunogenicity. Here we show, by generating a mouse model of sporadic cancer based on rare spontaneous activation of a dormant oncogene, that immunogenic tumours do not escape their recognition but induce tolerance. In this model, tumours derive from single cells and express a tumour-specific transplantation rejection antigen. Whereas vaccinated mice remain tumour-free throughout their lifetime, naive mice always develop a progressively growing tumour. We also show that despite specific recognition by T cells, the tumours do not lose their intrinsic immunogenicity and are rejected after transplantation in T-cell-competent recipients. Furthermore, in the primary host tumour-induced tolerance is associated with the expansion of non-functional T cells. Together, our data argue against immunosurveillance of spontaneous cancer.

    Topics: Animals; Antigens, Polyomavirus Transforming; Attachment Sites, Microbiological; Epigenesis, Genetic; Immune Tolerance; Integrases; Interferon-gamma; Lac Operon; Mice; Mice, Transgenic; Neoplasms; Oncogenes; T-Lymphocytes; Transforming Growth Factor beta; Transforming Growth Factor beta1; Viral Proteins

2005
Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation.
    The Journal of experimental medicine, 2005, Oct-03, Volume: 202, Issue:7

    The mechanisms through which regulatory T cells accumulate in lymphoid organs of tumor-bearing hosts remain elusive. Our experiments indicate that the accumulation of CD4+CD25+ regulatory T cells (T reg cells) expressing FoxP3 and exhibiting immunosuppressive function originates from the proliferation of naturally occurring CD25+ T cells and requires signaling through transforming growth factor (TGF)-beta receptor II. During tumor progression, a subset of dendritic cells (DCs) exhibiting a myeloid immature phenotype is recruited to draining lymph nodes. This DC subset selectively promotes the proliferation of T reg cells in a TGF-beta-dependent manner in mice and rats. Tumor cells are necessary and sufficient to convert DCs into regulatory cells that secrete bioactive TGF-beta and stimulate T reg cell proliferation. In conclusion, tumor expansion can stimulate T reg cells via a specific DC subset.

    Topics: Animals; Bromodeoxyuridine; Cell Differentiation; Cell Line, Tumor; Cell Proliferation; Dendritic Cells; DNA Primers; Forkhead Transcription Factors; Immunohistochemistry; Mice; Neoplasms; Rats; Rats, Inbred Strains; Receptors, Transforming Growth Factor beta; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2005
CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner.
    The Journal of experimental medicine, 2005, Oct-17, Volume: 202, Issue:8

    Tumor growth promotes the expansion of CD4+CD25+ regulatory T (T reg) cells that counteract T cell-mediated immune responses. An inverse correlation between natural killer (NK) cell activation and T reg cell expansion in tumor-bearing patients, shown here, prompted us to address the role of T reg cells in controlling innate antitumor immunity. Our experiments indicate that human T reg cells expressed membrane-bound transforming growth factor (TGF)-beta, which directly inhibited NK cell effector functions and down-regulated NKG2D receptors on the NK cell surface. Adoptive transfer of wild-type T reg cells but not TGF-beta-/- T reg cells into nude mice suppressed NK cell-mediated cytotoxicity, reduced NKG2D receptor expression, and accelerated the growth of tumors that are normally controlled by NK cells. Conversely, the depletion of mouse T reg cells exacerbated NK cell proliferation and cytotoxicity in vivo. Human NK cell-mediated tumor recognition could also be restored by depletion of T reg cells from tumor-infiltrating lymphocytes. These findings support a role for T reg cells in blunting the NK cell arm of the innate immune system.

    Topics: Animals; Cell Line, Tumor; Cell Proliferation; Cytokines; Cytotoxicity Tests, Immunologic; Flow Cytometry; France; Humans; Immunity, Innate; Killer Cells, Natural; Lymphocyte Activation; Mice; Mice, Inbred C57BL; Neoplasms; NK Cell Lectin-Like Receptor Subfamily K; Receptors, Immunologic; Receptors, Natural Killer Cell; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2005
Mucosal protection by cytokines.
    Current hematology reports, 2005, Volume: 4, Issue:6

    Oropharyngeal mucositis is a common, painful side effect seen in patients who receive aggressive chemotherapy, radiation, or both. Long thought to begin with direct epithelial cell damage, it now appears to be a complex process beginning in the submucosal endothelium and influenced by local cytokines. Previous attempts to use cytokines (including both topical and systemic transforming growth factor-beta, granulocyte- or granulocyte-macrophage colony-stimulating factors, and interleukin II) to prevent or treat mucositis were either inconclusive or, as in the case of interleukin II, associated with systemic toxicities. Recently palifermin (recombinant human keratinocyte growth factor, Kepivance [Amgen, Thousan Oaks, CA]) was approved as the first cytokine shown to decrease the incidence and duration of severe oral mucositis in patients with hematologic malignancies receiving myelotoxic therapy requiring hematopoietic stem cell support. Several other potentially effective agents are in clinical trials. After 40 years of painful mucositis, relief for patients finally is in sight.

    Topics: Antineoplastic Agents; Combined Modality Therapy; Cytokines; Granulocyte Colony-Stimulating Factor; Granulocyte-Macrophage Colony-Stimulating Factor; Hematopoietic Stem Cell Transplantation; Humans; Neoplasms; Risk Factors; Stomatitis; Transforming Growth Factor beta; Whole-Body Irradiation

2005
DNA methylation-associated inactivation of TGFbeta-related genes DRM/Gremlin, RUNX3, and HPP1 in human cancers.
    British journal of cancer, 2005, Oct-31, Volume: 93, Issue:9

    The transforming growth factor beta (TGFbeta)-signalling pathway is deregulated in many cancers. We examined the role of gene silencing via aberrant methylation of DRM/Gremlin and HPP1, which inhibit TGFbeta signalling, and RUNX3, which facilitates TGFbeta-signalling, of all genes that are thought to be tumour suppressors, are aberrantly expressed, and are thus thought to have important role in human cancers. We examined DRM/Gremlin mRNA expression in 44 cell lines and the promoter methylation status of DRM/Gremlin, HPP1, and RUNX3 in 44 cell lines and 511 primary tumours. The loss of DRM/Gremlin mRNA expression in human cancer cell lines is associated with DNA methylation, and treatment with the methylation inhibitor-reactivated mRNA expression (n=13). Methylation percentages of the three genes ranged from 0-83% in adult tumours and 0-50% in paediatric tumours. Methylation of DRM/Gremlin was more frequent in lung tumours in smokers, and methylation of all three genes was inversely correlated with prognosis in patients with bladder or prostate cancer. Our results provide strong evidence that these TGFbeta-related genes are frequently deregulated through aberrant methylation in many human malignancies.

    Topics: Aged; Core Binding Factor Alpha 3 Subunit; DNA Methylation; Down-Regulation; Female; Gene Expression Regulation, Neoplastic; Gene Silencing; Humans; Intercellular Signaling Peptides and Proteins; Male; Membrane Proteins; Neoplasm Proteins; Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured

2005
The ALK-5 inhibitor A-83-01 inhibits Smad signaling and epithelial-to-mesenchymal transition by transforming growth factor-beta.
    Cancer science, 2005, Volume: 96, Issue:11

    Transforming growth factor (TGF)-beta signaling facilitates tumor growth and metastasis in advanced cancer. Use of inhibitors of TGF-beta signaling may thus be a novel strategy for the treatment of patients with such cancer. In this study, we synthesized and characterized a small molecule inhibitor, A-83-01, which is structurally similar to previously reported ALK-5 inhibitors developed by Sawyer et al. (2003) and blocks signaling of type I serine/threonine kinase receptors for cytokines of the TGF-beta superfamily (known as activin receptor-like kinases; ALKs). Using a TGF-beta-responsive reporter construct in mammalian cells, we found that A-83-01 inhibited the transcriptional activity induced by TGF-beta type I receptor ALK-5 and that by activin type IB receptor ALK-4 and nodal type I receptor ALK-7, the kinase domains of which are structurally highly related to those of ALK-5. A-83-01 was found to be more potent in the inhibition of ALK5 than a previously described ALK-5 inhibitor, SB-431542, and also to prevent phosphorylation of Smad2/3 and the growth inhibition induced by TGF-beta. In contrast, A-83-01 had little or no effect on bone morphogenetic protein type I receptors, p38 mitogen-activated protein kinase, or extracellular regulated kinase. Consistent with these findings, A-83-01 inhibited the epithelial-to-mesenchymal transition induced by TGF-beta, suggesting that A-83-01 and related molecules may be useful for preventing the progression of advanced cancers.

    Topics: Activin Receptors, Type I; Animals; Humans; Lung; Mink; Neoplasms; Protein Serine-Threonine Kinases; Pyrazoles; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Thiocarbamates; Thiosemicarbazones; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured

2005
Analysis of host versus tumor interaction in cancer patients: opposing role of transforming growth factor-beta1 and interleukin-6 in the development of in situ tumor immunity.
    Immunobiology, 2005, Volume: 210, Issue:9

    A different degree of immunodeficiency is often found at tumor sites in cancer patients. At the late stage many patients develop malignant effusion that contains large numbers of tumor cells and host immune cells that constantly interact with each other. These sites may provide an ideal model to examine in situ anti-tumor immunity. The T cells in effusion were found to be immunodeficient, which suggested a defective anti-tumor cytotoxic T lymphocytes response. To pursue the mechanism for the T cell deficiency, we determined the production of immunomodulating cytokines in the effusion and detected the presence of transforming growth factor-beta1 (TGFbeta), prostaglandin E2, IL-6, IL-10, and IFNgamma. There was no detectable IL-2, IL-4, IL-12, or TNFalpha. The most prominent feature was the presence of TGFbeta and IL-6 at a very high level. Thus, the possible role of these two cytokines on T cell competence was further determined. TGFbeta was found to induce T cell anergy and reduced the production of perforin in T killer cells and their lytic activity. These events lead to the induction of peripheral T cell tolerance with profound T cell deficiency. IL-6 did not affect perforin production or cytolytic activity of the T killer cells. But the CD4+ CD25+ regulatory T cells (TR) that were often employed by TGFbeta to suppress T cell response were reduced in the malignant effusion, consistent with the fact that IL-6 down-regulates TR and this may represent the host's vigorous response to the tumor's subversion. These results show that TGFbeta and IL-6 might play pivotal but opposing roles in the host tumor interaction that, together with other immunomodulating components, determines the outcome for the development of local tumor immunity.

    Topics: Adult; Aged; Animals; Cells, Cultured; Female; Humans; Immunocompetence; Immunosuppression Therapy; Interleukin-6; Lymphocytes; Male; Membrane Glycoproteins; Mice; Mice, Inbred BALB C; Middle Aged; Neoplasms; Perforin; Pore Forming Cytotoxic Proteins; Transforming Growth Factor beta; Transforming Growth Factor beta1

2005
Cancer biology: emissaries set up new sites.
    Nature, 2005, Dec-08, Volume: 438, Issue:7069

    Topics: Animals; Cell Adhesion; Cell Movement; Fibronectins; Hematopoietic Stem Cells; Humans; Integrins; Mice; Neoplasm Metastasis; Neoplasms; Organ Specificity; Transforming Growth Factor beta; Vascular Endothelial Growth Factor Receptor-1

2005
Linking TGF-beta-mediated Cdc25A inhibition and cytoskeletal regulation through RhoA/p160(ROCK) signaling.
    Cell cycle (Georgetown, Tex.), 2004, Volume: 3, Issue:4

    Transforming growth factor-beta (TGF-beta) can mediate G(1)/S cell-cycle inhibition and changes in the cytoskeletal organization through multiple parallel downstream signaling pathways. Recent findings regarding TGF-beta-mediated cell-cycle checkpoint control and epithelial to mesenchymal transition have converged to the RhoA/p160(ROCK) signaling pathway. The activation of TGF-beta-mediated p160(ROCK)rapidly inhibits the Cdc25A phosphatase as a component of the G(1)/S checkpoint control at the time cytoskeletal re-organization occurs. This can be likened to the ability to preserve genomic integrity in circumstances of genotoxic stress. The inactivation of the RhoA/p160(ROCK) pathway may be a mechanism by which cancer cells bypass growth inhibition even in the presence of TGF-beta.

    Topics: Animals; cdc25 Phosphatases; Cell Cycle; Cell Division; Cytoskeleton; Disease Progression; G1 Phase; Humans; Intracellular Signaling Peptides and Proteins; Mice; Neoplasms; NIH 3T3 Cells; Phosphatidylinositol 3-Kinases; Phosphorylation; Protein Serine-Threonine Kinases; rho-Associated Kinases; rhoA GTP-Binding Protein; S Phase; Signal Transduction; Transforming Growth Factor beta

2004
beta-Catenin and TGFbeta signalling cooperate to maintain a mesenchymal phenotype after FosER-induced epithelial to mesenchymal transition.
    Oncogene, 2004, Apr-08, Volume: 23, Issue:15

    Several signalling pathways contribute to the regulation of epithelial to mesenchymal transition (EMT), either during developmentally regulated processes or in cancer progression and metastasis. Induction of EMT in fully polarized mouse mammary epithelial cells (EpH4) by an inducible c-fos estrogen receptor (FosER) oncoprotein involves loss of E-cadherin expression, nuclear translocation of beta-catenin, and autocrine production of TGFbeta. Reporter assays demonstrate that both beta-catenin/LEF-TCF- and TGFbeta-Smad-dependent signalling activities are upregulated, probably coregulating mesenchymal-specific gene expression during EMT. Stable expression of E-cadherin in mesenchymal FosER cells decreased beta-catenin activity and reduced cell proliferation. However, these cells still exhibited a defect in epithelial polarization and expressed E-cadherin/beta-catenin complexes in the entire plasma membrane. On the other hand, inhibition of TGFbeta-Smad signalling in mesenchymal FosER cells induced flat, cobblestone-like clusters of cells, which relocalized beta-catenin to the plasma membrane but still lacked detectable E-cadherin. Interestingly, inhibition of TGFbeta signalling in the E-cadherin-expressing mesenchymal FosER cells caused their reversion to a polarized epithelial phenotype, in which E-cadherin, beta-catenin, and ZO-1 were localized at their correct lateral plasma membrane domains. These results demonstrate that loss of E-cadherin can contribute to increased LEF/TCF-beta-catenin signalling, which in turn cooperates with autocrine TGFbeta signalling to maintain an undifferentiated mesenchymal phenotype.

    Topics: Active Transport, Cell Nucleus; Animals; beta Catenin; Cadherins; Cell Adhesion; Cell Differentiation; Cell Division; Cell Line; Cell Membrane; Cell Nucleus; Cytoskeletal Proteins; Disease Progression; DNA, Complementary; Enzyme-Linked Immunosorbent Assay; Epithelium; Genes, Reporter; Mesoderm; Mice; Microscopy, Fluorescence; Neoplasms; Phenotype; Protein Isoforms; Protein Structure, Tertiary; Proto-Oncogene Proteins c-fos; Receptors, Estrogen; Reverse Transcriptase Polymerase Chain Reaction; RNA; Signal Transduction; Trans-Activators; Transcription, Genetic; Transcriptional Activation; Transfection; Transforming Growth Factor beta; Up-Regulation

2004
Regulation of large-scale chromatin unfolding by Smad4.
    Biochemical and biophysical research communications, 2004, Mar-05, Volume: 315, Issue:2

    The tumor suppressor Smad4 plays a critical role in the transforming growth factor-beta (TGF-beta signaling pathway. Smad4 is an essential component of transcriptional complexes mediating the activation of Smad-dependent target genes. However, how Smad4 regulates various chromosomal events remains unknown. Here, we report that Smad4 induces large-scale chromatin unfolding by targeting Smad4 to an amplified, lac operator-containing chromosome region in mammalian cells. This unfolding activity maps to the subdomains within the transactivation domain of Smad4. Both the N-terminal missense and the C-terminal truncated tumor-derived mutations increased the chromatin unfolding activity of Smad4. These data identify a novel function of Smad4 and suggest that regulation of large-scale chromatin unfolding by Smad4 may be responsible for cancer development.

    Topics: Alleles; Animals; Blotting, Western; Cell Line; CHO Cells; Chromatin; Cricetinae; DNA-Binding Proteins; Humans; Luciferases; Mutation; Mutation, Missense; Neoplasms; Plasmids; Protein Denaturation; Protein Folding; Protein Structure, Tertiary; Signal Transduction; Smad4 Protein; Trans-Activators; Transcriptional Activation; Transfection; Transforming Growth Factor beta

2004
Suppression of macrophage function in AK-5 tumor transplanted animals: role of TGF-beta1.
    Immunology letters, 2004, Feb-15, Volume: 91, Issue:2-3

    Transforming growth factor-beta 1 (TGF-beta1), a multifunctional cytokine secreted by various cell types has been implicated in diverse physiologic and pathophysiologic functions, including immunological, inflammatory, and neoplastic processes. AK-5 tumor cells when injected intraperitoneally grow as ascites, causing 100% mortality. Peritoneal macrophages cocultured with AK-5 cells, in vitro showed specific suppression of tumoricidal function. We report that AK-5 cells secrete the latent form of TGF-beta1, which is converted to its active homodimeric form, causing suppression of the secretion of cytocidal molecule by macrophages thereby inhibiting their cytotoxic capabilities. When macrophages are cocultured with AK-5 cells in the presence of antibodies against TGF-beta1 or the receptor II for TGF-beta, there is significant recovery in the secretion of nitric oxide and macrophage cytotoxic potential. These results suggest a major role for TGF-beta produced by the tumor cells, in the immune escape mechanisms adopted by the AK-5 tumor cells.

    Topics: Animals; Antibodies; Cell Line, Tumor; Coculture Techniques; Macrophages, Peritoneal; Neoplasm Transplantation; Neoplasms; Nitric Oxide; Nitric Oxide Synthase; Nitric Oxide Synthase Type II; Nitrogen Oxides; Phagocytosis; Rats; Rats, Wistar; Superoxides; Transforming Growth Factor beta; Transforming Growth Factor beta1; Transforming Growth Factor beta2

2004
Antitumor histone deacetylase inhibitors suppress cutaneous radiation syndrome: Implications for increasing therapeutic gain in cancer radiotherapy.
    Molecular cancer therapeutics, 2004, Volume: 3, Issue:3

    Radiotherapy is an effective treatment for head and neck, skin, anogenital, and breast cancers. However, radiation-induced skin morbidity limits the therapeutic benefits. A low-toxicity approach to selectively reduce skin morbidity without compromising tumor killing by radiotherapy is needed. We found that the antitumor agents known as histone deacetylase (HDAC) inhibitors (phenylbutyrate, trichostatin A, and valproic acid) could suppress cutaneous radiation syndrome. The effects of HDAC inhibitors in promoting the healing of wounds caused by radiation and in decreasing later skin fibrosis and tumorigenesis were correlated with suppression of the aberrant expression of radiation-induced transforming growth factor beta and tumor necrosis factor alpha. Our findings implicate that the inhibition of HDAC may provide a novel strategy to increase the therapeutic gain in cancer radiotherapy by not only inhibiting tumor growth but also protecting normal tissues.

    Topics: Acetylation; Animals; Antineoplastic Agents; Blotting, Northern; Blotting, Western; Cell Line, Tumor; Enzyme Inhibitors; Female; Fibrosis; Histone Deacetylase Inhibitors; Histones; Humans; Hydroxamic Acids; Immunohistochemistry; Mice; Mice, Inbred BALB C; Neoplasm Transplantation; Neoplasms; Phenylbutyrates; Radiation Injuries; Radiotherapy; Rats; Rats, Sprague-Dawley; Ribonucleases; Skin; Time Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Valproic Acid; Wound Healing

2004
Cancer: dangerous liaisons.
    Nature, 2004, Mar-18, Volume: 428, Issue:6980

    Topics: Animals; Cell Division; Epithelial Cells; Hepatocyte Growth Factor; Mice; Neoplasms; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2004
Bone morphogenetic protein-2 stimulates angiogenesis in developing tumors.
    Molecular cancer research : MCR, 2004, Volume: 2, Issue:3

    Bone Morphogenetic Protein-2 (BMP-2) is highly overexpressed in the majority of patient-derived lung carcinomas. However, a mechanism revealing its role in cancer has not been established. Here we report that BMP-2 enhances the neovascularization of developing tumors. Recombinant BMP-2 stimulated blood vessel formation in tumors formed from A549 cells injected s.c. into thymic nude mice. Recombinant BMP-2 also enhanced angiogenesis in Matrigel plugs containing A549 cells in nude mice. The BMP-2 antagonist noggin abrogated BMP-2-induced angiogenic response. Furthermore, antisense transfection of BMP-2 cDNA resulted in a decrease in blood vessel formation in the Matrigel assays. BMP-2 induced tube formation in both human aortic endothelial cells (HAEC) and umbilical vein endothelial cells. BMP-2 also stimulated proliferation of HAEC. The ability of BMP-2 to activate endothelial cells was further demonstrated by its ability to phosphorylate Smad 1/5/8 and ERK-1/2 and to increase expression of Id1. This study reveals that BMP-2 enhanced the angiogenic response in developing tumors. Furthermore, these data suggest that BMP-2 stimulation of angiogenesis may involve the activation of endothelial cells.

    Topics: Active Transport, Cell Nucleus; Animals; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Cell Division; Cell Line; Cell Line, Tumor; Cell Nucleus; Collagen; DNA-Binding Proteins; Drug Combinations; Endothelial Cells; Enzyme Activation; Humans; Inhibitor of Differentiation Protein 1; Laminin; Mice; Mice, Nude; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Morphogenesis; Neoplasms; Neovascularization, Pathologic; Proteoglycans; Recombinant Proteins; Repressor Proteins; Smad Proteins; Smad1 Protein; Trans-Activators; Transcription Factors; Transforming Growth Factor beta

2004
Secreted transforming growth factor beta2 activates NF-kappaB, blocks apoptosis, and is essential for the survival of some tumor cells.
    Proceedings of the National Academy of Sciences of the United States of America, 2004, May-04, Volume: 101, Issue:18

    The basis of constitutive activation of NF-kappaB, essential for survival and resistance to apoptosis in many tumors, is not well understood. We find that transforming growth factor beta2 (TGFbeta2), predominantly in its latent form, is secreted by several different types of tumor cell lines that exhibit constitutively active NF-kappaB and that TGFbeta2 potently stimulates the activation of NF-kappaB in reporter cells. Suppression of TGFbeta2 expression by small interfering RNA kills prostate cancer PC3 cells, indicating that the TGFbeta2-NF-kappaB pathway is important for their viability. These findings identify TGFbeta2 as a potential target for therapeutic strategies to inhibit the growth of tumor cells that depend on constitutively active NF-kappaB, or to sensitize them to treatment with cytotoxic drugs.

    Topics: Apoptosis; Culture Media, Conditioned; Humans; In Vitro Techniques; Male; Neoplasms; NF-kappa B; Prostatic Neoplasms; Transforming Growth Factor beta; Transforming Growth Factor beta2; Tumor Cells, Cultured

2004
Snail and E47 repressors of E-cadherin induce distinct invasive and angiogenic properties in vivo.
    Journal of cell science, 2004, Jun-01, Volume: 117, Issue:Pt 13

    The transcription factors Snail and E47 are direct repressors of E-cadherin, with both inducing a full epithelial-mesenchymal transition and invasive behaviour in vitro when expressed in the prototypic epithelial MDCK cell line. The role of these repressors in the invasive process and in other tumorigenic properties is, nevertheless, still poorly understood. However, organotypic cultures and in vivo transplantation assays indicate that cells expressing MDCK-Snail and MDCK-E47 exhibit significant differences. MDCK-Snail cells have a higher infiltrative potential than MDCK-E47 cells. Interestingly, both cell types induce angiogenesis of the host stromal tissue in transplantation assays, but this property is greatly enhanced in transplants of MDCK-E47 cells. Xenografted tumours induced in nude mice also show signs of strong angiogenic potential, again markedly increased in tumours induced by MDCK-E47 which exhibit a higher vessel density and proliferation rate than those induced by MDCK-Snail cells. These results suggest differential roles for Snail and E47 E-cadherin repressors in tumour progression where Snail is implicated in promoting the initial invasion and E47 plays an active role in tumour cell growth by promoting angiogenesis.

    Topics: Actins; Animals; Cadherins; Cell Division; Cell Line; DNA-Binding Proteins; Dogs; Extracellular Matrix Proteins; Immunohistochemistry; Kinetics; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Transplantation; Neoplasms; Neovascularization, Pathologic; Platelet Endothelial Cell Adhesion Molecule-1; Repressor Proteins; RNA, Messenger; Snail Family Transcription Factors; TCF Transcription Factors; Transcription Factor 7-Like 1 Protein; Transcription Factors; Transforming Growth Factor beta; Transplantation, Heterologous; Vascular Endothelial Growth Factor A

2004
Interleukin-1beta system in anorectic catabolic tumor-bearing rats.
    Current opinion in clinical nutrition and metabolic care, 2004, Volume: 7, Issue:4

    The onset of cancer anorexia and the accompanying neurological symptoms and signs involve the general influence of cytokines on the brain. Using methylcholanthrene to induce tumors in Fischer 344 rats, we measured various specific components of the cytokine-induced anorectic reaction, including: (1) IL-1beta system components (ligand, signaling receptor, receptor accessory proteins, and receptor antagonist); (2) TNF-alpha; (3) TGF-beta1; and (4) IFN-gamma in the tumor tissue, the liver and the brain.. The data show that IL-1beta, TNF-alpha and IFN-gamma messenger RNA were detected in the tumor tissue of anorectic tumor-bearing rats. In brain regions, anorexia is associated with the upregulation of IL-1beta and its receptor mRNA. All other mRNA remained unchanged in the brain regions examined.. This suggests that IL-1beta and its receptor may play a significant role in this model of cancer-associated anorexia. In vivo, the characterization of cytokine components in the brain may provide data for potential pharmacological interventions to ameliorate the anorexia of disease.

    Topics: Animals; Anorexia; Brain; Cachexia; Cytokines; Interferon-gamma; Interleukin-1; Liver; Male; Neoplasms; Rats; Rats, Inbred F344; RNA, Messenger; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2004
CAR is a cell-cell adhesion protein in human cancer cells and is expressionally modulated by dexamethasone, TNFalpha, and TGFbeta.
    Gene therapy, 2003, Volume: 10, Issue:3

    The coxsackie adenovirus receptor (CAR) has become of interest for gene therapy due to its crucial function in adenoviral cell entry. In clinical trials with adenoviral vectors, dexamethasone is applied to reduce side effects such as inflammatory reactions or emesis. By using a beta-galactosidase-expressing adenovirus (AdGal), we observed that dexamethasone treatment resulted in decreased adenoviral gene transfer into human cancer cells. Expression of CAR and integrin alpha5beta1 was transcriptionally downregulated by dexamethasone as shown for HeLa cervical cancer cells and U87MG glioblastoma cells. TNFalpha increased CAR expression in HeLa and ovarian cancer cells but decreased CAR expression in U87MG cells. In all tested cancer cell lines, TNFalpha induced a significant increase in the expression of adenovirus-binding integrins alpha5beta1, alphavbeta3 and alphavbeta5. Pretreatment with TNFalpha increased AdGal gene transfer into cancer cells and enhanced the cytotoxic effect of a p53-expressing adenovirus. In contrast, TGFbeta reduced CAR expression level and adenoviral gene transfer into OV-UL-2 ovarian cancer cells. Confocal immunofluorescence analysis revealed localization of CAR at cell-cell adhesions in several human cancer cell lines and disruption of cell-cell contacts increased adenoviral gene transfer into human cancer cells. In clinical cancer gene therapy, efficiency of adenoviral gene delivery could be altered by cell adhesion, TNFalpha, TGFbeta, and dexamethasone.

    Topics: Adenoviridae; Anti-Inflammatory Agents; beta-Galactosidase; Calcium-Binding Proteins; Cell Adhesion; Cytokines; Dexamethasone; Eye Proteins; Female; Flow Cytometry; Fluorescent Antibody Technique; Gene Expression; Gene Expression Regulation; Genetic Therapy; Genetic Vectors; HeLa Cells; Hippocalcin; Humans; Integrin alpha5beta1; Integrin alphaVbeta3; Integrins; Lipoproteins; Neoplasms; Nerve Tissue Proteins; Receptors, Vitronectin; Recoverin; Reverse Transcriptase Polymerase Chain Reaction; Transduction, Genetic; Transforming Growth Factor beta; Transgenes; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

2003
p53 and TGF-beta in development: prelude to tumor suppression?
    Cell, 2003, May-02, Volume: 113, Issue:3

    Recent work in Xenopus embryos reveals an unexpected developmental role for the tumor suppressor gene p53. This finding may have implications for the evolution of p53, its interaction with Smads in TGF-beta dependent mesoderm specification, and the cooperation among p53 family members.

    Topics: Animals; DNA-Binding Proteins; Neoplasms; Smad Proteins; Trans-Activators; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Xenopus

2003
Decreased tumorigenicity in vivo when transforming growth factor beta treatment causes cancer cell senescence.
    Bioscience, biotechnology, and biochemistry, 2003, Volume: 67, Issue:4

    We have previously reported that transforming growth factor beta (TGF-beta) triggers two independent senescence programs, 1) replicative senescence dependent upon telomere shortening and 2) premature senescence independent of telomere shortening, in the cell line of A549 human lung adenocarcinoma. In this study, we examined the possibility that cancer cell tumor phenotypes could be suppressed by forced senescence. We used A549 cells treated with TGF-beta for a long time (over 50 days), where senescence was induced in a telomere-shortening-dependent or an independent way. Fully senescent A549 cells were elongated, acquired contact inhibition capabilities when reaching confluence, and secreted the senescence-associated cytokine IL-6. Furthermore, senescent A549 cells had no tumorigenicity in nude mice. These results indicate that the forced induction of senescence in cancer cells may be a novel and potentially powerful method for advancing anti-cancer therapy.

    Topics: Animals; Antineoplastic Agents; Cell Division; Cellular Senescence; Contact Inhibition; DNA-Binding Proteins; Humans; Interleukin-6; Mice; Mice, Nude; Neoplasm Transplantation; Neoplasms; Neoplasms, Experimental; Smad Proteins; Telomere; Trans-Activators; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
TIAF1 participates in the transforming growth factor beta1--mediated growth regulation.
    Annals of the New York Academy of Sciences, 2003, Volume: 995

    TGF-beta induces growth suppression and apoptosis of various types of cells, but supports fibroblast growth. We previously isolated TIAF1 (TGF-beta1-induced antiapoptotic factor 1), which protects murine L929 fibroblasts from TNF cytotoxicity. Here, we show that TIAF1 induced growth inhibition and apoptosis of monocytic U937 and other types of cells. In contrast, like TGF-beta1, TIAF1 supported transforming growth of L929 fibroblasts. TIAF1 increased the expression of p53, Cip1/p21, and Smad proteins; suppressed ERK phosphorylation; and altered TGF-beta1-mediated Smad2/3 phosphorylation in U937 cells. Antisense TIAF1 mRNA significantly enhanced the proliferation of mink lung Mv1Lu epithelial cells. Together, these observations indicate that TIAF1 participates in the TGF-beta-mediated growth regulation.

    Topics: Animals; Apoptosis; Apoptosis Regulatory Proteins; Carrier Proteins; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA-Binding Proteins; Fibroblasts; HL-60 Cells; Humans; Mice; Myosin Heavy Chains; Neoplasms; Nuclear Proteins; Signal Transduction; Smad2 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; U937 Cells

2003
VDUP1 upregulated by TGF-beta1 and 1,25-dihydorxyvitamin D3 inhibits tumor cell growth by blocking cell-cycle progression.
    Oncogene, 2003, Jun-26, Volume: 22, Issue:26

    Vitamin D(3) upregulated protein 1 (VDUP1) is a 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) upregulated protein, and it is induced by various stresses. In human tumor tissues, VDUP1 expression was downregulated. Upon stimulation by growth-inhibitory signals such as TGF-beta1 and 1,25(OH)(2)D(3), its expression was rapidly upregulated as the cell growth was retarded. The transfection of VDUP1 in tumor cells reduced cell growth. The VDUP1 expression was also increased when the cell-cycle progression was arrested. Transfection of VDUP1 induced cell-cycle arrest at the G0/G1 phase, indicating that VDUP1 possesses a tumor-suppressive activity. In addition, it was found that VDUP1 interacted with promyelocytic leukemia zinc-finger, Fanconi anemia zinc-finger, and histone deacetylase 1, which are known to be transcriptional corepressors. VDUP1 itself suppressed IL-3 receptor and cyclin A2 promoter activity. Taken together, these results suggest that VDUP1 is a novel antitumor gene which forms a transcriptional repressor complex.

    Topics: 3T3 Cells; Animals; Blotting, Northern; Calcitriol; Carrier Proteins; Cell Cycle; Cell Differentiation; Cell Division; Cell Line; Cyclin A; Cyclin A2; Disease Progression; DNA-Binding Proteins; Fanconi Anemia; Flow Cytometry; G1 Phase; Genes, Reporter; Histone Deacetylase 1; Histone Deacetylases; HL-60 Cells; Humans; In Situ Hybridization; Interleukin-3; Jurkat Cells; Kruppel-Like Transcription Factors; Mice; Microscopy, Fluorescence; Neoplasms; Promoter Regions, Genetic; Promyelocytic Leukemia Zinc Finger Protein; Repressor Proteins; Resting Phase, Cell Cycle; Retroviridae; Reverse Transcriptase Polymerase Chain Reaction; Thioredoxins; Time Factors; Transcription Factors; Transcription, Genetic; Transfection; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Up-Regulation; Zinc Fingers

2003
Tacrolimus enhances transforming growth factor-beta1 expression and promotes tumor progression.
    Transplantation, 2003, Aug-15, Volume: 76, Issue:3

    Immunosuppressive therapy is a risk factor for the increased incidence and metastatic progression of malignancies in organ graft recipients. Transforming growth factor (TGF)-beta(1) has been associated with tumor invasion and metastasis, and we have implicated cyclosporine-associated TGF-beta(1) hyperexpression in tumor progression in mice.. BALB/c mice or severe combined immunodeficient-beige mice were treated with 2 or 4 mg/kg of tacrolimus, and the effect of treatment on mouse renal cancer cell pulmonary metastasis was investigated. We also determined whether tacrolimus induces TGF-beta(1) expression. Spleens from tacrolimus-treated mice were analyzed for level of expression of TGF-beta(1) mRNA with the use of competitive-quantitative polymerase chain reaction assay, and circulating levels of TGF-beta(1) protein were measured with the use of an enzyme-linked immunosorbent assay.. Treatment with tacrolimus resulted in a dose-dependent increase in the number of pulmonary metastases in the BALB/c mice (197+/-16 in untreated mice, 281+/-26 in mice treated with 2 mg/kg of tacrolimus, and 339+/-25 in mice treated with 4 mg/kg of tacrolimus; no treatment vs. 4 mg/kg tacrolimus, Bonferroni's P<0.001) and in the severe combined immunodeficient-beige mice (117+/-18 in untreated mice, 137+/-19 in mice treated with 2 mg/kg of tacrolimus, and 216+/-29 in mice treated with 4 mg/kg of tacrolimus; no treatment vs. 4 mg/kg tacrolimus, P<0.05). Treatment with 4 mg/kg but not 2 mg/kg of tacrolimus resulted in a significant increase in the levels of expression of TGF-beta(1) mRNA and circulating levels of TGF-beta(1) protein.. Tacrolimus has a dose-dependent effect on tumor progression and TGF-beta(1) expression, and tacrolimus-induced TGF-beta(1) overexpression may be a pathogenetic mechanism in tumor progression.

    Topics: Animals; Carcinoma, Renal Cell; Disease Progression; Dose-Response Relationship, Drug; Immunosuppressive Agents; Kidney Neoplasms; Lung Neoplasms; Male; Mice; Mice, Inbred BALB C; Mice, SCID; Neoplasm Metastasis; Neoplasms; Spleen; Tacrolimus; Transforming Growth Factor beta; Transforming Growth Factor beta1

2003
Building better blood vessels: new insight on the molecular control of arteriogenesis.
    Cardiovascular research, 2003, Sep-01, Volume: 59, Issue:3

    Topics: Animals; Arterial Occlusive Diseases; Arteries; Carbonic Anhydrases; Collateral Circulation; DNA-Binding Proteins; Early Growth Response Protein 1; Endothelial Cells; Gene Expression; Humans; Immediate-Early Proteins; MAP Kinase Signaling System; Mice; Neoplasms; Neovascularization, Pathologic; Nerve Tissue Proteins; Stress, Mechanical; Transcription Factors; Transforming Growth Factor beta; Tunica Intima

2003
[Study on the angiogenic ability of tumor fibroblasts].
    Zhonghua gan zang bing za zhi = Zhonghua ganzangbing zazhi = Chinese journal of hepatology, 2003, Volume: 11, Issue:12

    To explore the role of fibroblasts derived from tumor in the tumor angiogenesis.. Two-well co-culture system were used to detect the expression of MMP-9, TGF-beta1, TN and bcl-2 in L929-H22 cells, and their ability of promoting angiogenesis of ECV304 cells and invasion of MDA-MB-231 cells respectively, which were established in our laboratory before. Then their adhesion and the effect of their supernatant on H22 cells proliferation were analysed.. Compared with L929 cells, the adhesion potential of L929-H22 cells increased (F>or=104.32, P<0.001), with the higher level of expression of MMP-9, bcl-2, TN, and TGF-beta1 in L929-H22 cells in creased (t>or=3.3055, P<0.01). L929-H22 and L929 cells enhanced the invasiveness of human mammary cancer MDA-MB-231 cells through artificial basement membrane (Matrigel) 1.21 and 0.48 times respectively (F=266.3, P<0.001). L929-H22 cells induced morphogenesis of ECV304 cells. L929-H22 stimulated endothelial cells to form more and longer tubes than L929 did (F>or=23.75, P<0.01). 25% CM of L929-H22 cells stimulated the growth of H22 cells (F=266.30, P<0.05).. The results suggested that fibroblasts in tumors secrete more growth factors and angiogenic factors to promote the angiogenesis and invasion of solid tumors.

    Topics: Animals; Cell Line, Tumor; Fibroblasts; Humans; Matrix Metalloproteinase 9; Mice; Neoplasm Invasiveness; Neoplasms; Neovascularization, Pathologic; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor beta

2003
An in vitro tumor model: analysis of angiogenic factor expression after chemotherapy.
    Cancer research, 2002, Oct-01, Volume: 62, Issue:19

    Tumor tissues include malignant cells and a stroma made up of mainly inflammatory cells, endothelial cells, and fibroblasts. To differentiate the effects of treatment on angiogenic cytokine secretion in tumor tissue, exponential and stationary phase human CaKi-1 renal cell carcinoma cells, human SW2 small cell lung carcinoma cells, human umbilical vein endothelial cells (HUVECs), murine NIH-3T3 fibroblasts, and murine RAW264.7 macrophages were exposed to gemcitabine, paclitaxel, carboplatin, and the protein kinase Cbeta inhibitor LY317615, and secretion (24 h) of tumor necrosis factor-alpha, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), and transforming growth factor (TGF)-beta was determined by a Luminex FlowMetrix assay. After 72 h of exposure, exponential RAW, 3T3, and SW2 cells were sensitive to gemcitabine; exponential and stationary SW2 and HUVECs were sensitive to paclitaxel; and exponential and stationary HUVECs were most sensitive to LY317615. None of the cells secreted detectable tumor necrosis factor-alpha. Generally, exponential cells secreted higher levels of cytokines than stationary cells (stationary cells secreted approximately 10 times less TGF-beta). Only malignant cells secreted VEGF (80-300 pg/10(6) cells). VEGF secretion by exponential SW2 cells decreased in an anticancer agent concentration-dependent manner. Every cell type secreted TGF-beta (40-700 pg/10(6) cells). Exponential 3T3, RAW, CaKi-1, and SW2 cells secreted the most TGF-beta, and levels did not decrease with treatment. Only CaKi-1, SW2, and HUVECs secreted bFGF (0.5-50 pg/10(6) cells). CaKi-1 cells increased secretion of bFGF with therapy. Although malignant cells alone secreted VEGF, stromal cells secreted TGF-beta and bFGF at levels comparable with or greater than malignant cells and thus may be important contributors to tumor growth and progression.

    Topics: 3T3 Cells; Animals; Carboplatin; Cytokines; Deoxycytidine; Drug Screening Assays, Antitumor; Endothelial Growth Factors; Fibroblast Growth Factor 2; Gemcitabine; Humans; Intercellular Signaling Peptides and Proteins; Lymphokines; Mice; Neoplasms; Paclitaxel; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2002
Activated human CD4+ T cells induced by dendritic cell stimulation are most sensitive to transforming growth factor-beta: implications for dendritic cell immunization against cancer.
    Clinical immunology (Orlando, Fla.), 2002, Volume: 102, Issue:1

    The secretion of immunosuppressive factors like transforming growth factor-beta (TGF-beta) by tumor cells has been recognized as one of the mechanisms involved in tumor immunological escape. This study aimed to examine whether dendritic cell (DC) immunization could reverse TGF-beta-induced immunosuppression by simulating the in vivo interaction among infused DCs, host T cells, and tumor-secreted TGF-beta in an in vitro study. We found that both immature and mature DCs were relatively resistant to TGF-beta. The addition of TGF-beta to naive human CD4+ T cells, which are required by genetically modified DC to elicit antitumor immunity, resulted in their hyporesponsiveness to DC stimulation in a dose-dependent manner. When activated by allogeneic DCs in the presence of TGF-beta, CD4+ T cells displayed a reduced capacity to proliferate. More importantly, activated CD4+ T cells induced by DC stimulation were very sensitive to TGF-beta, and this susceptibility was enhanced by their previous exposure to TGF-beta. The underlying mechanism was linked to TGF-beta-induced apoptosis of activated T cells. However, the presence of stimulation from DC or antibodies to CD3 plus CD28 could partly reverse the immunosuppressive effect of TGF-beta on activated CD4+ T cells. Taken together, our results indicate that the efficacy of DC immunization may be impaired by tumor-derived TGF-beta.

    Topics: Antigen Presentation; CD28 Antigens; CD3 Complex; CD4-Positive T-Lymphocytes; Cell Differentiation; Clonal Anergy; Dendritic Cells; Humans; Immune Tolerance; Lymphocyte Activation; Neoplasms; Transforming Growth Factor beta

2002
Modifying normal tissue damage postirradiation. Report of a workshop sponsored by the Radiation Research Program, National Cancer Institute, Bethesda, Maryland, September 6-8, 2000.
    Radiation research, 2002, Volume: 157, Issue:2

    Late effects that develop in normal tissues adjacent to the tumor site in the months to years after radiotherapy can reduce the quality of life of cancer survivors. They can be dose-limiting and debilitating or life-threatening. There is now evidence that some late effects may be preventable or partially reversible. A workshop, "Modifying Normal Tissue Damage Postirradiation", was sponsored by the Radiation Research Program of the National Cancer Institute to identify the current status of and research needs and opportunities in this area. Mechanistic, genetic and physiological studies of the development of late effects are needed and will provide a rational basis for development of treatments. Interdisciplinary teams will be needed to carry out this research, including pathologists, physiologists, geneticists, molecular biologists, experts in functional imaging, wound healing, burn injury, molecular biology, and medical oncology, in addition to radiation biologists, physicists and oncologists. The participants emphasized the need for developing and choosing appropriate models, and for radiation dose-response studies to determine whether interventions remain effective at the radiation doses used clinically. Both preclinical and clinical studies require long-term follow-up, and easier-to-use, more objective clinical scoring systems must be developed and standardized. New developments in biomedical imaging should provide useful tools in all these endeavors. The ultimate goals are to improve the quality of life and efficacy of treatment for cancer patients treated with radiotherapy.

    Topics: Animals; Cell Hypoxia; Chemokines; Cytokines; Diagnostic Imaging; Endopeptidases; Extracellular Matrix; Growth Substances; Humans; Hyperbaric Oxygenation; Mice; Mice, Inbred C3H; Mice, Inbred C57BL; Models, Animal; Neoplasms; Primates; Proto-Oncogenes; Quality of Life; Radiation Injuries; Radiation Injuries, Experimental; Radiation Tolerance; Radiotherapy; Rats; Receptors, Growth Factor; Renin-Angiotensin System; Severity of Illness Index; Swine; Time Factors; Transforming Growth Factor beta

2002
HOXD3 enhances motility and invasiveness through the TGF-beta-dependent and -independent pathways in A549 cells.
    Oncogene, 2002, Jan-24, Volume: 21, Issue:5

    Homeobox genes regulate sets of genes that determine cellular fates in embryonic morphogenesis and maintenance of adult tissue architecture by regulating cellular motility and cell-cell interactions. Our previous studies showed that a specific member, HOXD3, when overexpressed, upregulates integrin beta3 expression in human erythroleukemia HEL cells and lung cancer A549 cells, and enhances their motility and invasiveness. We performed a microarray study of over 7075 genes to determine the mechanisms underlying the HOXD3-enhanced motility and invasiveness in A549 cells. RT-PCR-based tracking gene analyses highlighted a set of TGF-beta-upregulated genes, which included matrix metalloproteinase-2, syndecan-1, CD44, and TGF-beta-induced 68 kDa protein. Exogenous TGF-beta also caused this pattern of upregulation in A549 cells and enhanced their migratory and invasive activity, confirming the involvement of TGF-beta signaling. However, HOXD3 reduced the expression of TGF-beta-independent genes coding for desmosomal components such as desmoglein, desmoplakin and plakoglobin which are known to suppress tumor invasion and metastasis. These results suggest that HOXD3 enhances the invasive and metastatic potential of cancer cells through the TGF-beta-dependent and -independent pathways.

    Topics: Cell Movement; DNA-Binding Proteins; Down-Regulation; Gene Expression Profiling; Homeodomain Proteins; Neoplasm Invasiveness; Neoplasms; Oligonucleotide Array Sequence Analysis; RNA, Neoplasm; Signal Transduction; Transcription Factors; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation

2002
Role of TGF-beta in oncogenesis.
    Microscopy research and technique, 2001, Feb-15, Volume: 52, Issue:4

    Topics: Animals; Humans; Mice; Neoplasms; Signal Transduction; Transforming Growth Factor beta

2001
Interleukin-10-deficient mice and inflammatory bowel disease associated cancer development.
    Carcinogenesis, 2001, Volume: 22, Issue:4

    Interleukin-10-deficient mice develop colitis and colorectal cancer similar to the inflammatory bowel disease associated cancer in humans. The aim of this study was to identify possible mutations of oncogenes and tumour suppressor genes involved in tumorigenesis in Interleukin-10 (IL-10)-deficient mice. Twenty colon carcinomas from IL-10-deficient mice were screened for mutations in the K-ras and p53 genes by 'cold' single-strand-conformation polymorphism. Immunohistochemical staining was performed to detect mutations in the proteins P53, APC and MSH2, and the transforming growth factor beta type II receptor. Microsatellite instability was analysed at eight chromosomal loci and plasma levels of transforming growth factor beta1 (TGF-beta1) were also measured. At 9 weeks, 14% of the animals developed colorectal cancer, and at 10-31 weeks the incidence of carcinoma was 65%. No mutations were detected in the analysed oncogene and tumour suppressor genes. Plasma TGF-beta1 levels in IL-10-deficient mice 10-31 weeks old were higher than in wild-type littermates e.g. 45.7 +/- 4.6 ng/ml versus 19.8 +/- 4.5 ng/ml (P<0.01). No alterations in K-ras, p53, APC: and Msh2 genes suggests that other genes are involved in the development of these tumours. Elevated TGF-beta1 plasma levels correspond to the high incidence of dysplasia and cancer. Normal expression of the TGF-beta II receptors hints at genetic alterations in other members of the TGF-beta receptor signal transduction pathway.

    Topics: Adenomatous Polyposis Coli Protein; Animals; Carcinoma; Colonic Neoplasms; Colorectal Neoplasms; Cytoskeletal Proteins; DNA-Binding Proteins; Genes, p53; Genes, ras; Humans; Immunohistochemistry; Inflammatory Bowel Diseases; Interleukin-10; Intestine, Large; Intestine, Small; Mice; Mice, Inbred C57BL; Microsatellite Repeats; Mutation; MutS Homolog 2 Protein; Neoplasms; Polymorphism, Single-Stranded Conformational; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Sequence Analysis, DNA; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factor beta1

2001
Re: Role of transforming growth factor-beta signaling in cancer.
    Journal of the National Cancer Institute, 2001, Apr-04, Volume: 93, Issue:7

    Topics: Animals; Antigens, CD; Endoglin; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Neovascularization, Pathologic; Receptors, Cell Surface; Signal Transduction; Transforming Growth Factor beta; Vascular Cell Adhesion Molecule-1

2001
CD3-mediated activation of tumor-reactive lymphocytes from patients with advanced cancer.
    Proceedings of the National Academy of Sciences of the United States of America, 2001, Jun-05, Volume: 98, Issue:12

    Lymphocytes from blood or tumors of patients with advanced cancer did not proliferate and produced very low levels of tumor necrosis factor and IFN-gamma when cultured with autologous tumor cells. Proliferation and lymphokine production dramatically increased in the presence of beads conjugated with mAbs to CD3 plus mAbs to CD28 and/or CD40, and the lymphocytes destroyed the tumor cells. Expression density of CD3 concomitantly increased from low to normal levels. Furthermore, beads providing a CD3 signal (in combination with CD28 or CD28 plus CD40) gave partial protection against the inhibitory effect of transforming growth factor type beta1 on lymphocyte proliferation and production of tumor necrosis factor and IFN-gamma. MHC class I-restricted cytolytic T cells lysing autologous tumor cells in a 4-h Cr(51) release assay were generated when peripheral blood leukocytes were activated in the presence of autologous tumor cells and anti-CD3/CD28 or anti-CD3/CD28/CD40 beads. Experiments performed in a model system using anti-V-beta1 or anti-V-beta2 mAbs to activate subsets of T cells expressing restricted T cell receptor showed that lymphocytes previously activated by anti-V-beta can respond to CD3 stimulation with vigorous proliferation and lymphokine production while retaining their specificity, also in the presence of transforming growth factor type beta1. Our results suggest that T lymphocytes from cancer patients can proliferate and form Th1 type lymphokines in the presence of autologous tumor cell when properly activated, and that antigen released from killed tumor cells and presented by antigen-presenting cells in the cultures facilitates the selective expansion of tumor-directed, CD8(+) cytolytic T cells.

    Topics: Antibodies, Monoclonal; CD28 Antigens; CD3 Complex; CD40 Antigens; Coculture Techniques; Histocompatibility Antigens Class I; Humans; Interferon-gamma; Lymphocyte Activation; Lymphocytes, Tumor-Infiltrating; Neoplasms; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2001
In vivo mechanisms by which tumors producing thrombospondin 1 bypass its inhibitory effects.
    Genes & development, 2001, Jun-01, Volume: 15, Issue:11

    Thrombospondin 1 (TSP1) is a multifunctional protein able to activate TGFbeta and to inhibit angiogenesis in vivo. Although usually thought of as an inhibitor of tumor growth, TSP1 may sometimes be present at high levels during tumor progression, suggesting that tumors can eventually overcome their anti-tumor effects. Using a tet-repressible expression system, we demonstrate that murine TSP1 delayed the onset of tumor growth when produced in the tumor bed by rat fibrosarcoma tumor cells or by stromal fibroblasts coinjected with unmodified C6 glioma tumor cells. Yet upon prolonged exposure to TSP1, tumors came to grow at the same rate in the presence as in the absence of TSP1 and transplantation experiments showed that they had become insensitive to inhibition by TSP1 in both syngeneic and immune compromised hosts. Tumor resistance to TSP1 developed as a result of the in vivo outgrowth of pre-existing tumor cell variants that (1) secreted increased amounts of angiogenic factors that counterbalanced the inhibitory effect of TSP1 on neovascularization and (2) grew more efficiently in the presence of TSP1-activated TGFbeta. These results indicate that prolonged and continuous local delivery of a single multifunctional angiogenesis inhibitor like TSP1 to fast-growing tumors can lead to tumor resistance in vivo by fostering the outgrowth of subpopulations that are a by-product of the genetic instability of the tumor cells themselves.

    Topics: Angiogenesis Inhibitors; Animals; Blotting, Northern; Fibrosarcoma; Glioblastoma; Immunoblotting; Immunohistochemistry; Mice; Mice, Nude; Neoplasm Transplantation; Neoplasms; Neovascularization, Pathologic; Rats; Rats, Inbred F344; Thrombospondin 1; Transforming Growth Factor beta; Tumor Cells, Cultured

2001
The retinoblastoma protein acts as a transcriptional coactivator required for osteogenic differentiation.
    Molecular cell, 2001, Volume: 8, Issue:2

    The incidence of osteosarcoma is increased 500-fold in patients who inherit mutations in the RB gene. To understand why the retinoblastoma protein (pRb) is specifically targeted in osteosarcoma, we studied its function in osteogenesis. Loss of pRb but not p107 or p130 blocks late osteoblast differentiation. pRb physically interacts with the osteoblast transcription factor, CBFA1, and associates with osteoblast-specific promoters in vivo in a CBFA1-dependent fashion. Association of pRb with CBFA1 and promoter sequences results in synergistic transactivation of an osteoblast-specific reporter. This transactivation function is lost in tumor-derived pRb mutants, underscoring a potential role in tumor suppression. Thus, pRb functions as a direct transcriptional coactivator promoting osteoblast differentiation, which may contribute to the targeting of pRb in osteosarcoma.

    Topics: Animals; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Cell Differentiation; Cells, Cultured; Core Binding Factor Alpha 1 Subunit; Genes, Reporter; Genes, Retinoblastoma; Humans; Mice; Mice, Knockout; Mutagenesis, Site-Directed; Neoplasm Proteins; Neoplasms; Oncogene Proteins, Viral; Osteoblasts; Osteocalcin; Osteogenesis; Osteosarcoma; Papillomavirus E7 Proteins; Promoter Regions, Genetic; Protein Binding; Recombinant Fusion Proteins; Recombinant Proteins; Retinoblastoma Protein; Reverse Transcriptase Polymerase Chain Reaction; Transcription Factors; Transcription, Genetic; Transcriptional Activation; Transforming Growth Factor beta

2001
RhoB is dispensable for mouse development, but it modifies susceptibility to tumor formation as well as cell adhesion and growth factor signaling in transformed cells.
    Molecular and cellular biology, 2001, Volume: 21, Issue:20

    RhoB is an endosomal small GTPase that is implicated in the response to growth factors, genotoxic stress, and farnesyltransferase inhibitors. To gain insight into its physiological functions we examined the consequences of homozygous gene deletion in the mouse. Loss of RhoB did not adversely affect mouse development, fertility, or wound healing. However, embryo fibroblasts cultured in vitro exhibited a defect in motility, suggesting that RhoB has a role in this process that is conditional on cell stress. Neoplastic transformation by adenovirus E1A and mutant Ras yielded differences in cell attachment and spreading that were not apparent in primary cells. In addition, transformed -/- cells displayed altered actin and proliferative responses to transforming growth factor beta. A negative modifier role in transformation was suggested by the increased susceptibility of -/- mice to 7,12-dimethylbenz[a]anthracene-induced skin carcinogenesis and by the increased efficiency of intraperitoneal tumor formation by -/- cells. Our findings suggest that RhoB is a negative regulator of integrin and growth factor signals that are involved in neoplastic transformation and possibly other stress or disease states.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Actins; Animals; Blotting, Western; Carcinogens; Cell Adhesion; Cell Line, Transformed; Dose-Response Relationship, Drug; Fibroblasts; Fibronectins; Gene Deletion; Genetic Predisposition to Disease; Growth Substances; Homozygote; Mice; Mice, Transgenic; Models, Genetic; Neoplasms; rhoB GTP-Binding Protein; Signal Transduction; Skin Neoplasms; Time Factors; Transforming Growth Factor beta

2001
Crystal structure of a phosphorylated Smad2. Recognition of phosphoserine by the MH2 domain and insights on Smad function in TGF-beta signaling.
    Molecular cell, 2001, Volume: 8, Issue:6

    Ligand-induced phosphorylation of the receptor-regulated Smads (R-Smads) is essential in the receptor Ser/Thr kinase-mediated TGF-beta signaling. The crystal structure of a phosphorylated Smad2, at 1.8 A resolution, reveals the formation of a homotrimer mediated by the C-terminal phosphoserine (pSer) residues. The pSer binding surface on the MH2 domain, frequently targeted for inactivation in cancers, is highly conserved among the Co- and R-Smads. This finding, together with mutagenesis data, pinpoints a functional interface between Smad2 and Smad4. In addition, the pSer binding surface on the MH2 domain coincides with the surface on R-Smads that is required for docking interactions with the serine-phosphorylated receptor kinases. These observations define a bifunctional role for the MH2 domain as a pSer-X-pSer binding module in receptor Ser/Thr kinase signaling pathways.

    Topics: Amino Acid Motifs; Amino Acid Sequence; Animals; Binding Sites; Crystallography, X-Ray; DNA-Binding Proteins; Humans; Models, Biological; Models, Molecular; Molecular Sequence Data; Mutation; Neoplasms; Phosphorylation; Phosphoserine; Protein Serine-Threonine Kinases; Protein Structure, Quaternary; Protein Structure, Tertiary; Sequence Alignment; Signal Transduction; Smad2 Protein; Smad4 Protein; Structure-Activity Relationship; Trans-Activators; Transforming Growth Factor beta

2001
Elevated levels of angiogenic cytokines in the plasma of cancer patients.
    International journal of cancer, 2000, Jan-01, Volume: 85, Issue:1

    Although in the normal healthy organism angiogenesis is a tightly regulated process, under a variety of circumstances it may contribute to disease states. These include the growth of solid tumors, the hematogenous spread of tumor cells and the growth of metastasis. Our aim was to measure the levels of 5 angiogenic cytokines in the plasma of patients with a variety of cancers, to establish a plasmatic angiogenic profile. We prospectively obtained blood samples in citrated tubes from 40 healthy individuals and 75 patients with a variety of solid tumors. Patients who had received any form of treatment in the preceeding 6 months were excluded from the study. Plasma levels of the following 5 cytokines were determined by ELISA: vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), basic fibroblast growth factor, transforming growth factor-beta and tumor necrosis factor-alpha. In some cases, additional samples were taken 4 and 15 days after surgical removal of the tumor. Our findings demonstrate, that firstly, compared to the tumor group VEGF was almost always undetectable or present at very low levels in healthy individuals; secondly, a threshold value for HGF was found to exist between the 2 groups (healthy vs. tumor); and thirdly, there was a clear relationship between plasma levels of VEGF and HGF and extension of disease (i.e., without or with metastases). The timing of blood sampling in the post-operative period was found to be critical, particularly with regard to VEGF and HGF. The existence of a systemic angiogenic profile in the plasma of cancer patients may be useful as a diagnostic and prognostic tool and may help in the future to monitor the responses of individual patients to anti-tumor and, particularly, anti-angiogenic therapy.

    Topics: Adult; Cytokines; Endothelial Growth Factors; Fibroblast Growth Factor 2; Hepatocyte Growth Factor; Humans; Lymphokines; Middle Aged; Neoplasms; Neovascularization, Pathologic; Prospective Studies; Reference Values; Time Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2000
Keystone Symposia on Molecular and Cellular Biology: 'The molecular basis of cancer'. Organizers: Carol L. Prives and George F. Vande Woude, Taos, NM, 15-21 March 1999.
    Biochimica et biophysica acta, 2000, Feb-14, Volume: 1470, Issue:1

    The Keystone Symposium on the Molecular Basis of Cancer was an excellent meeting, which stimulated the exchange of a great deal of information. This report was prepared to organize some of the results that provided new insights into the regulation of cell proliferation and apoptosis. We were unable to report on all of the talks and posters due mostly to our limited capacity to absorb and digest the large amount of results presented at the meeting. We apologize to those whose results were not covered in this report.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Apoptosis; Carrier Proteins; Cell Cycle; Cell Cycle Proteins; Cytoplasm; DNA Repair; DNA Replication; DNA-Binding Proteins; E2F Transcription Factors; Genes, Tumor Suppressor; Humans; Mice; MutL Protein Homolog 1; Neoplasm Proteins; Neoplasms; NF-kappa B; Nuclear Proteins; Proteins; Proto-Oncogene Proteins c-myc; Replication Protein A; Retinoblastoma-Binding Protein 1; Saccharomyces cerevisiae; Transcription Factor DP1; Transcription Factors; Transforming Growth Factor beta; Tumor Protein p73; Tumor Suppressor Proteins

2000
An acidic microenvironment impairs the generation of non-major histocompatibility complex-restricted killer cells.
    Immunology, 2000, Volume: 99, Issue:3

    The microenvironment within solid tumours has often been shown to exhibit an acidic local pH. In recent studies we could demonstrate that an acidic extracellular pH (pHe) inhibits the non-major histocompatibility complex (MHC) -restricted cytotoxicity of immunocompetent effector cells. However, within tumours the activation of cytotoxic cells may already be impaired by low pHe. Therefore, we investigated the influence of acidic conditions on the generation of active killer cells. The cytotoxic activity of natural killer (NK) as well as lymphokine-activated killer (LAK) cells against K562, Daudi and Raji cells was analysed after an activation period of 3 days at pHe 7.2-6.5. A minor reduction of pHe from 7.2 to 7.0 during the culture period resulted in a strong inhibition of the natural cytotoxicity of NK cells. Furthermore, acidic pHe below 7.2 prevented the generation of activated LAK cells by interleukin-2 (IL-2). The cytotoxic capacity could not be reconstituted if cells cultured at a pHe of 6.5 were returned to physiological pH for another 24 hr. Analysis of the cellular subtypes within the various cultures did not reveal differences regarding the frequencies of NK cells, CD8+ T cells, or CD4+ T cells. However, an acidic pHe clearly inhibited the activation-induced increase of relevant adhesion molecules. The production of cytokines which are involved in the regulation of the cytotoxic process (tumour necrosis factor-alpha, interferon-gamma, IL-10, IL-12 and transforming growth factor-beta1) was also affected by pHe, as their release was strongly inhibited at pHe 7.0. Furthermore, we observed a considerable decrease in the metabolic activity of effector cells at acidic pHe. In summary, our findings suggest that an acidic microenvironment impairs the induction of an anti-tumoral immune response within solid tumours.

    Topics: CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cell Line; Culture Media; Cytokines; Cytotoxicity Tests, Immunologic; Erythroblasts; Extracellular Space; Flow Cytometry; Humans; Hydrogen-Ion Concentration; Interferon-gamma; Interleukin-10; Interleukin-12; Interleukin-2; Killer Cells, Lymphokine-Activated; Killer Cells, Natural; Lymphocyte Activation; Neoplasms; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2000
Immunosuppression mediated by tumor cells: a challenge for immunotherapeutic approaches.
    Human gene therapy, 2000, Mar-20, Volume: 11, Issue:5

    Topics: Animals; Granulocyte-Macrophage Colony-Stimulating Factor; Immune Tolerance; Immunotherapy; Interleukin-10; Neoplasms; Neoplasms, Experimental; T-Lymphocytes; Transforming Growth Factor beta

2000
Role of transforming growth factor-beta signaling in cancer.
    Journal of the National Cancer Institute, 2000, Sep-06, Volume: 92, Issue:17

    Signaling from transforming growth factor-beta (TGF-beta) through its unique transmembrane receptor serine-threonine kinases plays a complex role in carcinogenesis, having both tumor suppressor and oncogenic activities. Tumor cells often escape from the antiproliferative effects of TGF-beta by mutational inactivation or dysregulated expression of components in its signaling pathway. Decreased receptor function and altered ratios of the TGF-beta type I and type II receptors found in many tumor cells compromise the tumor suppressor activities of TGF-beta and enable its oncogenic functions. Recent identification of a family of intracellular mediators, the Smads, has provided new paradigms for understanding mechanisms of subversion of TGF-beta signaling by tumor cells. In addition, several proteins recently have been identified that can modulate the Smad-signaling pathway and may also be targets for mutation in cancer. Other pathways such as various mitogen-activated protein kinase cascades also contribute substantially to TGF-beta signaling. Understanding the interplay between these signaling cascades as well as the complex patterns of cross-talk with other signaling pathways is an important area of investigation that will ultimately contribute to understanding of the bifunctional tumor suppressor/oncogene role of TGF-beta in carcinogenesis.

    Topics: Animals; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Humans; Mutation; Neoplasms; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad2 Protein; Smad3 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2000
Networks of tumor suppressors. Workshop: tumor suppressor networks.
    EMBO reports, 2000, Volume: 1, Issue:2

    Topics: Animals; beta Catenin; Cell Cycle Proteins; Cell Hypoxia; Cytoskeletal Proteins; Disease Models, Animal; DNA Damage; Genes, Tumor Suppressor; Humans; Models, Genetic; Neoplasms; Oncogenes; Signal Transduction; Trans-Activators; Transforming Growth Factor beta

2000
Genetic control of the circulating concentration of transforming growth factor type beta1.
    Human molecular genetics, 1999, Volume: 8, Issue:1

    The concentration of transforming growth factor beta (TGF-beta) in plasma has been correlated with the development of several diseases, including atherosclerosis and certain forms of cancer. However, the mechanisms that control the concentration of TGF-beta in plasma are poorly understood. In a study of 170 pairs of female twins (average age 57.7 years) we show that the concentration of active plus acid-activatable latent TGF-beta1 [(a+l) TGF-beta therefore is predominantly under genetic control (heritability estimate 0.54). Single strand conformation polymorphism (SSCP) mapping of the TGF-beta1 gene promoter has identified two single base substitution polymorphisms. The two polymorphisms (G-->A at position -800 bp and C-->T at position -509 bp) are in linkage disequilibrium (correlation coefficient Delta = 0.215, P < 0.01). The C-509T polymorphism is significantly associated with the plasma concentration of (a+l) TGF-beta1, explaining 8.2% of the additive genetic variance of (a+l) TGF-beta1 concentration. It is therefore possible that predisposition to atherosclerosis, bone diseases or various forms of cancer may be correlated with the presence of particular alleles at the TGFB1 locus.

    Topics: Adult; Aged; Alleles; Arteriosclerosis; Base Sequence; Bone Diseases; DNA; DNA Primers; Female; Genetic Variation; Humans; Linkage Disequilibrium; Middle Aged; Models, Genetic; Neoplasms; Polymorphism, Single-Stranded Conformational; Promoter Regions, Genetic; Transforming Growth Factor beta; Twins, Dizygotic; Twins, Monozygotic

1999
A transformed view of cyclosporine.
    Nature, 1999, Feb-11, Volume: 397, Issue:6719

    Topics: Animals; Carcinogens; Cyclosporine; Cytokines; Humans; Immunosuppressive Agents; Mice; Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured

1999
Transforming growth factor beta triggers two independent-senescence programs in cancer cells.
    Biochemical and biophysical research communications, 1999, Feb-05, Volume: 255, Issue:1

    Transforming growth factor-beta (TGF-beta)TG has been shown to play a multifunctional role in tumorigenesis. Here we demonstrate that TGF-beta induces a morphological change and expression of senescence-associated beta-galactosidase activity in the human lung adenocarcinoma cell line A549 cells within a week after the addition. These TGF-beta induced phenotypic changes are thought to characterize the rapid onset of senescence. When A549 cells were treated with TGF-beta, cell growth was not completely arrested, but the activity of telomerase was down regulated via transcriptional repression of telomerase reverse transcriptase, which led to a shortening of the telomere during long-term culture and finally resulted in replicative senescence. These results indicate that TGF-beta is able to induce a rapid senescence in A549 cells without significantly inhibiting cell growth and can further direct A549 cells to a replicative senescence state via the suppression of telomerase which culminates in telomere shortening. All these experimental results suggest that TGF-beta transmits several separate and independent signals to shift A549 cells back to a normal senescent cell.

    Topics: Cell Division; Cell Transformation, Neoplastic; Cellular Senescence; Enzyme Activation; Gene Expression Regulation, Neoplastic; Humans; Neoplasms; Telomerase; Transforming Growth Factor beta; Tumor Cells, Cultured

1999
Regulation of machinery for cancer cell growth, immortality, apoptosis and invasion--the Eighteenth International Symposium of Sapporo Cancer Seminar.
    Japanese journal of cancer research : Gann, 1999, Volume: 90, Issue:3

    Topics: Apoptosis; Cell Division; Cellular Senescence; Japan; Metalloendopeptidases; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Telomerase; Transforming Growth Factor beta

1999
Human cut-like repressor protein binds TGFbeta type II receptor gene promoter.
    Archives of biochemistry and biophysics, 1999, Nov-15, Volume: 371, Issue:2

    Resistance to the growth inhibitory effects of transforming growth factor beta (TGFbeta) has been associated with decreased levels of the TGFbeta type II receptor (TbetaR-II) and has been correlated with tumorigenicity. Previously, we reported an A --> G mutation at position -364 in the TbetaR-II promoter in A431 tumor cells which results in reduced TbetaR-II promoter activity. In this study, we show that the CDP/Cut (CCAAT displacement protein) transcription factor, a transcriptional repressor, binds both the wild type and the mutant TbetaR-II promoter. We also demonstrate that the A --> G mutation increases CDP/Cut binding affinity, and that overexpression of CDP/Cut reduces transcription from TbetaR-II promoter reporter constructs. Increased binding of the CDP/Cut repressor protein, as a result of a mutation at position -364, represents a novel mechanism of regulation in a neoplastic cell of the promoter of a tumor suppressor gene, TbetaR-II.

    Topics: Binding Sites; DNA Mutational Analysis; Female; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Neoplasms; Nuclear Proteins; Promoter Regions, Genetic; Protein Binding; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Repressor Proteins; Transcription Factors; Transforming Growth Factor beta; Tumor Cells, Cultured; Vulvar Neoplasms

1999
TbetaR-I(6A) is a candidate tumor susceptibility allele.
    Cancer research, 1999, Nov-15, Volume: 59, Issue:22

    We have previously described a type I transforming growth factor (TGF)-beta receptor (TbetaR-I) polymorphic allele, TbetaR-I(6A), that has a deletion of three alanines from a nine-alanine stretch. We observed a higher than expected number of TbetaR-I(6A) homozygotes among tumor and nontumor DNA from patients with a diagnosis of cancer. To test the hypothesis that TbetaR-I(6A) homozygosity is associated with cancer, we performed a case-control study in patients with a diagnosis of cancer and matched healthy individuals with no history of cancer and who were identical in their gender and their geographical and ethnic background to determine the relative germ-line frequencies of this allele. We found nine TbetaR-I(6A) homozygotes among 851 patients with cancer. In comparison, there were no TbetaR-I(6A) homozygotes among 735 healthy volunteers (P < 0.01). We also observed an excess of TbetaR-I(6A) heterozygotes in cancer cases compared to controls (14.6% versus 10.6%; P = 0.02, Fisher's exact test). A subset analysis revealed that 4 of 112 patients with colorectal cancer were TbetaR-I(6A) homozygotes (P < 0.01). Using mink lung epithelial cell lines devoid of TbetaR-I, we established stably transfected TbetaR-I and TbetaR-I(6A) cell lines. We found that, compared to TbetaR-I, TbetaR-I(6A) was impaired as a mediator of TGF-beta antiproliferative signals. We conclude that TbetaR-I(6A) acts as a tumor susceptibility allele that may contribute to the development of cancer, especially colon cancer, by means of reduced TGF-beta-mediated growth inhibition.

    Topics: Activin Receptors, Type I; Alleles; Analysis of Variance; Breast Neoplasms; Case-Control Studies; Colonic Neoplasms; Female; Genetic Predisposition to Disease; Germinoma; Heterozygote; Homozygote; Humans; Male; Neoplasms; Ovarian Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Transfection; Transforming Growth Factor beta

1999
Prolactin is an antagonist of TGF-beta activity and promotes proliferation of murine B cell hybridomas.
    Cellular immunology, 1998, Mar-15, Volume: 184, Issue:2

    Prolactin (PRL) is an immunomodulator that has been demonstrated to enhance immune responses both in vitro and in vivo. Prolactin enhances the proliferative response of lymphoid cells to both nonspecific mitogens and specific antigens and increases their production of IL-2 and interferon-gamma. Studies were performed to examine whether recombinant human prolactin (r-hPRL) also acts as a growth factor for B cell hybridomas. Prolactin was able to stimulate proliferation of murine B cell hybridomas in a dose-dependent manner and enhanced their proliferation in response to IL-4, IL-5, and IL-6. This increase in proliferation resulted in an overall increase in antibody production. Studies were also undertaken to examine the effect of PRL with transforming growth factor beta (TGF-beta), an immunosuppressive cytokine. Hybridoma cell lines incubated with TGF-beta demonstrated a dose-dependent decrease in proliferation. Variability in the degree of inhibition was observed among the various hybridomas in their responsiveness to TGF-beta. The addition of r-hPRL to the cultures reversed the antiproliferative effects of TGF-beta. The mechanism by which PRL can overcome the anti-proliferative effect of TGF-beta is under investigation. These findings provide an additional rationale for using r-hPRL clinically in immunosuppressed patients in certain disease settings such as AIDS and cancer, where overexpression of TGF-beta has been implicated in disease development and progression.

    Topics: Adjuvants, Immunologic; Animals; Antibody Formation; B-Lymphocytes; Cell Division; HIV Infections; Humans; Hybridomas; Interleukin-4; Interleukin-5; Interleukin-6; Mice; Neoplasms; Prolactin; Recombinant Proteins; Transforming Growth Factor beta

1998
Dpc4 transcriptional activation and dysfunction in cancer cells.
    Cancer research, 1998, Oct-15, Volume: 58, Issue:20

    Dpc4 (Smad4) is implicated in mediation of signals from transforming growth factor (TGF) beta and related ligands, and wild-type Dpc4 mediates TGF-beta-stimulated gene transcription at specific DNA sequences bound by Dpc4 [Smad binding element (SBE)]. We characterized panels of DPC4 tumor mutations and cancer cell lines. Amino acid substitutions within the NH2-terminal third of Dpc4 weakened or ablated SBE-mediated gene regulation by a disruption of DNA binding. An interaction of the COOH-terminal end with the DNA-binding domain of Dpc4 was evident but was not required to explain the functional impairment produced by NH2-terminal DPC4 mutations. Both substitution and truncation mutations of the COOH-terminal half of DPC4 lacked the ability to regulate transcription while retaining the sequence-specific DNA-binding function, but through differing mechanisms. A modular domain to redistribute Dpc4 to the nuclear compartment allowed SBE-mediated transcriptional activation in a cell line having a TGF-1 receptor defect and was sufficient to restore SBE-mediated transactivation ability to COOH-terminal DPC4 missense mutants. Cells harboring DPC4 alterations had a universal impairment of the TGF-beta-stimulated SBE transcriptional response. These studies identify the loss of SBE-mediated gene regulation as a uniform outcome of the selection for DPC4 alterations during tumorigenesis. They raise the possibility of restoration of some Dpc4-associated transcriptional events in cancer cells through the targeted redistribution of wild-type and some missense mutant forms of Dpc4 to the nucleus.

    Topics: DNA; DNA-Binding Proteins; Humans; Mutation; Neoplasms; Smad4 Protein; Structure-Activity Relationship; Trans-Activators; Transcriptional Activation; Transforming Growth Factor beta; Tumor Cells, Cultured

1998
[Increased possibility to affect cell signals following research break-through. Development of new therapeutic strategies].
    Lakartidningen, 1998, Oct-28, Volume: 95, Issue:44

    Topics: Animals; Cell Communication; Fibrosis; Humans; Neoplasms; Neoplasms, Experimental; Research; Signal Transduction; Transforming Growth Factor beta

1998
TEP1, encoded by a candidate tumor suppressor locus, is a novel protein tyrosine phosphatase regulated by transforming growth factor beta.
    Cancer research, 1997, Jun-01, Volume: 57, Issue:11

    It has long been postulated that protein tyrosine phosphatases may act as tumor suppressors because of their ability to counteract the oncogenic actions of protein tyrosine kinases. Here we report the cloning and characterization of a novel human protein tyrosine phosphatase, TEP1. TEP1 contains the protein tyrosine phosphatase signature motif, and we show that it possesses an intrinsic protein tyrosine phosphatase activity. TEP1 also shares extensive homology with tensin, a cytoskeletal protein localized to focal adhesions, and with auxilin, a protein involved in synaptic vesicle transport. Immunofluorescence studies show that TEP1 is a cytoplasmic protein. The abundance of TEP1 transcription is altered in many transformed cells. In the transforming growth factor beta-sensitive cells, TEP1 expression is rapidly down-regulated by transforming growth factor beta, a cytokine shown to be involved in regulating cell adhesion and cell motility. We have also mapped the gene encoding TEP1 to chromosome 10q23, a locus that is frequently deleted in a variety of human cancers. TEP1 protein is identical to the protein encoded by the candidate tumor suppressor gene PTEN/MMAC1. Our functional studies of the TEP1 protein suggest that its tumor suppressor function may associate with its intrinsic protein tyrosine phosphatase activity and its cytoplasmic localization.

    Topics: 3T3 Cells; Adaptor Proteins, Vesicular Transport; Amino Acid Sequence; Animals; Base Sequence; Cells, Cultured; Chromosome Mapping; Chromosomes, Human, Pair 10; Cloning, Molecular; Fibroblasts; Fluorescent Antibody Technique, Indirect; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; HeLa Cells; Humans; Mice; Microfilament Proteins; Molecular Sequence Data; Neoplasms; Nerve Tissue Proteins; Phosphoproteins; Phosphoric Monoester Hydrolases; Protein Tyrosine Phosphatases; PTEN Phosphohydrolase; Sequence Deletion; Sequence Homology, Amino Acid; Tensins; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Proteins

1997
Mutations increasing autoinhibition inactivate tumour suppressors Smad2 and Smad4.
    Nature, 1997, Jul-03, Volume: 388, Issue:6637

    Smad2 and Smad4 are related tumour-suppressor proteins, which, when stimulated by the growth factor TGF-beta, form a complex to inhibit growth. The effector function of Smad2 and Smad4 is located in the conserved carboxy-terminal domain (C domain) of these proteins and is inhibited by the presence of their amino-terminal domains (N domain). This inhibitory function of the N domain is shown here to involve an interaction with the C domain that prevents the association of Smad2 with Smad4. This inhibitory function is increased in tumour-derived forms of Smad2 and 4 that carry a missense mutation in a conserved N domain arginine residue. The mutant N domains have an increased affinity for their respective C domains, inhibit the Smad2-Smad4 interaction, and prevent TGF beta-induced Smad2-Smad4 association and signalling. Whereas mutations in the C domain disrupt the effector function of the Smad proteins, N-domain arginine mutations inhibit SMAD signalling through a gain of autoinhibitory function. Gain of autoinhibitory function is a new mechanism for inactivating tumour suppressors.

    Topics: Animals; Binding Sites; Cloning, Molecular; COS Cells; DNA-Binding Proteins; Escherichia coli; Gene Expression Regulation; Genes, Tumor Suppressor; Humans; Mutation; Neoplasms; Nerve Growth Factors; Saccharomyces cerevisiae; Signal Transduction; Smad Proteins; Smad2 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta; Xenopus; Xenopus Proteins

1997
Distinct tumor specific expression of TGFB4 (ebaf)*, a novel human gene of the TGF-beta superfamily.
    Frontiers in bioscience : a journal and virtual library, 1997, Jul-15, Volume: 2

    We recently identified a novel gene of the TGF-beta superfamily, endometrial bleeding associated factor, TGFB4 (ebaf), that, throughout the menstrual cycle, exhibited a defined expression in human endometrium. Here, we report on the expression of TGFB4 (ebaf) in normal and neoplastic human tissues. The expression of this gene was absent in a host of normal tissues including lung, stomach, small bowel, liver, kidney, breast, lymph node, spleen, ovary and fallopian tube. However, a weak expression of the 2.1 kb variant of the TGFB4 (ebaf) mRNA was observed in rectal, ovarian, and testicular tissues and the 2.1 and 2.5 kb TGFB4 (ebaf) mRNAs were observed in the pancreatic tissue. The expression of the mRNA of this gene was absent in sarcomas, Hodgkin's and non-Hodgkin's lymphomas, melanomas, squamous cell carcinomas, hepatocellular carcinomas, renal cell carcinomas, and adenocarcinomas of the breast, endometrium and lung. The expression of the TGFB4 (ebaf) mRNA was observed primarily in adenocarcinomas that exhibited a mucinous differentiation. This included colonic, duodenal, and ovarian adenocarcinomas. The expression of TGFB4 (ebaf) mRNA was absent in non- mucinous colonic, gastric and ovarian adenocarcinomas and adenocarcinomas of colon metastatic to the liver. However, some serous adenocarcinomas of the ovary also exhibited TGFB4 (ebaf) mRNA. The testicular tumors, seminomas and embryonal carcinomas, also expressed TGFB4 (ebaf) mRNA. These findings show that the TGFB4 (ebaf) mRNA has distinct tumor specific expression.

    Topics: Adenocarcinoma; Blotting, Northern; Female; Gene Expression Profiling; Humans; In Situ Hybridization; Left-Right Determination Factors; Male; Neoplasms; RNA, Messenger; Tissue Distribution; Transforming Growth Factor beta

1997
Renal pathophysiology.
    Current opinion in nephrology and hypertension, 1997, Volume: 6, Issue:4

    Topics: Aminoglycosides; Animals; Anti-Bacterial Agents; Endothelins; Humans; Hypertension; Ion Channels; Kidney; Neoplasms; Polycystic Kidney Diseases; Sodium Channels; Transforming Growth Factor beta; Water-Electrolyte Imbalance

1997
UICC Study Group on Basic and Clinical Cancer Research: mechanisms of growth factor and hormone insensitivity.
    International journal of cancer, 1997, Nov-14, Volume: 73, Issue:4

    Topics: Animals; Apoptosis; Cell Cycle; Cytoskeleton; Drug Resistance, Neoplasm; Growth Substances; Hormones; Humans; Neoplasms; Signal Transduction; Telomerase; Telomere; Transforming Growth Factor beta

1997
Tumour-derived, endocrine, exogenous and therapeutic factors differentially modulate cytokine secretion in whole blood cell culture.
    European journal of cancer (Oxford, England : 1990), 1997, Volume: 33, Issue:10

    Following our previous results which showed that TGF-beta 1 suppressed the secretion of certain cytokines, we investigated the effects of different endogenous and exogenous factors on cytokine secretion in whole blood cell culture by using an enzyme-linked immunosorbent assay (ELISA) for measurement of cytokine concentrations. Several molecules including dexamethasone, noradrenaline (NA) and ethanol differentially inhibited mitogen-induced cytokine secretion. Dexamethasone and noradrenaline suppressed secretion of IL-2, IFN alpha, IFN gamma, TNF alpha, IL-1 alpha and IL-1 beta. beta-Endorphin and Leu-Enkephalin had no significant influence on cytokine secretion. Suppression of cytokine secretion by TGF-beta 1 was further intensified significantly and dose dependently by addition of noradrenaline. GM-CSF stimulated the secretion of IL-1 alpha, IL-1 beta and TNF gamma, but had no influence on the secretion of IL-2, IFN alpha and IFN gamma. G-CSF, IL-3 and SCF did not significantly influence secretion of all cytokines tested. Thus, endogenous and exogenous factors differentially influence cytokine secretion by immunocompetent cells.

    Topics: Adult; Cell Culture Techniques; Colony-Stimulating Factors; Cytokines; Dexamethasone; Ethanol; Humans; Immune Tolerance; Immunosuppressive Agents; Neoplasms; Norepinephrine; Transforming Growth Factor beta

1997
Atherosclerosis, just another cancer?
    The Journal of clinical investigation, 1997, Nov-01, Volume: 100, Issue:9

    Topics: Arteriosclerosis; Humans; Mutation; Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Repetitive Sequences, Nucleic Acid; Transforming Growth Factor beta

1997
Transforming growth factor-beta 1 delays formation of granulocyte-macrophage colony-forming cells, but spares more primitive progenitors during ex vivo expansion of CD34+ haemopoietic progenitor cells.
    British journal of haematology, 1997, Volume: 99, Issue:4

    Ex vivo culture and expansion of autologous haemopoietic transplants has been developed to improve tumour cell purging and accelerate haemopoietic reconstitution by transplantation of increased progenitor cell numbers. We studied the effect of the negative haemopoietic regulator, transforming growth factor beta-1 (TGF-beta 1) on primitive precursors during ex vivo expansion of CD34+ cells. When added directly to methylcellulose colony-forming assays, TGF-beta 1 potently suppressed the development of granulocyte-macrophage colonies from CD34+ enriched peripheral blood progenitor cells (80-90% inhibition). In contrast, expansion of total nucleated cells and granulocyte-macrophage colony-forming cells (GM-CFC) from CD34+ progenitors in liquid culture in the presence of stem cell factor (SCF), interleukin (IL)-1 beta, IL-3, IL-6 and erythropoietin (EPO) was inhibited to 32-65% of control culture levels within 14 d when TGF-beta 1 was added, and still produced an average 3.3-fold absolute amplification of GM-CFC. The inhibitory effect of TGF-beta 1 on GM-CFC generation was reversed when it was washed out on day 6 of ex vivo expansion cultures, and total numbers of GM-CFC generated from expansion cultures then reached levels of untreated controls by day 16. Long-term bone marrow culture-initiating cell (LTCIC) numbers were preserved, at least at input levels, over a culture period of 14 d both in control and TGF-beta-1-treated expansion cultures. These findings suggest that TGF-beta 1, a cytokine which induces apoptosis or terminal differentiation in a number of malignant cell types, may be added to ex vivo expansion cultures without loss of primitive cells from autologous haemopoietic transplants.

    Topics: Apoptosis; Bone Marrow Cells; Cell Differentiation; Cell Division; Cells, Cultured; Granulocytes; Hematopoietic Stem Cells; Humans; Interleukins; Lymphatic Diseases; Macrophages; Neoplasms; Stem Cell Factor; Transforming Growth Factor beta

1997
Microsatellite instability: the mutator that mutates the other mutator.
    Nature medicine, 1996, Volume: 2, Issue:6

    Topics: Adenoma; Adenomatous Polyposis Coli; Adenomatous Polyposis Coli Protein; Animals; Biological Evolution; Colonic Neoplasms; Cytoskeletal Proteins; DNA Repair; DNA-Binding Proteins; DNA, Satellite; Fungal Proteins; Humans; Mice; Microsatellite Repeats; Models, Genetic; Mutation; MutS Homolog 2 Protein; Neoplasms; Transforming Growth Factor beta

1996
Polarity, proliferation and the hedgehog pathway.
    Nature genetics, 1996, Volume: 14, Issue:3

    Topics: Animals; Cell Differentiation; Cell Division; Cleft Palate; Drosophila; Embryonic Induction; Genes, Tumor Suppressor; Hedgehog Proteins; Holoprosencephaly; Humans; Mice; Mutation; Neoplasms; Proteins; Signal Transduction; Spinal Dysraphism; Trans-Activators; Transforming Growth Factor beta

1996
p53/TGF-beta/cancer: an intriguing connection.
    Cytokine & growth factor reviews, 1996, Volume: 7, Issue:4

    Topics: Animals; Humans; Neoplasms; Transforming Growth Factor beta; Tumor Suppressor Protein p53

1996
MAD-related proteins in TGF-beta signalling.
    Trends in genetics : TIG, 1996, Volume: 12, Issue:12

    Topics: Animals; Carrier Proteins; DNA-Binding Proteins; Humans; Neoplasms; Signal Transduction; Smad1 Protein; Transforming Growth Factor beta; Xenopus Proteins

1996
Short communication: normal tissue injury after cancer therapy is a local response exacerbated by an endocrine effect of TGF beta.
    The British journal of radiology, 1995, Volume: 68, Issue:807

    The sensitivity of normal tissues rather than of the tumour usually limits the effectiveness of cancer treatment. The normal tissue side effects from chemotherapy and/or radiotherapy result from both direct cellular loss and the extensive fibrosis that develops at the site of injury. Recent evidence suggests that the cytokine, transforming growth factor beta (TGF beta), mediates this fibrogenic process. Herein, we provide evidence in support of the hypothesis that the fibrosis formation following therapy results not only from TGF beta produced locally in the injured normal tissue, but also from circulating TGF beta released by the tumour. Thus, therapy-induced normal tissue damage appears in part to be a local manifestation of a systemic condition.

    Topics: Antineoplastic Agents; Fibrosis; Humans; Lung; Neoplasms; Radiation Injuries; Radiotherapy; Transforming Growth Factor beta

1995
Stromal expression of c-Ets1 transcription factor correlates with tumor invasion.
    Cancer research, 1994, Nov-01, Volume: 54, Issue:21

    The stroma reaction has an important role in tumor growth, invasion, and metastasis. In various invasive human carcinomas, as well as in a mouse model for tumor invasion, transcripts encoding the transcription factor c-Ets1 were detected within stromal fibroblasts, whereas they were absent in epithelial tumor cells. This expression of c-Ets1 was often increased in fibroblasts directly adjacent to neoplastic cells. Endothelial cells of stromal capillaries were also positive for c-Ets1 expression. In contrast, fibroblasts of corresponding noninvasive lesions and of normal tissues were consistently negative. In cultured human fibroblasts stimulated by basic fibroblast growth factor and tumor necrosis factor alpha, the expression of c-Ets1 correlated with the accumulation of transcripts for potential target genes, collagenase-1 and stromelysin-1. The same correlation was observed in some of the invasive carcinomas investigated. These results suggest that c-Ets1 participates in the regulation of tumor invasion in vivo.

    Topics: Adult; Aged; Animals; Collagenases; Female; Fibroblast Growth Factor 2; Fibroblasts; Humans; In Situ Hybridization; Male; Matrix Metalloproteinase 3; Metalloendopeptidases; Mice; Mice, SCID; Middle Aged; Neoplasm Invasiveness; Neoplasms; Oncogene Proteins; Platelet-Derived Growth Factor; Proto-Oncogene Proteins c-ets; RNA, Messenger; Transcription Factors; Transcription, Genetic; Transforming Growth Factor alpha; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator

1994
[Expression of transforming growth factor beta-3 (TGF-beta-3) on reactive and malignant cells in ascites and pleural effusion].
    Medizinische Klinik (Munich, Germany : 1983), 1994, May-15, Volume: 89, Issue:5

    The expression of TGF-beta-3 was examined in 64 patients with reactive and malignant effusion by immunocytochemistry.. In about half of the patients with malignant effusions (especially breast cancer, gastric cancer, and carcinomas of unknown primary) TGF-beta positive tumor cells could be detected. We could show here for the first time that reactive mesothelial cells could also express TGF-beta. Lymphatic cells were negative in all cases. TGF-beta-3 bioactivity could also be detected in the effusions studied. In our group of patients with far advanced cancer, the expression of TGF-beta had no clear-cut clinical or prognostic correlate. However, the expression of TGF-beta on tumor cells should be interpreted as a marker of tumor progression, taking into account the fibrogenic, angiogenic and immunosuppressive properties of TGF-beta.. Further research is necessary to answer the question if the 3 isoforms of TGF-beta are coordinately expressed and to elucidate the involvement of this cytokine in tumor progression and metastasis.

    Topics: Adult; Aged; Ascitic Fluid; Cell Division; Female; Humans; Male; Middle Aged; Neoplasms; Pleural Effusion, Malignant; Transforming Growth Factor beta; Tumor Cells, Cultured

1994
Growth modulation of human tumor cells by a growth-inhibiting activity derived from tumorigenic V79 Chinese hamster cells.
    In vitro cellular & developmental biology. Animal, 1993, Volume: 29A, Issue:4

    A growth-inhibiting activity was identified in supernatants of the neoplastic V79 Chinese hamster cell line based on its ability to inhibit the proliferation of the same cell line. The partially purified activity, provisionally termed "growth inhibiting factor" (GIF) activity, inhibited the growth of a wide variety of human tumor cells, but not various normal human fibroblasts. This species-nonspecific activity was reversible, saturable, and highly potent in tumorigenic cell lines, and was noted in both monolayer culture and in soft agar. The inhibitory activity of GIF was also exhibited in a chemically defined serum-free medium supplemented with insulin and transferrin. GIF activity was stable to acid, heat, trypsin, and dithiothreitol but sensitive to alpha-chymotrypsin. The pattern of growth modulation by GIF on V79 cells was apparently different from those exhibited by bifunctional peptides such as transforming growth factor-beta, tumor necrosis factor-alpha, and interleukin-1-alpha. In addition, GIF activity cannot be ascribed to these cytokines based on the physicochemical and immunologic properties. Although GIF has yet to be purified to homogeneity, these data suggest that GIF might be a novel growth regulator which has a critical role in regulating growth of V79 cells. The growth modulation of tumor cells by this tumor-derived growth inhibiting activity suggested the presence of an autocrine growth regulatory mechanism even in tumor cells.

    Topics: Animals; Cell Division; Cell Line; Chymotrypsin; Cricetinae; Cricetulus; Culture Media; Dose-Response Relationship, Drug; Fibroblasts; Growth Inhibitors; Hot Temperature; Humans; Interleukin-1; Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

1993
Structural and functional aspects of transforming growth factor-beta in prostate cancer and other human malignancies.
    Human pathology, 1993, Volume: 24, Issue:1

    Topics: Female; Humans; Male; Neoplasms; Prostatic Neoplasms; Transforming Growth Factor beta

1993
Phenotypic and functional analysis of lymphocytes infiltrating paediatric tumours, with a characterization of the tumour phenotype.
    Cancer immunology, immunotherapy : CII, 1992, Volume: 34, Issue:4

    Tumour-infiltrating lymphocytes (TIL) of paediatric tumours obtained from 37 lesions of different histotype (12 osteosarcomas, 5 Wilms' tumours, 7 soft-tissue sarcomas, 5 neuroblastomas and 8 miscellaneous) were studied to establish their potential for therapy. Fresh isolated TIL were cultured for the first 2 weeks with low doses of interleukin-2 (IL-2) (20 Cetus U/ml) to select for "tumour-specific" lymphocytes potentially present in the neoplastic lesion, followed by culture with high doses of IL-2 (1000 Cetus U/ml) to achieve TIL expansion. TIL were grown with more than 10-fold expansion in only 9 cases (mean expansion: 58-fold, range 13.5-346). In 17 cases no viable cells were obtained. After 30 days of culture with IL-2 the proliferative ability of TIL declined sharply in the majority of cases and TIL became refractory to any further stimulus, including addition of IL-4, tumour necrosis factor alpha (TNF alpha) or interferon gamma, and activation with OKT3 in solid phase. In 20 out of 37 cases TIL were available for phenotypic and functional analysis. TIL after long-term culture were predominantly CD3+ but 2 cases of osteosarcoma showed a predominance of CD3+TcR gamma/delta cells. The CD4/CD8 ratio was more than 1 in 10 cases, without correlation with tumour histology, site of lesion or TIL growth. The number of CD16+ and CD25+ lymphocytes decreased progressively during culture, the latter concomitantly with a reduction of TIL growth rate. The lytic pattern of TIL against allogenic and autologous tumour (Auto-Tu) cells was variable, but specific lysis of Auto-Tu was seen in only one case (Wilms' tumour) after culture with TNF alpha and irradiated Auto-Tu cells. The immunohistochemical analysis of tumour lesions revealed a limited lymphocyte infiltrate, a low expression of histocompatibility leukocyte antigens (HLA) class I and of the adhesion molecules ICAM1, LFA3, and a significant production of transforming growth factor beta (TGF beta). These data indicate that TIL obtained from paediatric patients are difficult to expand at levels required for immunotherapy and lack a significant number of tumour-specific T lymphocytes. A low expression of immunomodulatory molecules on tumour cells or the production of suppressive factors may prevent activation and expansion of TIL in paediatric tumours.

    Topics: Antigens, CD; Antigens, Differentiation, T-Lymphocyte; CD3 Complex; CD4-CD8 Ratio; Cell Adhesion Molecules; Cell Division; Child; Cytotoxicity, Immunologic; HLA Antigens; Humans; Interferon-gamma; Interleukin-2; Interleukin-4; Lymphocyte Subsets; Lymphocytes, Tumor-Infiltrating; Neoplasms; Receptors, Antigen, T-Cell; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha; Wilms Tumor

1992
Simultaneous assessment of TGFB and cell cycle kinetics using IUdR/BrdU infusions in human neoplasms from plastic-embedded tissue.
    The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society, 1992, Volume: 40, Issue:3

    We describe an immunohistochemical technique that makes use of two monoclonal antibodies (MAb), one to detect the transforming growth factor B (TGFB) and another that reacts with iodo- and bromodeoxyuridine. The purpose of this technique is to determine the relationship between TGFB expression and the S-phase cells in human tumors. Since both can be distinctly identified in situ from tissue embedded in plastic, in assessment of the geographic orientation of S-phase cells in relation to such factors as TGFB, contiguity to blood vessels, nerve fibers, and macrophages can also be achieved.

    Topics: Antibodies, Monoclonal; Biopsy; Bone Marrow; Breast Neoplasms; Bromodeoxyuridine; Cell Cycle; Colonic Neoplasms; Female; Glioblastoma; Histological Techniques; Humans; Idoxuridine; Indicators and Reagents; Kinetics; Neoplasms; S Phase; Transforming Growth Factor beta

1992
Transforming growth factor-beta 1 down-regulates expression of membrane-associated lymphotoxin and secretion of soluble lymphotoxin by human lymphokine-activated killer T cells in vitro.
    Lymphokine and cytokine research, 1992, Volume: 11, Issue:5

    Lymphokine-activated T killer lymphocytes (T-LAK) are important effector cells in various diseases of tissue destructive reactions. They require stimulation with various cytokines to proliferate and mature into function effector cells. We have examined the role of various endogenously and exogenously added cytokines, and tumor necrosis factor (TNF)/lymphotoxin (LT) receptors in this process in vitro. The present report is a continuation of these studies. We found that human T-LAK cells express membrane-associated LT (mLT) but not TNF, and secrete high amount of soluble LT (sLT) but low levels of TNF. When added to the initial cultures or immature T-LAK cells, transforming growth factor-beta 1 (TGF-beta 1) suppressed both mLT expression and sLT secretion by 30-40%. Coculture of mature T-LAK cells with TGF-beta 1 caused 35% down-regulation of both mLT expression and sLT secretion after 18 h of incubation. Kinetic experiments indicated reduction of LT mRNA synthesis could occur in as little as 1 h when cocultured with 5 ng/ml of TGF-beta 1. TGF-beta 1 also reduced mLT induced T-LAK cell cytolytic activity on L929 cells in vitro. It appears TGF-beta 1 can down-regulate LT mRNA syntheses, mLT expression, and sLT secretion of human T-LAK cells in vitro.

    Topics: Cells, Cultured; Dose-Response Relationship, Drug; Down-Regulation; Humans; Killer Cells, Lymphokine-Activated; Lymphotoxin-alpha; Neoplasms; RNA, Messenger; Transcription, Genetic; Transforming Growth Factor beta

1992
Serum growth factors and oncoproteins in firefighters.
    Occupational medicine (Oxford, England), 1992, Volume: 42, Issue:1

    Firefighters are potentially at increased risk for cancer and non-malignant respiratory disease due to their toxic exposures on the job. Growth factors and oncogene proteins are thought to play a role in the development of various malignancies and pulmonary fibrotic diseases. Therefore, a cohort of firefighters and matched controls have been screened for the presence of nine different growth factors and oncoproteins using an immunoblotting assay. Fourteen of the firefighters were found to be positive for beta-transforming growth factor (beta-TGF) related proteins compared to no positives in the controls (P = 0.0017). These results suggest that beta-TGF may be a possible biomarker for monitoring firefighters and other exposed workers for the potential development of cancer or non-malignant respiratory disease.

    Topics: Amino Acid Sequence; Fires; Humans; Mass Screening; Middle Aged; Molecular Sequence Data; Neoplasms; New York City; Occupational Exposure; Oncogene Proteins; Platelet-Derived Growth Factor; Population Surveillance; Pulmonary Fibrosis; Risk Factors; Transforming Growth Factor beta

1992
[The role of transforming growth factors in the growth control of cancer cells].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 1991, Volume: 36, Issue:7

    Topics: Animals; Cell Division; Neoplasms; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor alpha; Transforming Growth Factor beta

1991
[Involvement of cellular responses to growth factors in angiogenesis and transformation malignant].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 1991, Volume: 36, Issue:7

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Endothelium, Vascular; Epidermal Growth Factor; Fibroblast Growth Factors; Humans; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta

1991
Understanding autocrine growth regulation.
    The Western journal of medicine, 1991, Volume: 155, Issue:2

    Topics: Growth Substances; Humans; Neoplasms; Transforming Growth Factor alpha; Transforming Growth Factor beta; Transforming Growth Factors

1991
Modulation of tumoricidal activity, induced in bone-marrow-derived mononuclear phagocytes by interferon gamma or Corynebacterium parvum, by interferon beta, tumor necrosis factor, prostaglandin E2, and transforming growth factor beta.
    International journal of cancer, 1991, Nov-11, Volume: 49, Issue:5

    Among a series of agents, including various interleukins and growth factors, only interferon gamma (IFN gamma) and heat-killed Corynebacterium parvum (CP) organisms were able to elicit, within 24 hr, tumoricidal activity in bone-marrow-derived mononuclear (BMM) phagocytes. In subsequent experiments, the abilities of interferon beta (IFN beta), tumor necrosis factor alpha (TNF alpha), prostaglandin E2 (PGE2), and transforming growth factor beta (TFG beta), alone or in combinations of 2, to modulate tumoricidal activity triggered in BMM phagocytes by IFN gamma or CP, were compared. In concentrations secreted by macrophages under physiological conditions, these agents proved potent in modulating induction and/or expression of tumoricidal activity. However, their ability to interfere with tumoricidal activity varied considerably, depending on the extent of macrophage differentiation and/or functional responsiveness, the pathway of macrophage activation, the type, concentration and combination of the macrophage secretory molecules, and on whether the agents were present during induction and expression or only during expression of tumoricidal activity. In showing that IFN beta and TNF alpha were mostly enhancing and TGF beta mostly suppressive, whereas PGE2 suppressed induction but enhanced expression of tumoricidal activity, our findings provide further support for the concept that these macrophage-derived molecules have a key role in autocrine regulation of macrophage functional activities.

    Topics: Animals; Bone Marrow Cells; Cell Differentiation; Dinoprostone; Interferon-beta; Interferon-gamma; Macrophage Activation; Neoplasms; Phagocytes; Propionibacterium acnes; Rats; Recombinant Proteins; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

1991
[Growth factors and cell transformation].
    Nihon rinsho. Japanese journal of clinical medicine, 1990, Volume: 48, Issue:8

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Fibroblast Growth Factors; Humans; Molecular Sequence Data; Neoplasms; Platelet-Derived Growth Factor; Transforming Growth Factor alpha; Transforming Growth Factor beta

1990