transforming-growth-factor-beta and Cell-Transformation--Neoplastic

transforming-growth-factor-beta has been researched along with Cell-Transformation--Neoplastic* in 609 studies

Reviews

143 review(s) available for transforming-growth-factor-beta and Cell-Transformation--Neoplastic

ArticleYear
The function of LncRNA-ATB in cancer.
    Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 2023, Volume: 25, Issue:1

    Cancer as a progressive and complex disease is caused by early chromosomal changes and stimulated cellular transformation. Previous studies reported that long non-coding RNAs (lncRNAs) play pivotal roles in the initiation, maintenance, and progression of cancer cells. LncRNA activated by TGF-β (ATB) has been shown to be dysregulated in different types of cancer. Aberrant expression of lncRNA-ATB plays an important role in the progression of diverse malignancies. High expression of LncRNA-ATB is associated with cancer cell growth, proliferation, metastasis, and EMT. LncRNA-ATB by targeting various signaling pathways and microRNAs (miRNAs) can trigger cancer pathogenesis. Therefore, lncRNA-ATB can be a novel target for cancer prediction and diagnosis. In this review, we will focus on the function of lncRNA-ATB in various types of human cancers.

    Topics: Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; RNA, Long Noncoding; Signal Transduction; Transforming Growth Factor beta

2023
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 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
Molecular mechanisms underlying the action of carcinogens in gastric cancer with a glimpse into targeted therapy.
    Cellular oncology (Dordrecht), 2022, Volume: 45, Issue:6

    Gastric cancer imposes a substantial global health burden despite its overall incidence decrease. A broad spectrum of inherited, environmental and infectious factors contributes to the development of gastric cancer. A profound understanding of the molecular underpinnings of gastric cancer has lagged compared to several other tumors with similar incidence and morbidity rates, owing to our limited knowledge of the role of carcinogens in this malignancy. The International Agency for Research on Cancer (IARC) has classified gastric carcinogenic agents into four groups based on scientific evidence from human and experimental animal studies. This review aims to explore the potential comprehensive molecular and biological impacts of carcinogens on gastric cancer development and their interactions and interferences with various cellular signaling pathways.. In this review, we highlight recent clinical trial data reported in the literature dealing with different ways to target various carcinogens in gastric cancer. Moreover, we touch upon other multidisciplinary therapeutic approaches such as surgery, adjuvant and neoadjuvant chemotherapy. Rational clinical trials focusing on identifying suitable patient populations are imperative to the success of single-agent therapeutics. Novel insights regarding signaling pathways that regulate gastric cancer can potentially improve treatment responses to targeted therapy alone or in combination with other/conventional treatments. Preventive strategies such as control of H. pylori infection through eradication or immunization as well as dietary habit and lifestyle changes may reduce the incidence of this multifactorial disease, especially in high prevalence areas. Further in-depth understanding of the molecular mechanisms involved in the role of carcinogenic agents in gastric cancer development may offer valuable information and update state-of-the-art resources for physicians and researchers to explore novel ways to combat this disease, from bench to bedside. A schematic outlining of the interaction between gastric carcinogenic agents and intracellular pathways in gastric cancer H. pylori stimulates multiple intracellular pathways, including PI3K/AKT, NF-κB, Wnt, Shh, Ras/Raf, c-MET, and JAK/STAT, leading to epithelial cell proliferation and differentiation, apoptosis, survival, motility, and inflammatory cytokine release. EBV can stimulate intracellular pathways such as the PI3K/Akt, RAS/RAF, JAK/STAT, Notch, TGF-β, and NF-κB, leading to cell survival and motility, proliferation, invasion, metastasis, and the transcription of anti-apoptotic genes and pro-inflammatory cytokines. Nicotine and alcohol can lead to angiogenesis, metastasis, survival, proliferation, pro-inflammatory, migration, and chemotactic by stimulating various intracellular signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, ROS, and JAK/STAT. Processed meat contains numerous carcinogenic compounds that affect multiple intracellular pathways such as sGC/cGMP, p38 MAPK, ERK, and PI3K/AKT, leading to anti-apoptosis, angiogenesis, metastasis, inflammatory responses, proliferation, and invasion. Lead compounds may interact with multiple signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, DNA methylation-dependent, and epigenetic-dependent, leading to tumorigenesis, carcinogenesis, malignancy, angiogenesis

    Topics: Animals; Carcinogens; Cell Transformation, Neoplastic; Humans; NF-kappa B; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Reactive Oxygen Species; Stomach Neoplasms; Transforming Growth Factor beta

2022
TGF-β at the crossroads of multiple prognosis in breast cancer, and beyond.
    Life sciences, 2022, Dec-01, Volume: 310

    Transforming growth factor β (TGF-β), a pluripotent cytokine and a multifunctional growth factor has a crucial role in varied biological mechanisms like invasion, migration, epithelial-mesenchymal transition, apoptosis, wound healing, and immunosuppression. Moreover, it also has an imperative role both in normal mammary gland development as well as breast carcinogenesis. TGF-β has shown to have a paradoxical role in breast carcinogenesis, by transitioning from a growth inhibitor to a growth promoter with the disease advancement. The inter-communication and crosstalk of TGF-β with different signaling pathways has strengthened the likelihood to explore it as a comprehensive biomarker. In the last two decades, TGF-β has been studied extensively and has been found to be a promising biomarker for early detection, disease monitoring, treatment selection, and tumor progression making it beneficial for disease management. In this review, we focus on the signaling pathways and biological activities of the TGF-β family in breast cancer pathogenesis and its role as a circulatory and independent biomarker for breast cancer progression and metastasis. Moreover, this review highlights TGF-β as a drug target, and the underlying mechanisms through which it is involved in tumorigenesis that will aid in the development of varied therapies targeting the different stages of breast cancer.

    Topics: Breast; Breast Neoplasms; Carcinogenesis; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Female; Humans; Transforming Growth Factor beta

2022
TGF-β signaling: A recap of SMAD-independent and SMAD-dependent pathways.
    Journal of cellular physiology, 2022, Volume: 237, Issue:1

    Transforming growth factor-β (TGF-β) is a proinflammatory cytokine known to control a diverse array of pathological and physiological conditions during normal development and tumorigenesis. TGF-β-mediated physiological effects are heterogeneous and vary among different types of cells and environmental conditions. TGF-β serves as an antiproliferative agent and inhibits tumor development during primary stages of tumor progression; however, during the later stages, it encourages tumor development and mediates metastatic progression and chemoresistance. The fundamental elements of TGF-β signaling have been divulged more than a decade ago; however, the process by which the signals are relayed from cell surface to nucleus is very complex with additional layers added in tumor cell niches. Although the intricate understanding of TGF-β-mediated signaling pathways and their regulation are still evolving, we tried to make an attempt to summarize the TGF-β-mediated SMAD-dependent andSMAD-independent pathways. This manuscript emphasizes the functions of TGF-β as a metastatic promoter and tumor suppressor during the later and initial phases of tumor progression respectively.

    Topics: Cell Transformation, Neoplastic; Humans; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2022
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
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
Metabolic rewiring and redox alterations in malignant pleural mesothelioma.
    British journal of cancer, 2020, Volume: 122, Issue:1

    Malignant pleural mesothelioma (MPM) is a rare malignancy of mesothelial cells with increasing incidence, and in many cases, dismal prognosis due to its aggressiveness and lack of effective therapies. Environmental and occupational exposure to asbestos is considered the main aetiological factor for MPM. Inhaled asbestos fibres accumulate in the lungs and induce the generation of reactive oxygen species (ROS) due to the presence of iron associated with the fibrous silicates and to the activation of macrophages and inflammation. Chronic inflammation and a ROS-enriched microenvironment can foster the malignant transformation of mesothelial cells. In addition, MPM cells have a highly glycolytic metabolic profile and are positive in

    Topics: Animals; Antineoplastic Agents; Asbestos; Cell Transformation, Neoplastic; Cisplatin; Humans; Loss of Function Mutation; Lung Neoplasms; Mesothelioma; Mesothelioma, Malignant; Oxidation-Reduction; Pleural Neoplasms; Reactive Oxygen Species; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Suppressor Proteins; Ubiquitin Thiolesterase

2020
Cancer stem cells and their niche in the progression of squamous cell carcinoma.
    Cancer science, 2020, Volume: 111, Issue:11

    Most cancers harbor a small population of highly tumorigenic cells known as cancer stem cells (CSCs). Because of their stem cell-like properties and resistance to conventional therapies, CSCs are considered to be a rational target for curable cancer treatment. However, despite recent advances in CSC research, CSC-targeted therapies are not as successful as was initially hoped. The proliferative, invasive, and drug-resistant properties of CSCs are regulated by the tumor microenvironment associated with them, the so-called CSC niche. Thus, targeting tumor-promoting cellular crosstalk between CSCs and their niches is an attractive avenue for developing durable therapies. Using mouse models of squamous cell carcinoma (SCC), we have demonstrated that tumor cells responding to transforming growth factor β (TGF-β) function as drug-resistant CSCs. The gene expression signature of TGF-β-responding tumor cells has accelerated the identification of novel pathways that drive invasive tumor progression. Moreover, by focusing on the cytokine milieu and macrophages in the proximity of TGF-β-responding tumor cells, we recently uncovered the molecular basis of a CSC-niche interaction that emerges during early tumor development. This review article summarizes the specialized tumor microenvironment associated with CSCs and discusses mechanisms by which malignant properties of CSCs are maintained and promoted.

    Topics: Animals; Biomarkers, Tumor; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Disease Progression; Disease Susceptibility; Drug Resistance, Neoplasm; Humans; Neoplastic Stem Cells; Signal Transduction; Stem Cell Niche; Transforming Growth Factor beta; Tumor Microenvironment

2020
Interplay of TGFβ signaling and microRNA in thyroid cell loss of differentiation and cancer progression.
    Archives of endocrinology and metabolism, 2019, Volume: 63, Issue:5

    Thyroid cancer has been rapidly increasing in prevalence among humans in last 2 decades and is the most prevalent endocrine malignancy. Overall, thyroid-cancer patients have good rates of long-term survival, but a small percentage present poor outcome. Thyroid cancer aggressiveness is essentially related with thyroid follicular cell loss of differentiation and metastasis. The discovery of oncogenes that drive thyroid cancer (such as RET, RAS, and BRAF), and are aligned in the MAPK/ERK pathway has led to a new perspective of thyroid oncogenesis. The uncovering of additional oncogene-modulated signaling pathways revealed an intricate and active signaling cross-talk. Among these, microRNAs, which are a class of small, noncoding RNAs, expanded this cross-talk by modulating several components of the oncogenic network - thus establishing a new layer of regulation. In this context, TGFβ signaling plays an important role in cancer as a dual factor: it can exert an antimitogenic effect in normal thyroid follicular cells, and promote epithelial-to-mesenchymal transition, cell migration, and invasion in cancer cells. In this review, we explore how microRNAs influence the loss of thyroid differentiation and the increase in aggressiveness of thyroid cancers by regulating the dual function of TGFβ. This review provides directions for future research to encourage the development of new strategies and molecular approaches that can improve the treatment of aggressive thyroid cancer.

    Topics: Cell Transformation, Neoplastic; Disease Progression; Humans; MicroRNAs; Neoplasm Invasiveness; Neoplasm Metastasis; Signal Transduction; Thyroid Gland; Thyroid Neoplasms; Transforming Growth Factor beta

2019
Моlecular mechanisms of initiation of carcinogenesis in the testis.
    Experimental oncology, 2019, Volume: 41, Issue:3

    In this review, literature data on the study of precancerous changes in testicular tissue and molecular changes, as well as the influence of environmental factors that can initiate carcinogenesis, were analyzed and summarized for the future determination of early diagnosis of germ cell tumors of the testis and the development of preventive measures. The review also discusses the significant new changes presented in the Fourth Edition of the World Health Organization Classification of Urogenital Tumors, published in 2016, and modern concepts of the etiology and pathogenesis of these diseases. Among the environmental factors that can initiate carcinogenesis, the most noteworthy are the biological effects of low doses of ionizing radiation, such as the effect of radiation-induced genome instability, which increases the risk of carcinogenesis, the "bystander effect", and chronic oxidative stress. Disruption of ubiquitin-proteasomal proteolysis, impaired molecular-level components of the blood-testis barrier, and impaired regulatory action of TGF-β on the cell cycle can play a crucial role in the pathogenesis of male infertility and the initiation of carcinogenesis in the testis. The effect of low doses of ionizing radiation as an additional etiological factor leads to changes in the structural, as well as molecular, components of the testis, including epigenetic changes, which can be characterized as environmental pathomorphosis, which leads to impaired spermatogenesis and increased risk of malignancy. Summarizing the literature review data, we can state that patients with blocked spermatogenesis, in which atypical germ cell neoplasia in situ cells are detected in testicular tissue, constitute a group at increased risk of testicular carcinogenesis. The presence of additional etiological factors, such as chronic low doses of ionizing radiation, can initiate the progression of carcinogenesis in the testicle.

    Topics: Biomarkers; Blood-Testis Barrier; Cell Transformation, Neoplastic; Humans; Male; Models, Biological; Radiation, Ionizing; Spermatogenesis; Testicular Neoplasms; Testis; Tight Junction Proteins; Transforming Growth Factor beta; Ubiquitin

2019
Human Colorectal Cancer from the Perspective of Mouse Models.
    Genes, 2019, 10-11, Volume: 10, Issue:10

    Colorectal cancer (CRC) is a heterogeneous disease that includes both hereditary and sporadic types of tumors. Tumor initiation and growth is driven by mutational or epigenetic changes that alter the function or expression of multiple genes. The genes predominantly encode components of various intracellular signaling cascades. In this review, we present mouse intestinal cancer models that include alterations in the Wnt, Hippo, p53, epidermal growth factor (EGF), and transforming growth factor β (TGFβ) pathways; models of impaired DNA mismatch repair and chemically induced tumorigenesis are included. Based on their molecular biology characteristics and mutational and epigenetic status, human colorectal carcinomas were divided into four so-called consensus molecular subtype (CMS) groups. It was shown subsequently that the CMS classification system could be applied to various cell lines derived from intestinal tumors and tumor-derived organoids. Although the CMS system facilitates characterization of human CRC, individual mouse models were not assigned to some of the CMS groups. Thus, we also indicate the possible assignment of described animal models to the CMS group. This might be helpful for selection of a suitable mouse strain to study a particular type of CRC.

    Topics: Animals; Carcinogenesis; Cell Transformation, Neoplastic; Colonic Neoplasms; Colorectal Neoplasms; Disease Models, Animal; DNA Mismatch Repair; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Genes, p53; Hippo Signaling Pathway; Humans; Mice; Protein Serine-Threonine Kinases; Transforming Growth Factor beta; Wnt Signaling Pathway

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
The role of TGFβ in hematopoiesis and myeloid disorders.
    Leukemia, 2019, Volume: 33, Issue:5

    The role of transforming growth factor-β (TGFβ) signaling in embryological development and tissue homeostasis has been thoroughly characterized. Its canonical downstream cascade is well known, even though its true complexity and other non-canonical pathways are still being explored. TGFβ signaling has been described as an important pathway involved in carcinogenesis and cancer progression. In the hematopoietic compartment, the TGFβ pathway is an important regulator of proliferation and differentiation of different cell types and has been implicated in the pathogenesis of a diverse variety of bone marrow disorders. Due to its importance in hematological diseases, novel inhibitors of this pathway are being developed against a number of hematopoietic disorders, including myelodysplastic syndromes (MDS). In this review, we provide an overview of the TGFβ pathway, focusing on its role in hematopoiesis and impact on myeloid disorders. We will discuss therapeutic interventions with promising results against MDS.

    Topics: Animals; Cell Differentiation; Cell Transformation, Neoplastic; Clinical Studies as Topic; Disease Susceptibility; Drug Development; Hematopoiesis; Hematopoietic Stem Cells; Humans; Leukemia, Myeloid; Molecular Targeted Therapy; Myelodysplastic Syndromes; Signal Transduction; Transforming Growth Factor beta

2019
The concomitant apoptosis and EMT underlie the fundamental functions of TGF-β.
    Acta biochimica et biophysica Sinica, 2018, Jan-01, Volume: 50, Issue:1

    TGF-β's multipotent cellular effects and their relations are critical for TGF-β's pathophysiological functions. However, these effects may appear to be paradoxical in understanding TGF-β's functions. Apoptosis and epithelial-mesenchymal transition (EMT) are two fundamental events that are deeply linked to various physiological and disease-related processes. These two major cellular fates are subtly regulated and can be potently stimulated by TGF-β, which profoundly contribute to the biological roles of TGF-β. Moreover, these two events are also indirectly and directly correlated with TGF-β-mediated growth inhibition and are relevant to the current understanding of the roles of TGF-β in tumorigenesis and cancer progression. Although TGF-β-induced apoptosis and EMT can be singly independent cellular events, they can also be mutually exclusive but interrelated concomitant events in various cases. Thus, the modulation of apoptosis and EMT is essential for the seemingly paradoxical functions of TGF-β. However, the concomitant effect of TGF-β on apoptosis and EMT, the balance and regulated alterations of them are still been ignored or underestimated. This review focuses on the TGF-β-induced concomitant apoptosis and EMT. We aim to provide an insight in understanding their significance, balance, and modulation in TGF-β-mediated biological functions.

    Topics: Animals; Apoptosis; Cell Proliferation; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Humans; Models, Biological; Signal Transduction; Transforming Growth Factor beta

2018
TGF-beta signaling and its targeted therapy in gastrointestinal cancers.
    Discovery medicine, 2018, Volume: 26, Issue:142

    The transforming growth factor β (TGFβ) signaling pathway governs physiological homeostasis in the gastrointestinal system and its deregulation can lead to a diverse range of human pathologies including juvenile polyposis syndrome and tumor initiation, progression, and metastasis. In gastrointestinal malignancies, tumor cells evade the known tumor suppressive effects of TGFβ signaling through frequent inactivation of the pathway. Paradoxically, tumor cells utilize TGFβ-mediated regulation of epithelial-mesenchymal transition and immunomodulation to facilitate the invasive and migratory phenotype of gastrointestinal cancers and avoid immunosurveillance. The dichotomous role of TGFβ as both a tumor suppressor and tumor promoter has highly challenged research efforts to specifically target TGFβ signaling as a cancer therapy. The current preclinical approach is to inhibit TGFβ-mediated generation of a favorable microenvironment for tumor growth, invasion, and metastasis. Here, we overview the alterations of TGFβ signaling and its fundamental biological relevance in gastrointestinal tumorigenesis. We further discuss future perspectives for efficacious molecular targeted treatment of contextual TGFβ tumor-promoting effects in gastrointestinal cancers.

    Topics: Animals; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Gastrointestinal Neoplasms; Genes, Tumor Suppressor; Humans; Molecular Targeted Therapy; Signal Transduction; Transforming Growth Factor beta

2018
A Review of the Scaffold Protein Menin and its Role in Hepatobiliary Pathology.
    Gene expression, 2017, Jul-07, Volume: 17, Issue:3

    Multiple endocrine neoplasia type 1 (MEN1) is a familial cancer syndrome with neuroendocrine tumorigenesis of the parathyroid glands, pituitary gland, and pancreatic islet cells. The MEN1 gene codes for the canonical tumor suppressor protein, menin. Its protein structure has recently been crystallized, and it has been investigated in a multitude of other tissues. In this review, we summarize recent advancements in understanding the structure of the menin protein and its function as a scaffold protein in histone modification and epigenetic gene regulation. Furthermore, we explore its role in hepatobiliary autoimmune diseases, cancers, and metabolic diseases. In particular, we discuss how menin expression and function are regulated by extracellular signaling factors and nuclear receptor activation in various hepatic cell types. How the many signaling pathways and tissue types affect menin's diverse functions is not fully understood. We show that small-molecule inhibitors affecting menin function can shed light on menin's broad role in pathophysiology and elucidate distinct menin-dependent processes. This review reveals menin's often dichotomous function through analysis of its role in multiple disease processes and could potentially lead to novel small-molecule therapies in the treatment of cholangiocarcinoma or biliary autoimmune diseases.

    Topics: Animals; Autoimmune Diseases; Cell Transformation, Neoplastic; Cholangiocarcinoma; Epigenesis, Genetic; Fibrosis; Gene Expression Regulation, Neoplastic; Histone-Lysine N-Methyltransferase; Histones; Humans; Leukemia, Biphenotypic, Acute; Liver; Metabolic Diseases; MicroRNAs; Multiple Endocrine Neoplasia Type 1; Pancreas; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-jun; Signal Transduction; Transcription Factors; 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
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
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
Exosome-mediated microenvironment dysregulation in leukemia.
    Biochimica et biophysica acta, 2016, Volume: 1863, Issue:3

    The hematopoietic stem cell (HSC) niche is composed of a complex set of stromal support cells that maintain HSCs and promote normal hematopoiesis. We now know that molecular changes within the hematopoietic niche contribute to leukemia development. Leukemia cells often reorganize the hematopoietic niche to promote and support their own survival and growth. Here we will summarize recent works that decipher the normal hematopoietic niche cellular components and describe how the leukemia-transformed niche contributes to hematological malignances. Finally, we will discuss recent publications that highlight a possible role for exosomes in the leukemia-induced niche reorganization. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.

    Topics: Cell Transformation, Neoplastic; Exosomes; Hematopoietic Stem Cells; Humans; Leukemia; Models, Biological; Neoplastic Stem Cells; Receptor, Notch1; Stem Cell Niche; Transforming Growth Factor beta

2016
Keratoacanthoma: a distinct entity?
    Experimental dermatology, 2016, Volume: 25, Issue:2

    Keratoacanthoma (KA) are common but exceptional benign tumors, often appearing on sun-exposed areas of light skinned people and showing spontaneous resolution. The goal of this study was to review existing literature, to point out the etiological complexity of KA biology and to answer the controversial debate if or not KA is a distinct entity or a variant of squamous cell carcinoma (SCC). Relying on recent results, we highlight that KA is an individual lesion with a unique molecular signature caused by alterations in the TGFβ signalling pathway. These recent findings will help to understand the nature of KA and to develop new reliable diagnostic tools, simplifying the discrimination of the histologically similar KA and SCC.

    Topics: Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Comparative Genomic Hybridization; Diagnosis, Differential; Disease Progression; Genetic Predisposition to Disease; Humans; Keratoacanthoma; Neoplasm Proteins; Neoplasms, Radiation-Induced; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Skin Diseases; Skin Neoplasms; Sunlight; Transforming Growth Factor beta; Ultraviolet Rays

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
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
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
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
TAK1 regulates hepatic cell survival and carcinogenesis.
    Journal of gastroenterology, 2014, Volume: 49, Issue:2

    TGF-β-activated kinase 1 (TAK1 or MAP3K7) is an intracellular hub molecule that regulates both nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways that play key roles in development, cell survival, immune response, metabolism, and carcinogenesis. TAK1 activity is tightly regulated by its binding proteins, TAB1 and TAB2/TAB3, as well as by post-translational modification including ubiquitination and phosphorylation. Accumulating evidence demonstrates that TAK1 plays a role in tumor initiation, progression, and metastasis as a tumor prompter or tumor suppressor. An understanding of the role of TAK1 in liver physiology and diseases is required for the development of therapeutic agencies targeting TAK1. In this review, we highlight the activation mechanism and pathophysiological roles of TAK1 in the liver.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Cell Survival; Cell Transformation, Neoplastic; DNA Repair; Hepatocytes; Humans; Intracellular Signaling Peptides and Proteins; Liver Neoplasms; MAP Kinase Kinase Kinases; MAP Kinase Signaling System; Molecular Targeted Therapy; NF-kappa B; Transforming Growth Factor beta

2014
The roles of transforming growth factor-β, Wnt, Notch and hypoxia on liver progenitor cells in primary liver tumours (Review).
    International journal of oncology, 2014, Volume: 44, Issue:4

    Primary liver tumours have a high incidence and mortality. The most important forms are hepatocellular carcinoma and intrahepatic cholangiocarcinoma, both can occur together in the mixed phenotype hepatocellular-cholangiocarcinoma. Liver progenitor cells (LPCs) are bipotential stem cells activated in case of severe liver damage and are capable of forming both cholangiocytes and hepatocytes. Possibly, alterations in Wnt, transforming growth factor-β, Notch and hypoxia pathways in these LPCs can cause them to give rise to cancer stem cells, capable of driving tumourigenesis. In this review, we summarize and discuss current knowledge on the role of these pathways in LPC activation and differentiation during hepatocarcinogenesis.

    Topics: Bile Duct Neoplasms; Bile Ducts, Intrahepatic; Carcinoma, Hepatocellular; Cell Differentiation; Cell Hypoxia; Cell Transformation, Neoplastic; Cholangiocarcinoma; Humans; Liver; Liver Neoplasms; Receptors, Notch; Stem Cells; Transforming Growth Factor beta; Wnt Proteins; Wnt Signaling Pathway

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
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
HAS2 and CD44 in breast tumorigenesis.
    Advances in cancer research, 2014, Volume: 123

    Metastatic spread of breast cancer cells, facilitated by the epithelial-mesenchymal transition (EMT) process, is responsible for the majority of breast cancer mortality. Increased levels of hyaluronan due to deregulation of hyaluronan-synthesizing enzymes, like HAS2, and expression of CD44, the key receptor for hyaluronan, are correlated to poor outcome of patients with basal-like breast cancer. TGFβ induces HAS2 and CD44, both of which are required in the course of efficient TGFβ-induced EMT processes by mammary epithelial cells. Elucidation of the molecular mechanisms underlying tumor-stroma interactions in breast cancer including the regulation of HAS2 and CD44 expression may contribute to the development of better strategies to treat breast cancer patients.

    Topics: Breast Neoplasms; Cell Adhesion; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Disease Progression; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Glucuronosyltransferase; Humans; Hyaluronan Receptors; Hyaluronan Synthases; Hyaluronic Acid; Neoplasm Metastasis; Signal Transduction; 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
TRAF4 mediates activation of TGF-β signaling and is a biomarker for oncogenesis in breast cancer.
    Science China. Life sciences, 2014, Volume: 57, Issue:12

    The tumor-promoting arm of transforming growth factor beta (TGF-β) receptor signaling contributes to advanced cancer progression and is considered a master regulator of breast cancer metastasis. In mammals, there are six distinct members in the tumor-necrosis factor receptor (TNFR)-associated factor (TRAF) family (TRAF1-TRAF6), with the function of TRAF4 not being extensively studied in the past decade. Although numerous studies have suggested that there is elevated TRAF4 expression in human cancer, it is still unknown in which oncogenic pathway TRAF4 is mainly implicated. This review highlights TGF-β-induced SMAD-dependent signaling and non-SMAD signaling as the major pathways regulated by TRAF4 involved in breast cancer metastasis.

    Topics: Biomarkers, Tumor; Breast Neoplasms; Cell Transformation, Neoplastic; Female; Humans; Neoplasm Metastasis; Prognosis; Signal Transduction; Smad Proteins; TNF Receptor-Associated Factor 4; Transforming Growth Factor beta; Ubiquitin-Protein Ligases

2014
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
Cholangiocarcinoma: risk factors, environmental influences and oncogenesis.
    Annals of clinical and laboratory science, 2013,Spring, Volume: 43, Issue:2

    Cholangiocarcinoma (CCA) is one of the most frequent malignant epithelial liver tumors after hepatocellular carcinoma (HCC). Its incidence seems to be increasing worldwide, although risk factors are heterogeneous and differ globally. Although diagnostic and therapeutic medicine have advanced in several countries, tackling this tumor remains a challenge. The causes of CCA's increasing incidence are likely a differential increment of some factors according to the geographical area, which will be considered in this review. Environment-linked risk factors may play a critical role in the carcinogenesis. Liver flukes may play a major role in East Asia, while exposure to chemical compounds, such as naphthenic acids, has been postulated as a source of the rate increase in Western countries. Carcinogenesis is variable and confounding factors also need to be taken into account. Carcinogenesis depends on a sequential process and most probably involves both cholestasis and chronic inflammation as promoting steps after induction. The release and interaction of interleukin-6 (IL-6), transforming growth factor beta (TGF-beta), tumor necrosis factor alpha (TNF-alpha), and platelet-derived growth factor (PDGF) are at the basis of the proliferation of biliary epithelial cells or cholangiocytes. Additional steps for the final development of CCA may also involve an increase of the mutation rate of tumor suppressor genes, such as TP53, and the evasion of apoptosis.

    Topics: Animals; Bile Duct Neoplasms; Bile Ducts, Intrahepatic; Biliary Tract; Cell Transformation, Neoplastic; Cholangiocarcinoma; Cholelithiasis; Fasciola hepatica; Hepatitis C; Humans; Incidence; Interleukin-6; Liver; Petroleum; Platelet-Derived Growth Factor; Risk Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

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
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
Transforming growth factor-β signaling in tumor initiation, progression and therapy in breast cancer: an update.
    Cell and tissue research, 2012, Volume: 347, Issue:1

    Transforming growth factor-β (TGF-β) is a ubiquitous cytokine playing an essential role in cell proliferation, differentiation, apoptosis, adhesion and invasion, as well as in cellular microenvironment. In malignant diseases, TGF-β signaling features a growth inhibitory effect at an early stage but aggressive oncogenic activity at the advanced malignant state. Here, we update the current understanding of TGF-β signaling in cancer development and progression with a focus on breast cancer. We also review the current approaches of TGF-β signaling-targeted therapeutics for human malignancies.

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

2012
TGF-beta signalling in colon carcinogenesis.
    Cancer letters, 2012, Jan-01, Volume: 314, Issue:1

    Colorectal cancer remains the most common cancer and the second leading cause of cancer mortality in Europe. There are a number of pathways that have been implicated in colorectal carcinogenesis, including TGF-beta (TGF-β)/Smad signalling pathway. The TGF-β pathway is involved in several biological processes, including cell proliferation, differentiation, migration and apoptosis. Here we review the role of TGF-β signalling cascade in colorectal carcinogenesis and provide some new molecular insights that may aid efforts towards targeted antitumor therapies.

    Topics: Animals; Cell Transformation, Neoplastic; Colonic Neoplasms; Humans; Mutation; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2012
The roles of TGF-β signaling in carcinogenesis and breast cancer metastasis.
    Breast cancer (Tokyo, Japan), 2012, Volume: 19, Issue:2

    Transforming growth factor-β (TGF-β) ligand is a multifunctional growth factor that regulates various cell behavior, such as cell proliferation, differentiation, migration, and apoptosis. Because TGF-β is a potent growth inhibitor, abnormalities in TGF-β signaling result in carcinogenesis. In addition to tumor suppressor function, TGF-β acts as an oncogenic factor. In particular, TGF-β signaling plays an important role during metastasis of breast cancer. Recently, epithelial-mesenchymal transition (EMT) has been shown to confer malignant properties such as cell motility and invasiveness to cancer cells and plays crucial roles during cancer metastasis. Moreover, breast stem-like cells exhibit EMT properties. Because TGF-β is a potent regulator of EMT as well as cell stemness, TGF-β signaling might play a crucial role in the regulation of breast cancer stem cells.

    Topics: Breast Neoplasms; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Female; Humans; Neoplasm Metastasis; Signal Transduction; Transforming Growth Factor beta

2012
Crosstalk between TGF-β signaling and the microRNA machinery.
    Trends in pharmacological sciences, 2012, Volume: 33, Issue:7

    The activin/transforming growth factor-β (TGF-β) pathway plays an important role in tumorigenesis either by its tumor suppressor or tumor promoting effect. Loss of members of the TGF-β signaling by somatic mutations or epigenetic events, such as DNA methylation or regulation by microRNA (miRNA), may affect the signaling process. Most members of the TGF-β pathway are known to be targeted by one or more miRNAs. In addition, the biogenesis of miRNAs is also regulated by TGF-β both directly and through SMADs. Based on these interactions, it appears that autoregulatory feedback loops between TGF-β and miRNAs influence the fate of tumor cells. Our aim is to review the crosstalk between TGF-β signaling and the miRNA machinery to highlight potential novel therapeutic targets.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Epigenesis, Genetic; Humans; Mice; MicroRNAs; Mutation; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2012
Transforming growth Factor-Beta-Induced Protein (TGFBI)/(βig-H3): a matrix protein with dual functions in ovarian cancer.
    International journal of molecular sciences, 2012, Volume: 13, Issue:8

    Transforming growth factor-beta-induced protein (TGFBI, also known as βig-H3 and keratoepithelin) is an extracellular matrix protein that plays a role in a wide range of physiological and pathological conditions including diabetes, corneal dystrophy and tumorigenesis. Many reports indicate that βig-H3 functions as a tumor suppressor. Loss of βig-H3 expression has been described in several cancers including ovarian cancer and promoter hypermethylation has been identified as an important mechanism for the silencing of the TGFBI gene. Our recent findings that βig-H3 is down-regulated in ovarian cancer and that high concentrations of βig-H3 can induce ovarian cancer cell death support a tumor suppressor role. However, there is also convincing data in the literature reporting a tumor-promoting role for βig-H3. We have shown βig-H3 to be abundantly expressed by peritoneal cells and increase the metastatic potential of ovarian cancer cells by promoting cell motility, invasion, and adhesion to peritoneal cells. Our findings suggest that βig-H3 has dual functions and can act both as a tumor suppressor or tumor promoter depending on the tumor microenvironment. This article reviews the current understanding of βig-H3 function in cancer cells with particular focus on ovarian cancer.

    Topics: Cell Adhesion; Cell Movement; Cell Transformation, Neoplastic; DNA Methylation; Extracellular Matrix; Extracellular Matrix Proteins; Female; Humans; Ovarian Neoplasms; Transforming Growth Factor beta

2012
[Pathogenesis of colitis-associated neoplasms].
    Der Pathologe, 2012, Volume: 33 Suppl 2

    It is sometimes difficult but finally possible to distinguish colitis-associated neoplasms from sporadic neoplasms. The frequency of detection of precursor lesions of carcinoma (e.g. dysplasia, intraepithelial neoplasia and adenoma) has increased in recent years, which is most probably due to better endoscopic detection and thus improved histological diagnosis. Carcinogenesis of colitis-associated neoplasms is different from carcinogenesis in sporadic neoplasms because mutations and epigenetic changes are different or may occur at a different point in time. In the present article, these differences will be described and placed in context with carcinogenesis in ulcerative colitis.

    Topics: Adenoma; Carcinoma in Situ; Cell Transformation, Neoplastic; Chromosome Aberrations; Colitis, Ulcerative; Colonic Neoplasms; DNA Methylation; DNA Mutational Analysis; Epigenesis, Genetic; Humans; Intestinal Mucosa; Microsatellite Instability; Neoplasm Invasiveness; Neoplasm Staging; Oxidative Stress; Precancerous Conditions; Reactive Oxygen Species; Transforming Growth Factor beta

2012
TGF-β signaling in onset and progression of hepatocellular carcinoma.
    Digestive diseases (Basel, Switzerland), 2012, Volume: 30, Issue:5

    Transforming growth factor (TGF)-β is a central regulator in chronic liver disease, contributing to all stages of disease progression from initial liver injury through inflammation and fibrosis to cirrhosis and hepatocellular carcinoma. Liver damage-induced levels of active TGF-β enhance hepatocyte destruction and mediate hepatic stellate cell and fibroblast activation resulting in a wound-healing response, including myofibroblast generation and extracellular matrix deposition. Further evidence points to a decisive role of cytostatic and apoptotic functions mediated on hepatocytes, which is critical for the control of liver mass, with loss of TGF-β activities resulting in hyperproliferative disorders and cancer. This concept is based on studies that describe a bipartite role of TGF-β with tumor suppressor functions at early stages of liver damage and regeneration, whereas during cancer progression TGF-β may turn from a tumor suppressor into a tumor promoter that exacerbates invasive and metastatic behavior. We have delineated this molecular switch of the pathway from cytostatic to tumor promoting in further detail and identify activation of survival signaling pathways in hepatocytes as a most critical requirement. Targeting the TGF-β signaling pathway has been explored to inhibit liver disease progression. While interfering with TGF-β signaling in various short-term animal models has demonstrated promising results, liver disease progression in humans is a process of decades with different phases in which TGF-β or its targeting may have both beneficial and adverse outcomes. We emphasize that, in order to achieve therapeutic effects, targeting TGF-β signaling in the right cell type at the right time is required.

    Topics: Animals; Apoptosis; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Disease Progression; Humans; Liver Neoplasms; Mice; Rats; Signal Transduction; Transforming Growth Factor beta

2012
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
Noncanonical TGF-β signaling during mammary tumorigenesis.
    Journal of mammary gland biology and neoplasia, 2011, Volume: 16, Issue:2

    Breast cancer is a heterogeneous disease comprised of at least five major tumor subtypes that coalesce as the second leading cause of cancer death in women in the United States. Although metastasis clearly represents the most lethal characteristic of breast cancer, our understanding of the molecular mechanisms that govern this event remains inadequate. Clinically, ~30% of breast cancer patients diagnosed with early-stage disease undergo metastatic progression, an event that (a) severely limits treatment options, (b) typically results in chemoresistance and low response rates, and (c) greatly contributes to aggressive relapses and dismal survival rates. Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that regulates all phases of postnatal mammary gland development, including branching morphogenesis, lactation, and involution. TGF-β also plays a prominent role in suppressing mammary tumorigenesis by preventing mammary epithelial cell (MEC) proliferation, or by inducing MEC apoptosis. Genetic and epigenetic events that transpire during mammary tumorigenesis conspire to circumvent the tumor suppressing activities of TGF-β, thereby permitting late-stage breast cancer cells to acquire invasive and metastatic phenotypes in response to TGF-β. Metastatic progression stimulated by TGF-β also relies on its ability to induce epithelial-mesenchymal transition (EMT) and the expansion of chemoresistant breast cancer stem cells. Precisely how this metamorphosis in TGF-β function comes about remains incompletely understood; however, recent findings indicate that the initiation of oncogenic TGF-β activity is contingent upon imbalances between its canonical and noncanonical signaling systems. Here we review the molecular and cellular contributions of noncanonical TGF-β effectors to mammary tumorigenesis and metastatic progression.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Disease Progression; Epithelial-Mesenchymal Transition; Female; Humans; Mammary Glands, Animal; Mammary Glands, Human; Mammary Neoplasms, Experimental; Signal Transduction; 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
Molecular pathogenesis of hepatocellular carcinoma: altering transforming growth factor-β signaling in hepatocarcinogenesis.
    Digestive diseases (Basel, Switzerland), 2011, Volume: 29, Issue:3

    Hepatocellular carcinoma (HCC) occurs subsequent to liver injury, where regenerative hepatocytes develop into a dysplastic nodule and then early HCC, supporting the multistep hepatocarcinogenesis theory. Molecular alterations such as the p53 mutation, p16 gene silencing, and AKT signaling activation are found in the late stage of HCC progression. The overexpression of some marker molecules is observed at the early stage. Transforming growth factor-β (TGF-β), a potent inhibitor of cell proliferation, is frequently overexpressed in HCC, although the role of TGF-β signaling during HCC development remains controversial. We previously reported that HCC cells show TGF-β receptor-dependent growth inhibition in response to TGF-β. Also, reduced TGF-β receptor II in HCC correlates with intrahepatic metastasis and shorter time-to-recurrence, suggesting a role of TGF-β signaling in tumor suppression. In contrast, TGF-β overexpression in HCC is known to correlate with malignant potential, suggesting a role in tumor promotion. Enhanced formation of stroma is a feature of advanced HCC, and TGF-β also promotes the proliferation of stromal fibroblasts. The microenvironment produced via tumor-stromal interactions may be the key to the modulation of the dual roles of TGF-β signaling in HCC progression.

    Topics: Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Humans; Liver Neoplasms; Models, Biological; Signal Transduction; Transforming Growth Factor beta

2011
Intestinal inflammation and colorectal cancer: a double-edged sword?
    World journal of gastroenterology, 2011, Jul-14, Volume: 17, Issue:26

    Chronic inflammation is thought to be the leading cause of many human cancers including colorectal cancer (CRC). Accordingly, epidemiologic and clinical studies indicate that patients affected by ulcerative colitis and Crohn's disease, the two major forms of inflammatory bowel disease, have an increased risk of developing CRC. In recent years, the role of immune cells and their products have been shown to be pivotal in initiation and progression of colitis-associated CRC. On the other hand, activation of the immune system has been shown to cause dysplastic cell elimination and cancer suppression in other settings. Clinical and experimental data herein reviewed, while confirming chronic inflammation as a risk factor for colon carcinogenesis, do not completely rule out the possibility that under certain conditions the chronic activation of the mucosal immune system might protect from colonic dysplasia.

    Topics: Animals; Cell Transformation, Neoplastic; Chemokines; Colorectal Neoplasms; Humans; Immune System; Inflammation; Inflammatory Bowel Diseases; Interleukin-10; Interleukin-6; Intestines; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2011
Role of TGF-β and the tumor microenvironment during mammary tumorigenesis.
    Gene expression, 2011, Volume: 15, Issue:3

    Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that functions to inhibit mammary tumorigenesis by directly inducing mammary epithelial cells (MECs) to undergo cell cycle arrest or apoptosis, and to secrete a variety of cytokines, growth factors, and extracellular matrix proteins that maintain cell and tissue homeostasis. Genetic and epigenetic events that transpire during mammary tumorigenesis typically inactivate the tumor suppressing activities of TGF-beta and ultimately confer this cytokine with tumor promoting activities, including the ability to stimulate breast cancer invasion, metastasis, angiogenesis, and evasion from the immune system. This dramatic conversion in TGF-beta function is known as the "TGF-beta paradox" and reflects a variety of dynamic alterations that occur not only within the developing mammary carcinoma, but also within the cellular and structural composition of its accompanying tumor microenvironment. Recent studies have begun to elucidate the critical importance of mammary tumor microenvironments in manifesting the TGF-beta paradox and influencing the response of developing mammary carcinomas to TGF-beta. Here we highlight recent findings demonstrating the essential function of tumor microenvironments in regulating the oncogenic activities of TGF-beta and its stimulation of metastatic progression during mammary tumorigenesis.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Endothelial Cells; Epithelial Cells; Female; Fibroblasts; Humans; Mammary Neoplasms, Experimental; Neoplasm Metastasis; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2011
Targeting the transforming growth factor-beta signalling pathway in metastatic cancer.
    European journal of cancer (Oxford, England : 1990), 2010, Volume: 46, Issue:7

    Transforming growth factor (TGF)-beta signalling plays a dichotomous role in tumour progression, acting as a tumour suppressor early and as a pro-metastatic pathway in late-stages. There is accumulating evidence that advanced-stage tumours produce excessive levels of TGF-beta, which acts to promote tumour growth, invasion and colonisation of secondary organs. In light of the pro-metastasis function, many strategies are currently being explored to antagonise the TGF-beta pathway as a treatment for metastatic cancers. Strategies such as using large molecule ligand traps, reducing the translational efficiency of TGF-beta ligands using antisense technology, and antagonising TGF-beta receptor I/II kinase function using small molecule inhibitors are the most prominent methods being explored today. Administration of anti-TGF-beta therapies alone, or in combination with immunosuppressive or cytotoxic therapies, has yielded promising results in the preclinical and clinical settings. Despite these successes, the temporal- and context-dependent roles of TGF-beta signalling in cancer has made it challenging to define patient subgroups that are most likely to respond, and the therapeutic regimens that will be most effective in the clinic. Novel mouse models and diagnostic tools are being developed today to circumvent these issues, which may potentially expedite anti-TGF-beta drug development and clinical application.

    Topics: Animals; Antibodies, Monoclonal; Antineoplastic Agents; Antisense Elements (Genetics); Cell Transformation, Neoplastic; Combined Modality Therapy; Disease Progression; Humans; Mice; Neoplasm Metastasis; Neoplasm Proteins; Signal Transduction; Transforming Growth Factor beta

2010
SnoN in mammalian development, function and diseases.
    Current opinion in pharmacology, 2010, Volume: 10, Issue:6

    SnoN (Ski-novel protein) was discovered as a nuclear proto-oncogene on the basis of its ability to induce transformation of chicken and quail embryonic fibroblasts. As a crucial negative regulator of transforming growth factor-β (TGF-β) signaling and also an activator of p53, it plays an important role in regulating cell proliferation, senescence, apoptosis, and differentiation. Recent studies of its expression patterns and functions in mouse models and mammalian cells have revealed important functions of SnoN in normal epithelial development and tumorigenesis. Evidence suggests that SnoN has both pro-oncogenic and anti-oncogenic functions by modulating multiple signaling pathways. These studies suggest that SnoN may have broad functions in the development and homeostasis of embryonic and postnatal tissues.

    Topics: Animals; Cell Transformation, Neoplastic; Embryonic Development; Female; Gene Expression Regulation; Genes, p53; Humans; Intracellular Signaling Peptides and Proteins; Mice; Molecular Targeted Therapy; Proto-Oncogene Mas; Proto-Oncogene Proteins; Proto-Oncogenes; Signal Transduction; Trans-Activators; Transforming Growth Factor beta

2010
Role of transforming growth factor-β in inflammatory bowel disease and colitis-associated colon cancer.
    Inflammatory bowel diseases, 2010, Volume: 16, Issue:11

    Transforming growth factor-β (TGF-β) plays a central role in a wide array of cellular functions including control of cell growth and differentiation, embryonic development, wound healing, angiogenesis, and immune regulation. In the gastrointestinal tract, TGF-β can either promote or suppress inflammation and cancer formation. This report reviews recent data on the role of TGF-β in the pathogenesis of inflammatory bowel disease and how TGF-β might contribute to the cancer risk associated with chronic inflammation of the gut.

    Topics: Animals; Cell Proliferation; Cell Transformation, Neoplastic; Colitis; Colonic Neoplasms; Humans; Inflammatory Bowel Diseases; Lymphocyte Activation; Mice; Th17 Cells; Transforming Growth Factor beta

2010
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
The tale of transforming growth factor-beta (TGFbeta) signaling: a soigné enigma.
    IUBMB life, 2009, Volume: 61, Issue:10

    Transforming growth factor-beta (TGFbeta) is a secreted cytokine, which intricately controls a plethora of physiological and pathological processes during development and carcinogenesis. TGFbeta exerts antiproliferative effects and functions as a tumor suppressor during early stages of tumorigenesis, whereas at later stages it functions as a tumor promoter aiding in metastatic progression through an autocrine TGFbeta loop. Intricate knowledge of TGFbeta signaling and its regulation are still evolving. In this review, we make an attempt to showcase the associated enigma of TGFbeta signaling in its dual functional role as tumor suppressor and metastatic promoter during early and late stages of carcinogenesis, respectively.

    Topics: Animals; Cell Transformation, Neoplastic; Epithelial Cells; Forecasting; Humans; Mesoderm; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Suppressor Proteins

2009
Mechanisms of the epithelial-mesenchymal transition by TGF-beta.
    Future oncology (London, England), 2009, Volume: 5, Issue:8

    The formation of epithelial cell barriers results from the defined spatiotemporal differentiation of stem cells into a specialized and polarized epithelium, a process termed mesenchymal-epithelial transition. The reverse process, epithelial-mesenchymal transition (EMT), is a metastable process that enables polarized epithelial cells to acquire a motile fibroblastoid phenotype. Physiological EMT also plays an essential role in promoting tissue healing, remodeling or repair in response to a variety of pathological insults. On the other hand, pathophysiological EMT is a critical step in mediating the acquisition of metastatic phenotypes by localized carcinomas. Although metastasis clearly is the most lethal aspect of cancer, our knowledge of the molecular events that govern its development, including those underlying EMT, remain relatively undefined. Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that oversees and directs all aspects of cell development, differentiation and homeostasis, as well as suppresses their uncontrolled proliferation and transformation. Quite dichotomously, tumorigenesis subverts the tumor suppressing function of TGF-beta, and in doing so, converts TGF-beta to a tumor promoter that stimulates pathophysiological EMT and metastasis. It therefore stands to reason that determining how TGF-beta induces EMT in developing neoplasms will enable science and medicine to produce novel pharmacological agents capable of preventing its ability to do so, thereby improving the clinical course of cancer patients. Here we review the cellular, molecular and microenvironmental mechanisms used by TGF-beta to mediate its stimulation of EMT in normal and malignant cells.

    Topics: Animals; Cell Differentiation; Cell Transformation, Neoplastic; Epithelium; Humans; Mesoderm; Metaplasia; Transforming Growth Factor beta

2009
The relationship of endometriosis and ovarian malignancy: a review.
    Fertility and sterility, 2008, Volume: 90, Issue:5

    To review the malignant potential of endometriosis based on epidemiologic, histopathologic, and molecular data.. Literature review.. The pathogenesis of endometriosis remains unclear. The histopathologic development of endometriosis has undergone long-term investigation. Studies have confirmed histologic transition from benign endometriosis to ovarian malignancy, including malignant transformation of extraovarian endometriosis. The prevalence of endometriosis in patients with epithelial ovarian cancer, especially in endometrioid and clear cell types, has been confirmed to be higher than in the general population. Ovarian cancers and adjacent endometriotic lesions have shown common genetic alterations, such as PTEN, p53, and bcl gene mutations, suggesting a possible malignant genetic transition spectrum. Furthermore, endometriosis has been associated with a chronic inflammatory state leading to cytokine release. These cytokines act in a complex system in which they induce or repress their own synthesis and can cause unregulated mitotic division, growth and differentiation, and migration or apoptosis similar to malignant mechanisms.. The malignant potential of endometriosis holds serious implications for management, such as the need for earlier and more meticulous surgical intervention for complete disease treatment.

    Topics: Cell Transformation, Neoplastic; Endometriosis; Female; Gene Expression Regulation, Neoplastic; Genomic Instability; Humans; Incidence; Intercellular Signaling Peptides and Proteins; Interleukin-1; Interleukin-8; Mutation; Ovarian Neoplasms; Prevalence; Transforming Growth Factor beta; Treatment Outcome; Tumor Necrosis Factor-alpha

2008
TGF-beta in neural stem cells and in tumors of the central nervous system.
    Cell and tissue research, 2008, Volume: 331, Issue:1

    Mechanisms that regulate neural stem cell activity in the adult brain are tightly coordinated. They provide new neurons and glia in regions associated with high cellular and functional plasticity, after injury, or during neurodegeneration. Because of the proliferative and plastic potential of neural stem cells, they are currently thought to escape their physiological control mechanisms and transform to cancer stem cells. Signals provided by proteins of the transforming growth factor (TGF)-beta family might represent a system by which neural stem cells are controlled under physiological conditions but released from this control after transformation to cancer stem cells. TGF-beta is a multifunctional cytokine involved in various physiological and patho-physiological processes of the brain. It is induced in the adult brain after injury or hypoxia and during neurodegeneration when it modulates and dampens inflammatory responses. After injury, although TGF-beta is neuroprotective, it may limit the self-repair of the brain by inhibiting neural stem cell proliferation. Similar to its effect on neural stem cells, TGF-beta reveals anti-proliferative control on most cell types; however, paradoxically, many brain tumors escape from TGF-beta control. Moreover, brain tumors develop mechanisms that change the anti-proliferative influence of TGF-beta into oncogenic cues, mainly by orchestrating a multitude of TGF-beta-mediated effects upon matrix, migration and invasion, angiogenesis, and, most importantly, immune escape mechanisms. Thus, TGF-beta is involved in tumor progression. This review focuses on TGF-beta and its role in the regulation and control of neural and of brain-cancer stem cells.

    Topics: Animals; Cell Transformation, Neoplastic; Central Nervous System Neoplasms; Humans; Neurogenesis; Neurons; Stem Cells; Transforming Growth Factor beta

2008
Context-dependent regulation of cutaneous immunological responses by TGFbeta1 and its role in skin carcinogenesis.
    Carcinogenesis, 2008, Volume: 29, Issue:1

    Transforming growth factor beta1 (TGFbeta1) signaling plays a critical role in skin carcinogenesis. While most studies have focused on TGFbeta1 signaling and response in keratinocytes, it is now becoming clear that the interaction of keratinocyte-derived TGFbeta1 with cells of the immune system has an equally important role in tumor development. Tumors form within the context of innate and adaptive immune responses and studies in skin and skin carcinogenesis models have provided important insight into the impact of context-dependent pro-inflammatory and immunosuppressive actions of TGFbeta1 on tumor development. Indeed, the paradigm of TGFbeta1 duality is clearly evident in its ability to both promote and inhibit inflammatory responses. Recent studies have begun to shed new light on the molecular basis for these actions and to provide insight into how these may contribute to context-dependent effects of TGFbeta1 on carcinogenesis in the skin and other epithelial tissues.

    Topics: Animals; Cell Transformation, Neoplastic; Humans; Skin Neoplasms; Transforming Growth Factor beta

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
Nodal signaling: developmental roles and regulation.
    Development (Cambridge, England), 2007, Volume: 134, Issue:6

    Nodal-related ligands of the transforming growth factor-beta (TGFbeta) superfamily play central roles in patterning the early embryo during the induction of mesoderm and endoderm and the specification of left-right asymmetry. Additional roles for this pathway in the maintenance of embryonic stem cell pluripotency and in carcinogenesis have been uncovered more recently. Consistent with its crucial developmental functions, Nodal signaling is tightly regulated by diverse mechanisms including the control of ligand processing, utilization of co-receptors, expression of soluble antagonists, as well as positive- and negative-feedback activities.

    Topics: Animals; Body Patterning; Cell Transformation, Neoplastic; Embryonic Development; Ligands; Mice; Nodal Protein; Receptors, Growth Factor; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2007
Chemokines and squamous cancer of the head and neck: targets for therapeutic intervention?
    Expert review of anticancer therapy, 2007, Volume: 7, Issue:3

    The biological properties of squamous carcinoma cells are intimately regulated by a multitude of cytokines and growth factors; the most well studied of these include epidermal growth factor receptor agonists and members of the transforming growth factor-beta family. The recent explosion of research in the field of chemokine function as a mediator of tumor progression has led to the possibility that these small, immunomodulatory proteins also play key roles in squamous carcinogenesis and may, therefore, be potential targets for novel therapeutic approaches.

    Topics: Amino Acid Motifs; Amino Acid Sequence; Antineoplastic Agents; Carcinoma, Squamous Cell; Cell Survival; Cell Transformation, Neoplastic; Chemokines; Chemokines, CXC; Disease Progression; Drug Design; ErbB Receptors; Head and Neck Neoplasms; Humans; Molecular Sequence Data; Neoplasm Invasiveness; Neoplasm Proteins; Neovascularization, Pathologic; Receptors, Chemokine; Sequence Alignment; Sequence Homology, Amino Acid; Signal Transduction; 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
Roles of Smad3 in TGF-beta signaling during carcinogenesis.
    Critical reviews in eukaryotic gene expression, 2007, Volume: 17, Issue:4

    Signaling of transforming growth factor beta (TGF-beta) is mediated through a heteromeric complex of two types of transmembrane receptors and downstream intracellular proteins known as Smads. Alterations of TGF-beta signaling underlie various forms of human cancer and developmental diseases. Human genetic studies have revealed both point mutations and deletions of Smad2 or Smad4 in several types of cancers. However, the role of Smad3 in tumorigenesis is not clear. Recent data indicate that Smad3 also functions as a tumor suppressor by inhibiting cell proliferation and promoting apoptosis. In addition, Smad3 is essential for TGF-beta-mediated immune suppression, and it plays an important role in regulating transcriptional responses that are favorable to metastasis. Therefore, through regulating different transcriptional responses, Smad3 functions as both a negative and positive regulator of carcinogenesis depending on cell type and clinical stage of the tumor.

    Topics: Animals; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Humans; Liver Neoplasms; Mice; Mutation; Neoplasm Metastasis; Signal Transduction; Smad3 Protein; 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
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
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
Growth factor signalling in prostatic growth: significance in tumour development and therapeutic targeting.
    British journal of pharmacology, 2006, Volume: 147 Suppl 2

    The intricate balance maintained between cell growth and proliferation factors and apoptosis-inducing factors is fundamental to the regulation of prostate growth. Disruptions in this homeostasis often trigger the loss of apoptosis and the over-expression of factors promoting cell survival and proliferation, inevitably leading to tumorigenesis and cancer. Deregulation of prostate growth during prostate cancer development and progression is characterized by apoptotic evasion, uncontrolled proliferation, and increased invasive potential. Thus, in advanced stages of disease progression, surviving prostate tumour cells acquire the ability to migrate and invade heterotopic tissues, with the bone and lymph nodes being the most common sites for human prostate cancer metastasis. The challenges in the implementation of effective therapeutic strategies for the treatment of advanced metastatic prostate cancer reflect the multidimensional nature and functional significance of antiapoptotic pathways in the emergence of therapeutic resistance of prostate tumours. In this chapter, we discuss the current understanding of the molecular mechanisms governing growth factor signalling pathways with often overlapping functions that contribute to loss of apoptosis control and activation of cell proliferation towards aggressive prostate tumorigenic growth and metastatic behaviour. While a full understanding of the prosurvival characteristics of these growth factor pathways is still evolving, the impact that growth factors such a epidermal growth factor and transforming growth factor-beta can be recognized by the vigorous attempts at therapeutic targeting of their key signalling steps.

    Topics: Animals; Antineoplastic Agents; Apoptosis; Cell Proliferation; Cell Transformation, Neoplastic; Fibroblast Growth Factors; Humans; Insulin-Like Growth Factor I; Intercellular Signaling Peptides and Proteins; Male; Neovascularization, Pathologic; Prostate; Prostatic Neoplasms; Signal Transduction; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2006
Smad3 phosphoisoform-mediated signaling during sporadic human colorectal carcinogenesis.
    Histology and histopathology, 2006, Volume: 21, Issue:6

    Transforming growth factor-beta (TGF-beta) signaling occurring during human colorectal carcinogenesis involves a shift in TGF-beta function, reducing the cytokine's antiproliferative effect, while increasing actions that promote invasion and metastasis. TGF-beta signaling involves phosphorylation of Smad3 at serine residues 208 and 213 in the linker region and serine residues 423 and 425 in the C-terminal region. Exogenous TGF-beta activates not only TGF-beta type I receptor (TbetaRI) but also c-Jun N-terminal kinase (JNK), changing unphosphorylated Smad3 to its phosphoisoforms: C-terminally phosphorylated Smad3 (pSmad3C) and linker phosphorylated Smad3 (pSmad3L). Either pSmad3C or pSmad3L oligomerizes with Smad4, and translocates into nuclei. While the TbetaRI/pSmad3C pathway inhibits growth of normal epithelial cells in vivo, JNK/pSmad3L-mediated signaling promotes tumor cell invasion and extracellular matrix synthesis by activated mesenchymal cells. Furthermore, hepatocyte growth factor signaling interacts with TGF-beta to activate the JNK/pSmad3L pathway, accelerating nuclear transport of cytoplasmic pSmad3L. This reduces accessibility of unphosphorylated Smad3 to membrane-anchored TbetaRI, preventing Smad3C phosphorylation, pSmad3C-mediated transcription, and antiproliferative effects of TGF-beta on epithelial cells. As neoplasia progresses from normal colorectal epithelium through adenoma to invasive adenocarcinoma with distant metastasis, nuclear pSmad3L gradually increases while pSmad3C decreases. The shift from TbetaRI/pSmad3C-mediated to JNK/pSmad3L-mediated signaling is a major mechanism orchestrating a complex transition of TGF-beta signaling during sporadic human colorectal carcinogenesis. This review summarizes the recent understanding of Smad3 phosphoisoform-mediated signaling, particularly 'cross-talk' between Smad3 and JNK pathways that cooperatively promote oncogenic activities. Understanding of these actions should help to develop more effective therapy against human colorectal cancer, involving inhibition of JNK/pSmad3L pathway.

    Topics: Adenocarcinoma; Adenoma; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Colorectal Neoplasms; Disease Progression; Humans; MAP Kinase Kinase 4; Neoplasm Invasiveness; Phosphorylation; Protein Isoforms; Serine; Signal Transduction; Smad3 Protein; Transforming Growth Factor beta; Tumor Cells, Cultured

2006
Role of TGFbeta in skin inflammation and carcinogenesis.
    Molecular carcinogenesis, 2006, Volume: 45, Issue:6

    The functions of transforming growth factor beta-1(TGFbeta1) are cell-context specific. We have found that TGFbeta1 expression in human skin squamous cell carcinoma (SCC) samples has two distinct distribution patterns: (1) either predominantly in suprabasal layers or (2) throughout tumor epithelia including basal proliferative cells. To understand whether the spatial TGFbeta1 expression patterns affect its functions, we have generated several keratinocyte-specific transgenic mouse models in which TGFbeta1 overexpression can be induced either predominantly in the suprabasal epidermis or in the basal layer of the epidermis and hair follicles. Suprabasal TGFbeta1 overexpression inhibits keratinocyte proliferation, suppresses skin carcinogenesis at early stages, but promotes tumor invasion at later stages. In contrast, TGFbeta1 overexpression in the basal layer of the epidermis and hair follicles causes a severe inflammatory skin disorder and epidermal hyperproliferation. Given the importance of inflammation in cancer development, our data suggest that TGFbeta1-induced skin inflammation may override its tumor suppressive effect at early stages during skin carcinogenesis. This hypothesis is further suggested by our recent study that Smad3 knockout mice are resistant to skin chemical carcinogenesis at least in part via abrogation of endogenous TGFbeta1-induced inflammation. This review intends to summarize current insights into the role of TGFbeta1 in skin inflammation and carcinogenesis.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Dermatitis; Homeostasis; Humans; Mice; Mice, Transgenic; Skin Neoplasms; Transforming Growth Factor beta

2006
Colorectal cancer: genetics of development and metastasis.
    Journal of gastroenterology, 2006, Volume: 41, Issue:3

    It has been well documented that there are two major pathways in colorectal carcinogenesis. One is the chromosomal instability pathway (adenoma-carcinoma sequence), which is characterized by allelic losses on chromosome 5q (APC), 17p (p53), and 18q (DCC/SMAD4), and the other is a pathway that involves microsatellite instability. Recent progress in molecular biology, however, has shown that colorectal carcinogenesis is not necessarily clearly divided into these two pathways, but is in fact more complicated. Other routes, including the transforming growth factor-beta/SMAD pathway, the serrated pathway, and the epigenetic pathway, have been reported. Cross talk among these pathways has also been reported. In the invasion and metastasis steps of colorectal cancers, many more genes have now been identified as being involved in proteolysis, adhesion, angiogenesis, and cell growth. Recently accumulated evidence indicates that colorectal cancer is a genetically heterogeneous and complicated disease.

    Topics: Adenocarcinoma; Carcinoma; Cell Transformation, Neoplastic; Chromosomal Instability; Colorectal Neoplasms; Epigenesis, Genetic; Gene Expression Regulation, Neoplastic; Genes, ras; Genes, Tumor Suppressor; Genetic Predisposition to Disease; Humans; Microsatellite Instability; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta; Tumor Suppressor Proteins

2006
The pleiotropic roles of transforming growth factor beta in homeostasis and carcinogenesis of endocrine organs.
    Endocrine-related cancer, 2006, Volume: 13, Issue:2

    Transforming growth factor beta (TGF-beta) is a ubiquitous cytokine that plays a critical role in numerous pathways regulating cellular and tissue homeostasis. TGF-beta is regulated by hormones and is a primary mediator of hormone response in uterus, prostate and mammary glands. This review will address the role of TGF-beta in regulating hormone-dependent proliferation and morphogenesis. The subversion of TGF-beta regulation during the processes of carcinogenesis, with particular emphasis on its effects on genetic stability and epithelial to mesenchymal transition, will also be examined. An understanding of the multiple and complex mechanisms of TGF-beta regulation of epithelial function, and the ultimate loss of TGF-beta function during carcinogenesis, will be critical in the design of novel therapeutic interventions for endocrine-related cancers.

    Topics: Animals; Cell Transformation, Neoplastic; Drug Design; Endocrine Gland Neoplasms; Homeostasis; Hormones; Humans; Mice; 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
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
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
New highlights on stroma-epithelial interactions in breast cancer.
    Breast cancer research : BCR, 2005, Volume: 7, Issue:1

    Although the stroma in which carcinomas arise has been previously regarded as a bystander to the clonal expansion and acquisition of malignant characteristics of tumor cells, it is now generally acknowledged that stromal changes are required for the establishment of cancer. In the present article, we discuss three recent publications that highlight the complex role the stroma has during the development of cancer and the potential for targeting the stroma by therapeutic approaches.

    Topics: Animals; Breast Neoplasms; Cell Communication; Cell Transformation, Neoplastic; Epithelial Cells; Female; Humans; Mammary Glands, Animal; Mammary Glands, Human; Mice; Signal Transduction; Stromal Cells; 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
The links between axin and carcinogenesis.
    Journal of clinical pathology, 2005, Volume: 58, Issue:3

    The products of the two mammalian Axin genes (Axin1 and its homologue Axin2) are essential for the degradation of beta catenin, a component of Wnt signalling that is frequently dysregulated in cancer cells. Axin is a multidomain scaffold protein that has many functions in biological signalling pathways. Overexpression of mutant [corrected] axin results in axis duplication in mouse embryos. Wnt signalling activity determines dorsal-ventral axis formation in vertebrates, implicating axin as a negative regulator of this signalling pathway. In addition, Wnts modulate pattern formation and the morphogenesis of most organs by influencing and controlling cell proliferation, motility, and fate. Defects in different components of the Wnt signalling pathway promote tumorigenesis and tumour progression. Recent biochemical studies of axins indicate that these molecules are the primary limiting components of this pathway. This review explores the intriguing connections between defects in axin function and human diseases.

    Topics: Animals; Axin Protein; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; Intercellular Signaling Peptides and Proteins; JNK Mitogen-Activated Protein Kinases; Mice; Neoplasm Proteins; Repressor Proteins; Signal Transduction; Transforming Growth Factor beta; Wnt Proteins

2005
Functions and regulation of transforming growth factor-beta (TGF-beta) in the prostate.
    European journal of cancer (Oxford, England : 1990), 2005, Volume: 41, Issue:6

    The prostate is a highly androgen-dependent tissue that in humans exhibits marked susceptibility to carcinogenesis. The malignant epithelium generated from this tissue ultimately loses dependence on androgens despite retention or amplification of the androgen receptor. Accumulating evidence support that transforming growth factor-beta (TGF-beta) plays key roles in the control of androgen dependence and acquisition of resistance to such hormonal control. Although TGF-beta functions as a key tumour suppressor of the prostate, it can also promote malignant progression and metastasis of the advanced disease, through undefined mechanisms. In addition to giving an overview of the TGF-beta field as related to its function in prostate cancer, this Review focuses on novel findings that support the tumour suppressor function of TGF-beta is lost or altered by changes in the activity of the androgen receptor, insulin-like growth factor-I, Akt, and mTOR during malignant progression. Understanding the mechanisms of cross-talk between TGF-beta and such growth modulators has important implications for the rational therapeutics of prostate cancer.

    Topics: Apoptosis; Cell Communication; Cell Cycle; Cell Transformation, Neoplastic; Humans; Male; Prostate; Prostatic Neoplasms; Receptors, Androgen; Signal Transduction; Transforming Growth Factor beta

2005
Transforming growth factor-beta and malignant melanoma: molecular mechanisms.
    Journal of cutaneous pathology, 2005, Volume: 32, Issue:6

    Transforming growth factor family members (TGF-beta) are secretory polypeptides that have dual tumor-suppressor and oncogenic effects. They signal through kinase receptor complexes on the cell surface, which phosphorylate cytoplasmic mediators (SMADs). Upon phosphorylation, SMADs march to the nucleus and interact with coactivators or corepressors to mediate the transcriptional regulation of several genes resulting in diverse effects. In tumorigenesis, malignant cells escape from the tumor-suppressive effects of TGF-beta by mutational inactivation or dysregulated expression of the molecular components in TGF-beta signaling pathway. Although melanoma cells are resistant to the tumor-suppressive effects of TGF-beta, there are no detectable defects at the receptor/SMAD level. Therefore, in these lesions, it is possible that TGF-beta effects occur independently of TGF-beta receptor/SMAD pathway. This review seeks to examine the present knowledge about TGF-beta receptor/SMAD signaling pathway and its related genes (SMADs, SKI, Filamin, endoglin, Follistatin, and other molecules) in melanomas.

    Topics: Animals; Cell Transformation, Neoplastic; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Receptor Cross-Talk; Signal Transduction; Smad Proteins; Trans-Activators; 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
Proteomics of TGF-beta signaling and its impact on breast cancer.
    Expert review of proteomics, 2005, Volume: 2, Issue:6

    The complexity of mechanisms leading to the appearance and progression of cancer is a challenge being addressed by large-scale studies, such as proteomics. Simultaneous monitoring of thousands of proteins uncovers novel signaling mechanisms, thus revising our knowledge of tumorigenesis. Transforming growth factor (TGF)-beta is a secreted polypeptide that is known to inhibit tumor growth at the early stages of cancer, but promote metastasis at the later stages. Proteomics-based studies have significantly widened our knowledge of TGF-beta-dependent regulation of cell proliferation, apoptosis, DNA damage repair and transcription. This leads to better understanding of the TGF-beta role in human breast tumorigenesis, and opens the way for the development of novel anticancer treatments and drugs, with some of the drugs already entering clinics. This review discusses recent advances in proteomics studies of TGF-beta signaling and its contribution to the understanding and treatment of breast cancer.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Humans; Protein Binding; Proteomics; Signal Transduction; Transforming Growth Factor beta

2005
Drosophila invasive tumors: a model for understanding metastasis.
    Cell cycle (Georgetown, Tex.), 2004, Volume: 3, Issue:1

    Inactivation of Drosophila tumor suppressor genes can cause excessive proliferation and, in some cases, neoplastic growth. Neoplastic growth in Drosophila tissues can also be followed by metastasis upon transplantation into hosts or in vivo. Recently, we have shown that metastatic tumors of Drosophila can provide a model in which to identify genes that are involved in the metastatic process.

    Topics: Animals; Cell Transformation, Neoplastic; Drosophila; Drosophila Proteins; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Humans; Membrane Glycoproteins; Mutation; Neoplasm Invasiveness; Neoplasm Transplantation; Neoplasms, Experimental; Semaphorins; Transforming Growth Factor beta; Tumor Suppressor Proteins

2004
What is transforming growth factor-beta (TGF-beta)?
    British journal of plastic surgery, 2004, Volume: 57, Issue:3

    The TGF-beta superfamily of proteins produces a wide range of frequently opposing effects in different cells and tissues in the body. However, its activation and mode of action are only partially understood because of its complexity in structure and functions and the variability in its downstream targets. Current work on these cytokines focuses on their receptors and the intercellular signalling pathways, comparing bioactivities between cell types and tracking their physiological and immunological effects in vivo. Future research will yield important therapeutic applications and the ability to manipulate these proteins in vivo.

    Topics: Animals; Cell Physiological Phenomena; Cell Transformation, Neoplastic; Cicatrix; DNA-Binding Proteins; Humans; Osteogenesis; Protein Isoforms; Rats; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Trans-Activators; Transforming Growth Factor beta; Wound Healing

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
FoxO: linking new signaling pathways.
    Molecular cell, 2004, May-21, Volume: 14, Issue:4

    Two recent reports reveal new roles for FoxO proteins in cell proliferation and tumorigenesis. Seoane and colleagues show that FoxO proteins play key roles in the TGFbeta-dependent activation of p21Cip1 by partnering with Smad3 and Smad4. FoxG1, a protein from a distinct Fox subfamily, binds FoxO/Smad complexes and blocks p21Cip1 expression. These interactions establish a relationship between the PI3K pathway, FoxG1, and the TGFbeta/Smad pathways. The second report identifies IkappaB kinase as a negative regulator of FoxO proteins, suggesting a mechanism for relieving negative regulation of cell cycle and promoting tumor cell proliferation.

    Topics: Animals; Apoptosis Regulatory Proteins; Carrier Proteins; Cell Division; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; DNA-Binding Proteins; Forkhead Box Protein O1; Forkhead Transcription Factors; Humans; Intracellular Signaling Peptides and Proteins; Mitochondrial Proteins; Nerve Tissue Proteins; Phosphatidylinositol 3-Kinases; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2004
Convergence of p53 and TGF-beta signaling networks.
    Cancer letters, 2004, Sep-30, Volume: 213, Issue:2

    p53 is a protein with many talents. One of the most fundamental is the ability to act as essential growth checkpoint that protects cells against cellular transformation. p53 does so through the induction of genes leading to growth arrest or apoptosis. Most of the studies focusing on the mechanisms of p53 activity have been performed in cultured cells upon treatment with well-established p53-activating inputs, such as high doses of radiations, DNA-damaging drugs and activated oncogenes. However, how the tumor suppressive functions of p53 become concerted with the extracellular cues arriving at the cell surface during tissue homeostasis, remains largely unknown. Intriguingly, two recent papers have shed new light into this unexplored field, indicating that p53 plays a key role in TGF-beta-induced growth arrest and, unexpectedly, in the developmental effects of TGF-beta in early embryos. Here we review and comment on these findings and on their implications for cancer biology.

    Topics: Cell Division; Cell Transformation, Neoplastic; DNA Damage; Embryonic and Fetal Development; Gene Expression Regulation, Developmental; Growth; Homeostasis; Humans; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53

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
The role of TGF-beta in epithelial malignancy and its relevance to the pathogenesis of oral cancer (part II).
    Critical reviews in oral biology and medicine : an official publication of the American Association of Oral Biologists, 2004, Nov-01, Volume: 15, Issue:6

    The role of transforming growth factor-beta (TGF-beta) in epithelial malignancy is complex, but it is becoming clear that, in the early stages of carcinogenesis, the protein acts as a potent tumor suppressor, while later, TGF-beta can function to advance tumor progression. We review the evidence to show that the pro-oncogenic functions of TGF-beta are associated with (1) a partial loss of response to the ligand, (2) defects of components of the TGF-beta signal transduction pathway, (3) over-expression and/or activation of the latent complex, (4) epithelial-mesenchymal transition, and (5) recruitment of signaling pathways which act in concert with TGF-beta to facilitate the metastatic phenotype. These changes are viewed in the context of what is known about the pathogenesis of oral cancer and whether this knowledge can be translated into the development of new therapeutic modalities.

    Topics: Animals; Carcinoma; Cell Transformation, Neoplastic; DNA-Binding Proteins; Epithelial Cells; Gene Expression Regulation, Neoplastic; Humans; Mouth Neoplasms; Neoplasm Metastasis; Signal Transduction; Smad Proteins; Trans-Activators; Transforming Growth Factor beta

2004
The role of transforming growth factor-beta in primary brain tumors.
    Frontiers in bioscience : a journal and virtual library, 2003, Jan-01, Volume: 8

    Dramatic therapeutic benefits of targeting specific signal transduction pathways in some cancers have pushed rational molecular targeting to the forefront of cutting-edge cancer therapy. The identification and targeting of pathways critical to the phenotype of cancers offers new hope in the treatment of many patients. Transforming growth factor beta (TGF-beta) is a multifunctional cytokine that is frequently expressed in multiple types of malignant brain tumors. TGF-beta exerts a complex set of effects in cancers with an early tumor suppressive effect through growth inhibition but later effects in cancer development that are tumorigenic - including increased tumor cell motility and invasion, induction of angiogenesis, and immune suppression. Early preclinical and clinical studies have shown promise of anti-TGF-beta strategies in the treatment of malignant gliomas suggesting TGF-beta may be a potential new therapeutic target in neuro-oncology.

    Topics: Animals; Brain Neoplasms; Cell Transformation, Neoplastic; Humans; Transforming Growth Factor beta

2003
The not-so innocent bystander: the microenvironment as a therapeutic target in cancer.
    Expert opinion on therapeutic targets, 2003, Volume: 7, Issue:1

    The microenvironment in which cancer arises is often regarded as a bystander to the clonal expansion and acquisition of malignant characteristics of the tumour. However, a major function of the microenvironment is to suppress cancer, and its disruption is required for the establishment of cancer. In addition, tumour cells can further distort the microenvironment to promote growth, recruit non-malignant cells that provide physiological resources, and facilitate invasion. In this review, the authors discuss the contribution of the microenvironment, i.e., the stroma and its resident vasculature, inflammatory cells, growth factors and the extracellular matrix (ECM), in the development of cancer, and focus on two components as potential therapeutic targets in breast cancer. First, the ECM, which imparts crucial signalling via integrins and other receptors, is a first-line barrier to invasion, modulates aggressive behaviour and may be manipulated to provide novel impediments to tumour growth. Second, the authors discuss the involvement of TGF-beta1 as an example of one of many growth factors that can regulate ECM composition and degradation and that play complex roles in cancer. Compared to the variable routes taken by cells to become cancers, the response of tissues to cancer is relatively consistent. Therefore, controlling and eliminating cancer may be more readily achieved indirectly via the tissue microenvironment.

    Topics: Breast Neoplasms; Cell Communication; Cell Transformation, Neoplastic; Cells, Cultured; Cytokines; Drug Design; Extracellular Matrix; Female; Fibronectins; Growth Substances; Humans; Laminin; Matrix Metalloproteinases; Models, Biological; Neoplasm Proteins; Proteoglycans; Stromal Cells; Tenascin; Transforming Growth Factor beta

2003
Growth regulatory factors and signalling proteins in testicular germ cell tumours.
    APMIS : acta pathologica, microbiologica, et immunologica Scandinavica, 2003, Volume: 111, Issue:1

    The molecular basis of testicular germ cell tumourigenesis are not well elucidated. Growth factors regulate cell growth, differentiation and apoptosis. Major families of growth factors are present in the male gonad from early fetal development to adult life. They are involved in germ cell proliferation and differentiation. Growth signalling pathways suffer deregulation in many human malignancies. Given the importance of growth signals in normal testicular development and their acquired deregulation in most human cancers, growth factors and signalling molecules that have been implicated in the genesis of testicular germ cell tumours, are reviewed. We detected a somatic mutation of SMAD4 gene, responsible for loss of protein function in seminomas. This mutational inactivation may affect the activity of several members of TGFbeta superfamily (TGFbeta, activin, inhibin, BMP). VEGF expression has been shown to predict metastasis in seminomas. A significant association of HST-1 expression, a member of fibroblast growth factors, with the nonseminomatous phenotype and with tumour stage has been described. In contrast, C-KIT is expressed by seminomas only, from the preinvasive stage. Despite intense expression in almost all seminomas, activating mutation of C-KIT gene is seldom reported. Recently, the first animal model of classical testicular seminoma has been identified in transgenic mouse overexpressing GDNF. RET (GDNF receptor) expression is demonstrated in human seminomas, and not in nonseminomatous tumours. However, the exact molecular alterations of GDNF/RET/GFRalpha1 complex in germ cell tumours are not known. Finally, beside growth factors, other signalling molecules such as peptide hormones may be involved in testicular carcinogenesis. We have demonstrated a specific pattern of somatostatin receptors expression in each type of testicular germ cell tumours, with a loss of sst3 and sst4 in seminomas and loss of sst4 and expression of sst1 in nonseminomas only. These data suggest an antiproliferative action of somatostatin in testicular cancers. In summary, many growth factors and signalling molecules seem to represent specific markers for different histological types of germ cell tumours (seminomas versus nonseminomas) and may play a role in the differentiation of germ cell tumours. Despite a complex signalling pathway involved in the physiological functions of male gonad, little is known about the implication of this signalling network in testicular malig

    Topics: Animals; Cell Transformation, Neoplastic; DNA-Binding Proteins; Endothelial Growth Factors; ErbB Receptors; Fibroblast Growth Factors; Germinoma; Glial Cell Line-Derived Neurotrophic Factor; Growth Substances; Humans; Male; Nerve Growth Factors; Proto-Oncogene Proteins c-kit; Receptors, Somatostatin; Signal Transduction; Smad Proteins; Stem Cell Factor; Testicular Neoplasms; Testis; Trans-Activators; Transforming Growth Factor beta

2003
Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis.
    Nature reviews. Molecular cell biology, 2003, Volume: 4, Issue:8

    In contrast to the aberrant control of proliferation, apoptosis, angiogenesis and lifespan, the cellular mechanisms that cause local invasion and metastasis of tumour cells are still poorly understood. New experimental approaches have identified different types of epithelial-plasticity changes in tumour cells towards fibroblastoid phenotypes as crucial events that occur during metastasis, and many molecules and signalling pathways cooperate to trigger these processes.

    Topics: Animals; Cell Culture Techniques; Cell Transformation, Neoplastic; Cells, Cultured; Epithelial Cells; Gene Expression Profiling; Humans; Neoplasm Metastasis; Phenotype; ras Proteins; Receptor Protein-Tyrosine Kinases; Signal Transduction; 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
Molecular changes accompanying senescence and immortalization of cultured human mammary epithelial cells.
    The international journal of biochemistry & cell biology, 2002, Volume: 34, Issue:11

    Limits on the proliferative potential of cultured normal human cells may be consequences of pathways that exist to suppress tumorigenicity. Human mammary epithelial cells (HMEC) employ several mechanisms to prevent unlimited growth. One mechanism may be activated by stress, and is associated with upregulated expression of p16(INK4a). In serum-free medium, some HMEC arise spontaneously which do not express p16. These "post-selection" HMEC are capable of long-term proliferation, but ultimately cease growth when their telomeres become very short. As they approach a growth plateau, termed agonescence, post-selection HMEC populations accumulate chromosome abnormalities. In contrast to the crisis exhibited by cells lacking functional p53, agonescent cells can be maintained as viable cultures. Although transduction of hTERT, the catalytic subunit of telomerase, into post-selection cells can, by itself, efficiently produce immortality and avoid agonescence, the errors that produce telomerase reactivation during carcinogenesis are not known. The block to endogenous telomerase reactivation in HMEC is extremely stringent. However, if one predisposing error is present, the probability greatly increases that additional error(s) required for immortalization may be generated by genomic instability encountered during agonescence. In p53(+) HMEC immortalized after chemical carcinogen exposure, the events involved in overcoming agonescence can be temporally separated from activation of telomerase. We have used the term "conversion" to describe the gradual process that leads to telomerase activation, telomere length stabilization, decreased p57 (KIP2) expression, and increased ability to grow uniformly well in the presence or absence of TGF beta. In the presence of active p53, conversion may represent a rate-limiting step in immortal transformation.

    Topics: Breast; Cell Size; Cell Transformation, Neoplastic; Cells, Cultured; Cellular Senescence; Cyclin-Dependent Kinase Inhibitor p16; Cyclin-Dependent Kinase Inhibitor p57; Epithelial Cells; Genes, myc; Humans; Nuclear Proteins; Telomerase; Trans-Activators; Transforming Growth Factor beta; Tumor Suppressor Protein p53

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
[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
Function of cytokines within the TGF-beta superfamily as determined from transgenic and gene knockout studies in mice.
    Current molecular medicine, 2002, Volume: 2, Issue:3

    Several major conceptual problems regarding specific in vivo functions of the TGF-beta family members remain the key focus of many researchers studying the biology of these secreted signaling molecules. More than 45 members of this family of growth factors have been identified and partially characterized for their molecular roles in numerous processes such as cell proliferation and differentiation, embryonic development, carcinogenesis, immune dysfunction, inflammation and wound healing. The high degree of similarity that exists at the structural level among the isoforms of these growth factors is accompanied by a significant overlap in function, as defined by many in vitro model systems and in vivo systems involving administration of exogenous ligand or of ligand-specific blocking antibodies. The ability to discern the critical functions of these molecules based on patterns of expression has also often been quite difficult. The evolution of more sophisticated functional genomics approaches has been recently instrumental in generating unique perspectives into the mechanisms governing the activity of the members of the TGF-beta family. The studies outlined in this review are significant in that they not only support working hypotheses regarding the activities of TGF-beta generated through extensive in vitro studies but also raise new questions regarding the role of each isoform in numerous processes. With the rapid advances in these approaches to probe activity in a more cell and time-dependent fashion, we will gain valuable insights for designing approaches for targeting the complex cellular pathways mediating their responses and will also help us develop novel therapies to treat disease processes.

    Topics: Animals; Cell Transformation, Neoplastic; DNA-Binding Proteins; Homeostasis; Leukocytes; Ligands; Mice; Mice, Knockout; Mice, Transgenic; Phenotype; Phylogeny; Receptors, Transforming Growth Factor beta; Smad Proteins; Trans-Activators; Transforming Growth Factor beta; Wound Healing

2002
RUNX: a trilogy of cancer genes.
    Cancer cell, 2002, Volume: 1, Issue:3

    The RUNX family of transcription factors plays pivotal roles during normal development and in neoplasias. Recent data involve RUNX3 as an important tumor suppressor in gastric cancers and pose interesting questions about how perturbed levels and interspecific competition among RUNX family members may contribute to tumorigenesis.

    Topics: Animals; Cell Transformation, Neoplastic; Core Binding Factor Alpha 2 Subunit; Core Binding Factor Alpha 3 Subunit; Core Binding Factor alpha Subunits; Disease Progression; Disease Susceptibility; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Humans; Loss of Heterozygosity; Mice; Mice, Knockout; Neoplasm Proteins; Proto-Oncogene Proteins; Stomach Neoplasms; Transcription Factors; 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
Hamartomatous polyposis syndromes: molecular genetics, neoplastic risk, and surveillance recommendations.
    Annals of surgical oncology, 2001, Volume: 8, Issue:4

    Hamartomatous polyposis syndromes are characterized by an overgrowth of cells or tissues native to the area in which they normally occur. Juvenile polyposis syndrome (JPS) results from germ-line mutations in the SMAD-4 gene (18q21.1) that encodes for an enzyme involved in transforming growth factor beta(TGF-beta) signal transduction. The increased neoplastic risk may result from SMAD-4 mutations in the stromal component, which stimulate epithelial dysplasia and progression to invasive malignancy. Peutz-Jeghers syndrome (PJS) is associated with germ-line mutations in the LKB1 gene (19p13.3) that encodes a multifunctional serine-threonine kinase. These mutations occur in the epithelial component, suggesting a direct tumor suppressor effect. Patients are at an increased risk of intestinal and extraintestinal malignancies, including breast, pancreatic, ovarian, testicular, and cervical cancer. Cowden's disease is associated with germ-line mutations in the PTEN gene (10q22-23) and an increased risk of breast and thyroid malignancies. Ruvalcaba-Myhre-Smith syndrome is less common; controversy suggests that it may represent a variant of Cowden's disease.. Genetic alterations underlying hamartomatous polyposis syndromes are diverse. Carcinogenesis may result from either germ-line mutations in the stroma (JPS) or as a direct result of functional deletion of tumor suppressor genes (PJS). Diagnosis depends on clinical presentation and patterns of inheritance within families. Suggested surveillance guidelines for the proband and first-degree relatives are outlined.

    Topics: Adenomatous Polyposis Coli; Adolescent; Adult; Age of Onset; Aged; Cell Transformation, Neoplastic; Diagnosis, Differential; Female; Genes, Tumor Suppressor; Germ-Line Mutation; Hamartoma Syndrome, Multiple; Humans; Male; Middle Aged; Neoplasm Invasiveness; Pedigree; Peutz-Jeghers Syndrome; Point Mutation; Risk Factors; Signal Transduction; Transforming Growth Factor beta

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
Tumour-stromal interactions. Role of the stroma in mammary development.
    Breast cancer research : BCR, 2001, Volume: 3, Issue:4

    Mammary development depends on branching morphogenesis, namely the bifurcation and extension of ductal growth points (end buds) and secretory lobules into a more or less fatty stroma. Because breast carcinomas are overwhelmingly ductal in origin, this review focuses on stromal influences guiding postnatal ductal development and there is only the briefest account of the role of embryonic stroma (mesenchyme). The stroma as the necessary target for endocrine mammogens and the source of stimulatory growth factors is described and the importance of mammary epithelium-induced modifications of the periductal stroma is emphasized. Evidence is presented that if they are to grow, end buds must condition proximal fatty stroma by recruiting white blood cells as well as inducing stromal cell division and, possibly, estrogen receptors. The induction of a fibrous stromal tunic around the end bud is described and its likely role as a complex ductal morphogen is discussed; a possible role in growth inhibition is also considered. Although the signals governing fibrotic induction, ductal morphogenesis, and growth inhibition are unknown, a role for transforming growth factor-beta is highly likely and is discussed. Finally, a need for new conceptual and experimental approaches to understanding stromal-epithelial signaling is discussed.

    Topics: Breast; Breast Neoplasms; Carcinoma, Ductal, Breast; Cell Communication; Cell Division; Cell Transformation, Neoplastic; Female; Humans; Leukocytes; Receptors, Estrogen; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2001
Oncogenic mechanisms of Evi-1 protein.
    Cancer chemotherapy and pharmacology, 2001, Volume: 48 Suppl 1

    Although Evi-1 is thought to promote growth or block differentiation in some cell types, its biological functions have not been elucidated. To explore the mechanisms underlying Evi-1-induced oncogenesis, we investigated whether Evi-1 affects the signaling of transforming growth factor beta (TGF-beta), which inhibits proliferation of a wide range of cell types and is one of the most studied growth regulatory factors. We demonstrated that Evi-1 represses TGF-beta signaling and antagonizes its growth-inhibitory effects. Two separate regions of Evi-1 are responsible for this repression, one of which is the first zinc-finger domain. Through this domain, Evi-1 physically interacts with Smad3, an intracellular mediator of TGF-beta signaling, thereby suppressing the transcriptional activity of Smad3. These results define a novel function of Evi-1 as a repressor of signaling components of TGF-beta. We also demonstrated that Evi-1 represses Smad-induced transcriptional activation by recruiting CtBP as a corepressor. Evi-1 associates with CtBP1 through one of the CtBP-binding consensus motifs within the region from amino acid 544 to 607, and this association is required for the efficient inhibition of TGF-beta signaling. A specific histone deacetylase (HDAc) inhibitor, trichostatin A (TSA), alleviates Evi-1-mediated repression of TGF-beta signaling, suggesting that HDAc is involved in transcriptional repression by Evi-1. This identifies a novel function of Evi-1 as a member of corepressor complexes and suggests that aberrant recruitment of corepressors is one of the mechanisms involved in Evi-1-induced leukemogenesis. These results indicate that specific HDAc inhibitors may be useful in the treatment of Evi-1-induced neoplastic tumors, including myeloid leukemias.

    Topics: Animals; Cell Transformation, Neoplastic; DNA-Binding Proteins; Humans; MDS1 and EVI1 Complex Locus Protein; Proto-Oncogenes; Signal Transduction; Smad3 Protein; Trans-Activators; Transcription Factors; Transforming Growth Factor beta; Zinc Fingers

2001
Culture models of human mammary epithelial cell transformation.
    Journal of mammary gland biology and neoplasia, 2000, Volume: 5, Issue:4

    Human pre-malignant breast diseases, particularly ductal carcinoma in situ (DCIS) already display several of the aberrant phenotypes found in primary breast cancers, including chromosomal abnormalities, telomerase activity, inactivation of the p53 gene, and overexpression of some oncogenes. Efforts to model early breast carcinogenesis in human cell cultures have largely involved studies of in vitro transformation of normal finite lifespan human mammary epithelial cells (HMEC) to immortality and malignancy. We present a model of HMEC immortal transformation consistent with the known in vivo data. This model includes a recently described, presumably epigenetic process, termed conversion, which occurs in cells that have overcome stringent replicative senescence and are thus able to maintain proliferation with critically short telomeres. The conversion process involves reactivation of telomerase activity, and acquisition of good uniform growth in the absence and presence of TGFbeta. We propose that overcoming the proliferative constraints set by senescence, and undergoing conversion, represent key rate-limiting steps in human breast carcinogenesis, and occur during early stage breast cancer progression.

    Topics: Breast Neoplasms; Cell Transformation, Neoplastic; Cells, Cultured; Cellular Senescence; Disease Models, Animal; DNA-Binding Proteins; Epithelial Cells; Humans; Mammary Glands, Human; Models, Biological; Phenotype; Telomerase; Time Factors; Transforming Growth Factor beta; Tumor Suppressor Protein p53

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
[Angiogenesis and tumor progression in melanoma].
    Recenti progressi in medicina, 2000, Volume: 91, Issue:11

    Angiogenesis is defined as the formation of new blood capillaries from preexisting vessels. It takes place in physiological and pathological conditions, such as cancer. Tumor angiogenesis depends on the release of angiogenic growth factors by tumor cells and infiltrating inflammatory cells, and from the extracellular matrix following degradation by tumor proteases. Human melanoma progresses through different steps: nevocellular nevi, dysplastic nevi, in situ melanoma, radial growth phase melanoma (Breslow index < or = 0.75 mm), vertical growth phase melanoma (Breslow index > 0.75 mm), and metastatic melanoma. In agreement with progression, it acquires a rich vascular network, whereas an increasing proportion of tumor cells express the laminin receptor, which enables their adhesion to the vascular wall. Hence, both phenomena favour tumour cell extravasation and metastases. Melanocytic cells produce and release Fibroblast Growth Factor-2 (FGF-2), mainly in the steps of dysplastic nevus and melanoma in vertical growth phase. Melanoma cells also secrete the Vascular Endothelial Growth Factor (VEGF), in parallel with the switch from the radial to the vertical growth phase and the metastatic phase. It is becoming clear that anti-angiogenic agents will interfere with or block melanoma progression.

    Topics: Cell Division; Cell Transformation, Neoplastic; Disease Progression; Endothelial Growth Factors; Fibroblast Growth Factor 2; Hepatocyte Growth Factor; Humans; Lymphokines; Melanoma; Neoplasm Invasiveness; Neoplasm Proteins; Neovascularization, Pathologic; Receptors, Laminin; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2000
[Adaptive reactions of the cell as the staring point of tumor progression].
    Voprosy onkologii, 2000, Volume: 46, Issue:5

    Topics: Animals; Cell Transformation, Neoplastic; DNA, Neoplasm; Drug Resistance, Multiple; Drug Resistance, Neoplasm; Fibroblast Growth Factors; Growth Substances; Humans; Receptors, Growth Factor; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2000
[Bright note on the molecular mechanisms of oncogenic transformation].
    Bulletin du cancer, 1999, Volume: 86, Issue:1

    Topics: Animals; Cell Cycle; Cell Hypoxia; Cell Transformation, Neoplastic; DNA Damage; Gene Expression Regulation; Genes, APC; Genes, myc; Genes, p53; Humans; Mice; Molecular Biology; Retinoblastoma Protein; Transforming Growth Factor beta

1999
Genetic events and the role of TGF beta in epithelial tumour progression.
    The Journal of pathology, 1999, Volume: 187, Issue:1

    The mouse skin model of chemical carcinogenesis has been very well characterized with respect to epigenetic changes, which occur during tumour cell initiation, promotion and progression. The use of transgenic and gene knock-out mice has contributed greatly to knowledge in this area. The H-ras genetic locus has been shown to undergo multiple genetic changes, including mutagenic activation, amplification of the mutant gene, and loss of the normal allele. These different genetic events lead to thresholds of ras activity which contribute to different stages along the pathway to neoplasia. The genetic and epigenetic events which lead to tumour invasion and metastasis have been less well characterized than studies on tumour initiation and promotion, despite the fact that it is metastases which ultimately kill the animal/patient. In the mouse skin model, loss of p53 contributes to malignant conversion. Gene deletion of the INK4 locus is associated with transformation to a highly invasive spindle cell tumor phenotype. This spindle cell transformation can also be induced in vitro or in vivo by TGF beta 1, possible by synergizing with mutant H-ras. TGF beta can have both positive and negative effects on tumourigenesis, acting early as a tumour suppresser, but later as a stimulator of tumour invasion. It is this latter effect which may be clinically more significant, since many human tumours overexpress TGF beta, yet the majority still retain the intracellular signaling systems necessary for the cell to respond to this growth factor.

    Topics: Animals; Cell Transformation, Neoplastic; Disease Progression; Epithelium; Genes, p53; Mice; Neoplasm Invasiveness; Neoplasm Metastasis; Skin Neoplasms; Transforming Growth Factor beta

1999
Going mad with Smads.
    The New England journal of medicine, 1999, Oct-07, Volume: 341, Issue:15

    Topics: Adenoma; Animals; Cell Division; Cell Transformation, Neoplastic; Colorectal Neoplasms; DNA-Binding Proteins; Gene Expression Regulation; Humans; Intestinal Polyps; Mice; Mutation; Signal Transduction; Smad3 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

1999
P15INK4b gene methylation and myelodysplastic syndromes.
    Leukemia & lymphoma, 1999, Volume: 35, Issue:5-6

    Myelodysplastic syndromes (MDS) are clonal disorders, which frequently undergo leukemic transformation. It was recently shown that the promoter of the p15INK4b but not the p16INK4a gene is frequently and selectively hypermethylated in MDS. The p15INK4b gene is a cyclin dependent kinase inhibitor gene, which is actively transcribed after TGFbeta exposure. Methylation of the p15INK4b gene is significantly correlated with blastic bone marrow involvement, and sequential analyses have shown that methylation increases with disease evolution toward AML. These data strongly suggest that p15INK4b gene methylation is a mechanism allowing leukemic cells to escape to inhibitory signals from the bone marrow environment, however the exact role of p15INK4b gene methylation in disruption of the signal mediated by TGFbeta remains to be investigated.

    Topics: Acute Disease; Animals; Antimetabolites, Antineoplastic; Azacitidine; Bone Marrow; Carrier Proteins; Cell Cycle; Cell Cycle Proteins; Cell Differentiation; Cell Transformation, Neoplastic; Chromosomes, Human, Pair 9; Clinical Trials, Phase II as Topic; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p16; Cyclin-Dependent Kinases; Decitabine; Disease Progression; DNA Methylation; Genes, p16; Genes, Tumor Suppressor; Hematopoiesis; Humans; Leukemia, Myeloid; Mice; Myelodysplastic Syndromes; Neoplasm Proteins; Precancerous Conditions; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Transforming Growth Factor beta; Tumor Suppressor Proteins

1999
The role of cytokines in both the normal and malignant ovary.
    Endocrine-related cancer, 1999, Volume: 6, Issue:1

    Normal ovarian tissue is rich in cytokines. Cytokines and chemokines are important in the physiology of ovarian function and of ovulation. Cytokines and chemokines may recruit cytokine-producing lymphocytes to the site of a developing follicle, and cytokines appear to play an important role in pre and post follicle development. Most of the same cytokines that are found in normal ovarian tissue are also found in association with malignancy in contrast to their functions in normal tissues. It is reasonable to assume that the functions of cytokines associated with malignancy may serve to promote the unregulated growth if tumor cells and metastasis. It is also likely that cytokines produced by tumors will modulate immune responses that favor tumor progression. In the following review, we have highlighted those functions of cytokines that have been identified as having the most significant impact on tumor growth and development. By examining activities of these cytokines in normal and in malignant ovarian tissues, it is hoped that future possible avenues for investigation may be opened up and that the results of these investigations will lead to strategies that can modulate the production or the activity of the cytokines leading to the growth of tumors or their metastases. Such strategies now fall under the general discipline of bioimmunotherapy. This is an expanding discipline as more is learned about growth regulation in cancer, and with the availability and rapid development of new molecules for therapeutic approaches.

    Topics: Animals; Antigen Presentation; Apoptosis; Biomarkers, Tumor; Cell Transformation, Neoplastic; Cytokines; Disease Progression; Female; Humans; Immunotherapy; Interferon-gamma; Interleukin-12; Interleukin-6; Macrophage Colony-Stimulating Factor; Mice; Neutropenia; Ovarian Neoplasms; Ovary; Ovulation; Signal Transduction; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

1999
The role of transforming growth factor beta in glioma progression.
    Journal of neuro-oncology, 1998, Volume: 36, Issue:2

    This review examines the apparently paradoxical conversion of transforming growth factor beta's (TGFbeta) regulatory role as a growth inhibitor among normal glial cells to that of a progression factor among glioblastomas (GM). In vitro, TGFbeta functions as an autocrine growth inhibitor of near-diploid gliomas of any grade. In contrast, hyperdiploid glioblastoma multiforme (HD-GM) cultures proliferate in response to TGFbeta, which is mediated by induction of platelet-derived growth factor B chain (PDGF-BB). The dominant hypothesis of TGFbeta's pathogenetic association with malignant transformation has been predicated upon acquisition of resistance to its growth inhibitory effects. However, the lack of obvious correlation with TGFbeta receptor (TbetaR) expression (or loss) between the HD-GM and the TGFbeta-inhibited GM cultures suggests the existence of intrinsically opposed regulatory mechanisms influenced by TGFbeta. The mechanism of conversion might be explained either by the loss of a putative tumor suppressor gene (TSG) which mediates TGFbeta's inhibition of growth or by enhancement of an active oncogenic pathway among the HD-GM. The frequency of mutations within glioma-associated TSG, such as TP53 and RB, suggests that defects in TGFbeta's inhibitory signaling pathway may have analogous effects in the progression to HD-GM, and TGFbeta's conversion to a mitogen. Alternative sites of inactivation which might explain the loss of TGFbeta's inhibitory effect include inactivating mutation/loss of the TbetaR type II, alterations in post-receptor signal transmission or the cyclin/cyclin dependent kinase system which regulates the phosphorylation of pRB. Loss or inactivation of a glial TSG with a consequent failure of inhibition appears to allow TGFbeta's other constitutive effects, such as induction of c-sis, to become functionally dominant. Mechanistically, TGFbeta's conversion from autocrine inhibitor to mitogen promotes 'clonal dominance' by conferring a Darwinian advantage to the hyperdiploid subpopulations through qualitative and quantitative differences in its modulation of PDGF-A and c-sis, with concomitant paracrine inhibition of competing, near-diploid elements.

    Topics: Animals; Brain Neoplasms; Cell Transformation, Neoplastic; Disease Progression; Glioma; Humans; 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 signaling].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 1997, Volume: 42, Issue:10 Suppl

    Topics: Animals; Basic Helix-Loop-Helix Leucine Zipper Transcription Factors; Calcium-Calmodulin-Dependent Protein Kinases; Cell Transformation, Neoplastic; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Humans; Repressor Proteins; Signal Transduction; Transforming Growth Factor beta

1997
TGF-beta signalling from cell membrane to nucleus through SMAD proteins.
    Nature, 1997, Dec-04, Volume: 390, Issue:6659

    The recent identification of the SMAD family of signal transducer proteins has unravelled the mechanisms by which transforming growth factor-beta (TGF-beta) signals from the cell membrane to the nucleus. Pathway-restricted SMADs are phosphorylated by specific cell-surface receptors that have serine/threonine kinase activity, then they oligomerize with the common mediator Smad4 and translocate to the nucleus where they direct transcription to effect the cell's response to TGF-beta. Inhibitory SMADs have been identified that block the activation of these pathway-restricted SMADs.

    Topics: Animals; Cell Membrane; Cell Nucleus; Cell Transformation, Neoplastic; DNA-Binding Proteins; Gene Expression Regulation; Humans; Signal Transduction; Smad1 Protein; Transforming Growth Factor beta

1997
Liver tumors and host defense.
    Seminars in liver disease, 1997, Volume: 17, Issue:4

    Complex molecular and cellular mechanisms exist to protect cells against tumor formation and to protect the entire organism against further development and spread of established tumors. The p53 tumor suppressor gene controls the cell cycle through at least two mechanisms, namely, mitotic arrest and apoptosis. Human hepatocellular carcinomas (HCCs) are often found to have mutant p53, or sometimes may have dysfunctional p53 as a result of its being bound by viral or cellular proteins. Another mechanism of host response is the production of transforming growth factor beta 1, which acts on receptors in normal hepatocytes to cause inhibition of DNA synthesis; abnormalities of transforming growth factor beta 1 have been documented in HCCs, but their biologic significance is unclear. Other host defense mechanisms include cellular responses to the tumor and the proliferation of substances with anticoagulant properties.

    Topics: Antibody Formation; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Genes, Tumor Suppressor; Humans; Immunity, Cellular; Liver Neoplasms; Transforming Growth Factor beta

1997
The cellular effects of E2F overexpression.
    Current topics in microbiology and immunology, 1996, Volume: 208

    The product of the retinoblastoma tumor-suppressor gene (RB) is a ubiquitously expressed, 105-kDa nuclear phosphoprotein (pRB). The pRB protein negatively regulates the cellular G1/S phase transition, and it is at this point in the cell cycle that it is thought to play its role as a tumor suppressor. The growth-inhibitory effects of pRB are exerted, at least in part, through the E2F family of transcription factors. This chapter reviews the insights into the mechanism of action of the E2F family members that have been obtained through overexpression studies. Studies in RB-/- SAOS-2 cells have provided evidence in support of the hypothesis that the E2F family members are negatively regulated by pRB and the related protein p130. In particular, the results obtained are consistent with the earlier biochemical data which suggested that E2F1 is regulated primarily by pRB, and E2F4 by p130. Results relating to p107 are also discussed. Consistent with the proposed role of pRB and E2F1 as coregulators of entry into S phase, experiments have demonstrated that overexpression of E2F1 is sufficient to override the cell cycle arrests caused by serum deprivation of fibroblasts or transforming growth factor-beta (TGF-beta) treatment of mink lung epithelial cells. However, at least in the case of the serum deprivation induced arrest, the ultimate result of E2F1 overexpression is death by p53-dependent apoptosis. In light of this and other data, a model is discussed as to how functional inactivation of pRB and p53 might cooperate to promote tumorigenesis. A number of studies have demonstrated the oncogenic potential of E2F family members, at least under certain conditions. This is, again, in keeping with the notion that these proteins play a critical role in controlling proliferation.

    Topics: Animals; Apoptosis; Carrier Proteins; Cell Cycle; Cell Cycle Proteins; Cell Transformation, Neoplastic; DNA-Binding Proteins; E2F Transcription Factors; E2F1 Transcription Factor; E2F4 Transcription Factor; Humans; Mutation; Retinoblastoma-Binding Protein 1; Transcription Factor DP1; Transcription Factors; Transforming Growth Factor beta; Tumor Suppressor Protein p53

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
Transforming growth factor-beta system and its regulation by members of the steroid-thyroid hormone superfamily.
    Advances in cancer research, 1996, Volume: 70

    TGF-beta s and their receptors are expressed ubiquitously, and they act as key regulators of many aspects of cell growth, differentiation, and function. Steroid action on target tissues is often associated with increase in TGF-beta isoforms. Regulation of TGF-beta expression and activation is crucial for normal development and growth control. The loss of responsiveness of different tumor cells to the antiproliferative effects of TGF-beta is a common feature in carcinogenesis. Multiple changes are required for the cells to gain complete resistance to TGF-beta growth inhibition (Fynan and Reiss, 1993; Kimchi et al., 1988; Samuel et al., 1992). Although many tumor cells are not growth inhibited by TGF-beta, they respond to TGF-beta treatment by changes in the expression of matrix components and enhanced proteolytic activity (KeskiOja et al., 1988). Agents that induce TGF-beta production in target tissues can have a chemopreventive or chemotherapeutic value for the management of epithelial malignancies. Conversely, data supporting a positive role for TGF-beta in established tumor progression are beginning to emerge (Arteaga et al., 1993a,b; Barrett-Lee et al., 1990; Arrick et al., 1992 ; E. A. Thompson et al., 1991). In later stages of tumor development, cell proliferation is often not inhibited by TGF-beta, and tumor cells secrete large amounts of this growth factor (Fynan and Reiss, 1993). In vivo TGF-beta secreted by tumor or stromal cells can influence host responses such as a natural killer cell function and thus indirctly support tumor cell viability (Arteaga et al., 1993b). TGF-beta may also affect tumor growth indirectly by stromal effects and promotion of angiogenesis. TGF-beta may also be involved in the progression of breast tumors from the steroid-sensitive to steroid-insensitive state (King et al., 1989). Understanding of the net effect of TGF-beta in different stages of tumor development is critical for the evaluation of its therapeutic value in cancer treatment.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Female; Humans; Multigene Family; Plasminogen Activators; Signal Transduction; Steroids; Thyroid Hormones; Transforming Growth Factor beta

1996
[Relevance of apoptosis for carcinogenesis].
    Verhandlungen der Deutschen Gesellschaft fur Pathologie, 1994, Volume: 78

    Apoptosis is a type of active, genetically programmed cell death. It occurs under specific conditions and is characterized by it's morphology. It is controlled by genes, hormones and other factors regulating the growth of organs and cells in the organism. In the liver and some other epithelial tissues transforming growth factor beta 1 and related peptides seem to be involved in the homeostasis of cell multiplication and cell death. In the course of carcinogenesis, initiated, preneoplastic and neoplastic cells and cell foci in the liver show enhanced DNA synthesis and also enhanced apoptosis. Tumor promoters inhibit apoptosis and increase cell replication and can thereby shift the balance between birth and death of cells accelerating tumor development. Fasting can have the opposite effect.

    Topics: Animals; Apoptosis; Carcinogens; Cell Division; Cell Transformation, Neoplastic; DNA Replication; Epithelial Cells; Epithelium; Humans; Liver; Transforming Growth Factor beta

1994
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
[Effects of cell differentiation on the induction and inhibition of cancers].
    Zhongguo yi xue ke xue yuan xue bao. Acta Academiae Medicinae Sinicae, 1994, Volume: 16, Issue:2

    Topics: Animals; Cell Transformation, Neoplastic; Humans; Transforming Growth Factor beta; Tretinoin; 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 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
Transforming growth factor regulation of keratinocyte growth.
    Recent results in cancer research. Fortschritte der Krebsforschung. Progres dans les recherches sur le cancer, 1993, Volume: 128

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Genes, myc; Humans; Keratinocytes; Retinoblastoma Protein; Transforming Growth Factor beta; Tumor Cells, Cultured

1993
Positive and negative growth control in multistage skin carcinogenesis.
    Recent results in cancer research. Fortschritte der Krebsforschung. Progres dans les recherches sur le cancer, 1993, Volume: 128

    Topics: Animals; Cell Transformation, Neoplastic; Disease Models, Animal; Genes, Tumor Suppressor; Mice; Skin Neoplasms; Transforming Growth Factor beta

1993
[Transforming growth factor beta. A potent multifunctional growth factor for normal and malignant cells].
    Ugeskrift for laeger, 1992, Nov-30, Volume: 154, Issue:49

    The polypeptide growth factor transforming growth factor-beta (TGF-beta) is a multifunctional regulator of basic cellular functions: proliferation, differentiation, cell adhesion and interactions with the extracellular matrix. TGF-beta is part of a regulatory network of which our knowledge is still incomplete, together with other substances such as steroid hormones, oncogene products and integrins. Five isoforms for TGF-beta and five different TGF-beta receptors have been described. TGF-beta exhibits an antiproliferative effect in vitro and in vivo on many cells of epthelial, myeloid, lymphoid and mesenchymal origin together with a growth-stimulating effect on various cells like endothelial cells and epidermal keratinocytes. Production of TGF-beta and receptors for TGF-beta has been found in many cell types, both normal and malignant. Nevertheless the amount of in vivo data is too limited to identify possibilities for therapeutic intervention in the physiological and patophysiological functions of TGF-beta.

    Topics: Breast Neoplasms; Cell Adhesion; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Female; Humans; Lung Neoplasms; Receptors, Cell Surface; Transforming Growth Factor beta

1992
Transforming growth factors and related peptides in gastrointestinal neoplasia.
    Journal of cellular biochemistry. Supplement, 1992, Volume: 16G

    Transforming growth factor alpha and beta 1 (TGF alpha and TGF beta 1) are representative members of two distinct and expanding families of polypeptide growth factors. TGF alpha is an epithelial cell mitogen, whereas TGF beta 1 inhibits epithelial cell growth; the role of these factors in contributing to the transformed phenotype is uncertain. Steady state mRNA expression for these growth factors and their receptors in a panel of human colon cancers and adjacent normal mucosa is presented. Based in part on results from transgenic mice in which TGF alpha is selectively overproduced in the mammary gland, a possible role for TGF alpha as a tumor promoter in the process of transformation is discussed.

    Topics: Animals; Cell Transformation, Neoplastic; Colon; Colonic Neoplasms; Female; Gastrointestinal Neoplasms; Humans; Male; Mice; Mice, Transgenic; Multigene Family; Proto-Oncogenes; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transforming Growth Factor alpha; Transforming Growth Factor beta

1992
Polypeptide modulators of prostatic growth and development.
    Cancer surveys, 1991, Volume: 11

    Normal and abnormal developmental events in the prostate are strongly influenced by androgens. There is abundant evidence, however, that androgens are not the only substances present that have the capacity to influence prostatic growth. A number of polypeptides that either stimulate or inhibit growth have now been identified in the prostate. These include members of the HBGF family, TGF-beta family, EGF and TGF-alpha, PDGF, NGF, and the less well characterized osteoblast growth factors. In some cases, the prostatic cell population, stromal or epithelial, that synthesizes the growth factor and its receptor is known. This information and the properties of the growth factors suggest ways in which these polypeptides may be involved in regulating growth of the prostate, including benign prostatic hyperplasia and prostate cancer.

    Topics: Bone Neoplasms; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblast Growth Factors; Growth Substances; Humans; Male; Neovascularization, Pathologic; Prostate; Receptors, Fibroblast Growth Factor; Transforming Growth Factor alpha; Transforming Growth Factor beta; Wound Healing

1991
Abnormalities of growth factor systems in transformed airway epithelial cells.
    Princess Takamatsu symposia, 1991, Volume: 22

    The role of the peptide growth factors transforming growth factor alpha (TGF alpha) and transforming growth factor beta (TGF beta) in the regulation of proliferation of normal and transformed airway epithelial cells was studied. Normal as well as transformed rat tracheal epithelial (RTE) cell cultures secrete similar amounts of TGF alpha during logarithmic growth. Once normal RTE cell cultures reach the plateau phase of growth, they down-regulate TGF alpha expression at the RNA and protein level; in contrast, transformed cells do not down-regulate TGF alpha. Using neutralizing TGF alpha antiserum and a tyrphostin TGF alpha/EGF receptor tyrosine kinase inhibitor, we show that the secreted TGF alpha is utilized by both normal and transformed cells as an autocrine mitogenic factor. Normal RTE cells are highly sensitive to the growth inhibitory effects of TGF beta, particularly during early phases of logarithmic growth. At late logarithmic and plateau phases of proliferation, cultures of normal RTE cells secrete large amounts of TGF beta. That the endogenous TGF beta is exerting growth inhibitory effects can be demonstrated by adding TGF beta antisera to the cultures which causes a burst of proliferation. Many transformed RTE cell lines exhibit a markedly reduced sensitivity to the growth inhibitory effects of TGF beta. However, the cells remain responsive to regulation of ECM genes by TGF beta. The transformed cell lines examined secrete less than one tenth the amount of TGF beta of normal cells. Our studies show that the autocrine TGF beta growth restraining mechanism is inoperative in many of the RTE cell transformants. We conclude, therefore, that alterations in TGF alpha and TGF beta regulation of cell proliferation are important factors contributing to the abnormal growth behavior of transformed RTE cells.

    Topics: Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Epithelium; Rats; Trachea; Transforming Growth Factor alpha; Transforming Growth Factor beta

1991
Functional loss of tumour suppressor genes in multistage chemical carcinogenesis.
    Princess Takamatsu symposia, 1991, Volume: 22

    Studies of multistage carcinogenesis in mouse skin have provided many of the early concepts of tumour initiation, promotion and progression. Genetic approaches have led to the identification of a number of mutational alterations in proto-oncogenes and tumour suppressor genes which take place at specific stages of carcinogenesis in this particular system. Initiation involves, at least in a proportion of tumours, mutational activation of the cellular H-ras proto-oncogene. Trisomy of chromosome 7, which develops during the premalignant clonal expansion phase, possibly as a consequence of tumour promoter treatment, is followed by further alterations on chromosome 7 which lead to a relative increase in the expression of mutant ras alleles. The p53 tumour suppressor gene undergoes mutational alteration and loss of heterozygosity in a proportion of squamous carcinomas but this particular gene does not appear to be involved in the further transition of squamous carcinomas to highly undifferentiated spindle cell tumours. The latter transition appears to be a recessive event which can be complemented by fusion with cells at earlier stages of malignancy. Mouse skin carcinogenesis therefore continues to provide invaluable information on the nature of the genetic and biological transitions which occur during the step-wise progression of normal cells to malignancy.

    Topics: Animals; Carcinoma; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Genes, ras; Genes, Tumor Suppressor; Humans; Mice; Mutation; Proto-Oncogene Mas; Skin Neoplasms; Transforming Growth Factor beta

1991
Positive and negative regulation of proliferation and differentiation in tracheobronchial epithelial cells.
    The American review of respiratory disease, 1990, Volume: 142, Issue:6 Pt 2

    The lung (in particular the bronchial epithelium) is a major site for tumor formation in humans. Environmental factors in conjunction with genetic factors are important determinants in this disease. The acquisition of defects in the control of proliferation and differentiation appears to constitute crucial steps in the transition of a normal to a neoplastic cell. Several factors have been identified that control positively or negatively the proliferation and differentiation of tracheobronchial epithelial cells. These factors include EGF/TGF alpha, TGF beta, insulin/IGFI, KGF, certain cytokines, retinoids, and activators of protein kinase C. Studies with neoplastic cells have identified several protooncogenes and tumor suppressor genes whose gene products are involved in the regulation of cell growth of normal tracheobronchial epithelial cells, and when mutated, lost, or activated, bring about a neoplastic phenotype. Future studies on the precise function of these genes will help to elucidate the mechanisms by which proliferation and differentiation in normal tracheobronchial epithelial cells are regulated and help to understand the molecular changes involved in diseases such as cancer.

    Topics: Bronchi; Cell Division; Cell Transformation, Neoplastic; Epithelial Cells; Genes, Tumor Suppressor; Growth Substances; Humans; Lung Neoplasms; Proto-Oncogenes; Retinoids; Trachea; Transforming Growth Factor beta

1990
Transforming growth factor-beta and suppression of carcinogenesis.
    Princess Takamatsu symposia, 1989, Volume: 20

    Transforming growth factor-beta (TGF-beta) plays an important role in controlling proliferation or differentiation in almost all epithelial tissues. The pathophysiology of TGF-beta during carcinogenesis is now an important area of investigation, since it appears that as the process of carcinogenesis progresses, epithelial cells often become refractory to the growth-regulatory actions of TGF-beta. In this article we consider the possible cellular and molecular bases for this phenomenon, and then discuss some pharmacological approaches to enhancing the synthesis or activity of TGF-beta. These approaches may provide new modalities for prevention of carcinogenesis, if they can be applied during the early stages of the disease process, before cells become refractory. We give particular attention to tamoxifen and retinoic acid, since it has been shown that these agents, which are of known efficacy for prevention of cancer, can markedly enhance the secretion of specific isotypes of TGF-beta by several types of cells.

    Topics: Amino Acid Sequence; Animals; Blood Platelets; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epithelium; Growth Inhibitors; Humans; Molecular Sequence Data; Rats; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Sequence Homology, Nucleic Acid; Species Specificity; Tamoxifen; Transforming Growth Factor beta; Tretinoin; Tumor Cells, Cultured

1989
[Tumor and transforming growth factor].
    Rinsho byori. The Japanese journal of clinical pathology, 1989, Issue:Spec No 82

    Topics: Amino Acid Sequence; Animals; Carrier Proteins; Cell Division; Cell Transformation, Neoplastic; Latent TGF-beta Binding Proteins; Molecular Sequence Data; Neoplasms, Experimental; Rats; Transforming Growth Factor beta; Transforming Growth Factors

1989
Control of human breast cancer by estrogen, growth factors, and oncogenes.
    Cancer treatment and research, 1988, Volume: 40

    Topics: Breast Neoplasms; Cell Transformation, Neoplastic; Estrogen Antagonists; Estrogens; Female; Genes, ras; Humans; Neoplasms, Hormone-Dependent; Oncogenes; Phenotype; Platelet-Derived Growth Factor; Receptors, Estrogen; Somatomedins; Transforming Growth Factor alpha; Transforming Growth Factor beta; Transforming Growth Factors; Tumor Cells, Cultured

1988

Other Studies

466 other study(ies) available for transforming-growth-factor-beta and Cell-Transformation--Neoplastic

ArticleYear
ATOH8 binds SMAD3 to induce cellular senescence and prevent Ras-driven malignant transformation.
    Proceedings of the National Academy of Sciences of the United States of America, 2023, 01-17, Volume: 120, Issue:3

    Topics: Basic Helix-Loop-Helix Transcription Factors; Cell Transformation, Neoplastic; Cellular Senescence; Genes, ras; Genes, Tumor Suppressor; Humans; Smad3 Protein; Transforming Growth Factor beta

2023
A positive TGF-β/miR-9 regulatory loop promotes the expansion and activity of tumour-initiating cells in breast cancer.
    British journal of pharmacology, 2023, Volume: 180, Issue:17

    MicroRNA-9 (miR-9) has previously been described as a dual-functional RNA during breast cancer progression and its roles need to be clarified thoroughly.. A miR-9 knockout mode of mouse breast cancer, the MMTV-PyMT model (PyMT-miR-9. MiR-9 is ectopically expressed in breast cancer and its level is negatively correlated with the prognosis, especially in basal-like breast cancer patients. Additionally, miR-9 is essential for breast cancer progression by promoting the expansion and activity of tumour-initiating cells (TICs) in preneoplastic glands, established tumours and xenograft modes. Mechanistically, the activity of TICs hinges on a positive TGF-β/miR-9 regulatory loop mediated by the STARD13/YAP axis.. These findings demonstrate that miR-9 is an oncogenic miRNA rather than a tumour-suppressor in breast cancer, calling for rectification of the model for this conserved and highly abundant miRNA.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Female; Gene Expression Regulation, Neoplastic; Humans; Mice; MicroRNAs; Neoplastic Stem Cells; Transforming Growth Factor beta

2023
Transforming Growth Factor Beta Promotes Inflammation and Tumorigenesis in Smad4-Deficient Intestinal Epithelium in a YAP-Dependent Manner.
    Advanced science (Weinheim, Baden-Wurttemberg, Germany), 2023, Volume: 10, Issue:23

    Topics: Animals; Carcinogenesis; Cell Transformation, Neoplastic; Colitis; Colitis-Associated Neoplasms; Dextran Sulfate; Inflammation; Intestinal Mucosa; Mice; Transforming Growth Factor beta; Transforming Growth Factor beta1

2023
FOXA1-induced LINC00621 promotes lung adenocarcinoma progression via activating the TGF-β signaling pathway.
    Thoracic cancer, 2023, Volume: 14, Issue:21

    Lung adenocarcinoma (LUAD) is highly malignant and associated with poor prognoses in patients worldwide. There has been widespread recognition that lncRNAs are tightly linked to LUAD tumorigenesis and development. Here, we identified that the LINC00621 level was increased in LUAD tissues and concerned with the poor prognoses in LUAD patients.. Bioinformatical analysis and RT-qPCR determined the level of LINC00621 in LUAD tissues and cell lines. The admeasurement of the proliferation, migration, and invasion abilities of LUAD cells was utilized in the CCK8 and Transwell formulas. Luciferase reporter assay was used to corroborate the downstream target genes of LINC00621. The phosphorylated SMAD3 protein was tested by Western blotting assay. The impression of LINC00621 knockdown on LUAD tumor growth and metastasis put into effect by murine models. ChIP-qPCR assay was carried out to verify the transcriptional regulation by FOXA1 on LINC00621.. In vitro, the knockdown of LINC00621 significantly reduced the proliferative, migrating, and invasive abilities, the same was true for tumorigenesis and metastasis in vivo. MiR-34a-5p as a straight target of LINC00621 was ascertained, and LUAD patients with inferior miR-34a-5p levels had undesirable prognoses. Furthermore, TGFBR1 is an immediate and functional connection site of miR-34a-5p. Collectively, LINC00621 can sponge miR-34a-5p and upregulate TGFBR1 levels, which further sensitized TGF-β signaling pathway. Finally, it was revealed that FOXA1 transcriptionally upregulated LINC00621.. This study uncovered that FOXA1-induced LINC00621 promotes LUAD progression via the miR-34a-5p/TGFBR1/TGF-β axis, and is one novel therapeutic target that may be used in LUAD treatment.

    Topics: Adenocarcinoma; Animals; Carcinogenesis; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; Lung; Lung Neoplasms; Mice; MicroRNAs; Receptor, Transforming Growth Factor-beta Type I; RNA, Long Noncoding; Signal Transduction; Transforming Growth Factor beta

2023
Heterogeneous response to TGF-β1/3 isoforms in fibroblasts of different origins: implications for wound healing and tumorigenesis.
    Histochemistry and cell biology, 2023, Volume: 160, Issue:6

    Identification of therapeutic targets for treating fibrotic diseases and cancer remains challenging. Our study aimed to investigate the effects of TGF-β1 and TGF-β3 on myofibroblast differentiation and extracellular matrix deposition in different types of fibroblasts, including normal/dermal, cancer-associated, and scar-derived fibroblasts. When comparing the phenotype and signaling pathways activation we observed extreme heterogeneity of studied markers across different fibroblast populations, even within those isolated from the same tissue. Specifically, the presence of myofibroblast and deposition of extracellular matrix were dependent on the origin of the fibroblasts and the type of treatment they received (TGF-β1 vs. TGF-β3). In parallel, we detected activation of canonical signaling (pSMAD2/3) across all studied fibroblasts, albeit to various extents. Treatment with TGF-β1 and TGF-β3 resulted in the activation of canonical and several non-canonical pathways, including AKT, ERK, and ROCK. Among studied cells, cancer-associated fibroblasts displayed the most heterogenic response to TGF-β1/3 treatments. In general, TGF-β1 demonstrated a more potent activation of signaling pathways compared to TGF-β3, whereas TGF-β3 exhibited rather an inhibitory effect in keloid- and hypertrophic scar-derived fibroblasts suggesting its clinical potential for scar treatment. In summary, our study has implications for comprehending the role of TGF-β signaling in fibroblast biology, fibrotic diseases, and cancer. Future research should focus on unraveling the mechanisms beyond differential fibroblast responses to TGF-β isomers considering inherent fibroblast heterogeneity.

    Topics: Carcinogenesis; Cell Transformation, Neoplastic; Cells, Cultured; Cicatrix, Hypertrophic; Fibroblasts; Humans; Protein Isoforms; Transforming Growth Factor beta; Transforming Growth Factor beta1; Transforming Growth Factor beta3; Wound Healing

2023
The LIN28B/TGF-β/TGFBI feedback loop promotes cell migration and tumour initiation potential in cholangiocarcinoma.
    Cancer gene therapy, 2022, Volume: 29, Issue:5

    Cholangiocarcinoma (CCA), a lethal malignancy of the biliary epithelium, is the second most common primary liver cancer. The poor prognosis of CCA is due to the high rate of tumour invasion and distant metastasis. We found that the RNA-binding protein LIN28B, a known regulator of microRNA biogenesis, stem cell maintenance, and oncogenesis, is expressed in a subpopulation of CCA patients. To further investigate the potential role of LIN28B in CCA pathogenesis, we studied the effect of LIN28B overexpression in the cholangiocyte cell line MMNK-1 and cholangiocarcinoma cell lines HuCCT-1 and KKU-214. Here, we show that enhanced LIN28B expression promoted cancer stem cell-like properties in CCA, including enhanced cell migration, epithelial-to-mesenchymal transition (EMT), increased cell proliferation and spheroid formation. Proteomic analysis revealed TGF-β-induced protein (TGFBI) as a novel LIN28B target gene, and further analysis showed upregulation of other components of the TGF-β signalling pathway, including TGF-β receptor type I (TGFBRI) expression and cytokine TGFB-I, II and III secretion. Importantly, the small molecule TGF-β inhibitor SB431542 negated the effects of LIN28B on both cell migration and clonogenic potential. Overexpression of TGFBI alone promoted cholangiocarcinoma cell migration and EMT changes, but not spheroid formation, suggesting that TGFBI partially contributes to LIN28B-mediated aggressive cell behaviour. These observations are consistent with a model in which TGF-β and LIN28B work together to form a positive feedback loop during cholangiocarcinoma metastasis and provide a therapeutic intervention opportunity.

    Topics: Bile Duct Neoplasms; Bile Ducts, Intrahepatic; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Cholangiocarcinoma; Epithelial-Mesenchymal Transition; Feedback; Humans; Proteomics; RNA-Binding Proteins; Transforming Growth Factor beta

2022
Canonical TGFβ signaling induces collective invasion in colorectal carcinogenesis through a Snail1- and Zeb1-independent partial EMT.
    Oncogene, 2022, Volume: 41, Issue:10

    Local invasion is the initial step towards metastasis, the main cause of cancer mortality. In human colorectal cancer (CRC), malignant cells predominantly invade as cohesive collectives and may undergo partial epithelial-mesenchymal transition (pEMT) at the invasive front. How this particular mode of stromal infiltration is generated is unknown. Here we investigated the impact of oncogenic transformation and the microenvironment on tumor cell invasion using genetically engineered organoids as CRC models. We found that inactivation of the Apc tumor suppressor combined with expression of oncogenic Kras

    Topics: Cell Line, Tumor; Cell Transformation, Neoplastic; Colorectal Neoplasms; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Snail Family Transcription Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Microenvironment; Zinc Finger E-box-Binding Homeobox 1

2022
Multiple transcriptome analysis of Piwil2-induced cancer stem cells, including piRNAs, mRNAs and miRNAs reveals the mechanism of tumorigenesis and development.
    Molecular biology reports, 2022, Volume: 49, Issue:7

    Cancer stem cells play important roles in the process of tumorigenesis. Our research group obtained cancer stem cell-like cells named Piwil2-iCSCs by reprogramming human preputial fibroblasts (FBs) with the PIWIL2 gene, but the mechanism of Piwil2-iCSCs is still unclear.. We sequenced the piRNAs, miRNAs and mRNAs of Piwil2-iCSCs and FBs, and analyzed the differences. Gene Ontology (GO) and, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses and gene set enrichment analysis (GSEA) were performed on the differentially expressed (DE) mRNAs. In addition, we analyzed the variable shear events and fusion genes in the Piwil2-iCSCs. Target gene prediction and functional enrichment analysis were performed for the DE miRNAs.. A total of 1119 DE mRNAs, 220 DE piRNAs, and 440 DE miRNAs were obtained between the Piwil2-iCSCs and FBs. Functional enrichment analysis showed that the genes with upregulated expression were mainly involved in DNA repair, mismatch repair, base excision repair, and nucleotide excision repair. Genes with downregulated expression were mainly involved in the TGF-β receptor signaling pathway, senescence and autophagy in cancer. More frequent shear events occurred in Piwil2-iCSCs and FBs, especially in intron retention (IR) events. We also identified three fusion genes MCM3AP-C21orf58, LRRFIP2-CAV3 and TMEM184B-DMC1. Enrichment analysis of DE miRNAs showed that they were associated with apoptosis, the TGF-β signaling pathway, and the stem cell regulatory signaling pathway. In particular, target gene prediction of the top three miRNAs with upregulated expression showed that they targeted SMAD, GREM1 and other genes to participate in the regulation of TGF-β and other pathways.. PIWIL2-induced cancer stem cells have significantly altered levels of miRNAs, piRNAs and mRNAs.TGF-β, autophagy, apoptosis and other pathways may play an important role in stem cell development. The occurrence of alternative splicing and fusion genes may be related to the occurrence of cancer stem cells.

    Topics: Acetyltransferases; Argonaute Proteins; Carcinogenesis; Cell Transformation, Neoplastic; Gene Expression Profiling; Gene Regulatory Networks; Humans; Intracellular Signaling Peptides and Proteins; MicroRNAs; Neoplastic Stem Cells; RNA, Messenger; RNA, Small Interfering; Transcriptome; Transforming Growth Factor beta

2022
Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells.
    Cell reports, 2022, 06-21, Volume: 39, Issue:12

    Regulatory T (Treg) cells play a vital role in maintaining the immunosuppressive tumor microenvironment. Lactate is a crucial metabolite in cancer and is related to tumor prognosis, metastasis, and overall survival. In this study, we focus on the effects of lactate on Treg cells. In vitro, lactate improves Treg cell stability and function, whereas lactate degradation reduces Treg cell induction, increases antitumor immunity, and decreases tumor growth in mice. Mechanistically, lactate modulates Treg cell generation through lactylation of Lys72 in MOESIN, which improves MOESIN interaction with transforming growth factor β (TGF-β) receptor I and downstream SMAD3 signaling. Cotreatment with anti-PD-1 and a lactate dehydrogenase inhibitor has a stronger antitumor effect than anti-PD-1 alone. Individuals with hepatocellular carcinoma who responded to anti-PD-1 treatment have lower levels of MOESIN lactylation in Treg cells than nonresponding individuals. Thus, we identify lactate as an essential small molecule that reinforces Treg cells in the tumor microenvironment through lactylation.

    Topics: Animals; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Lactic Acid; Liver Neoplasms; Mice; Mice, Knockout; Microfilament Proteins; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Microenvironment

2022
UPF1/circRPPH1/ATF3 feedback loop promotes the malignant phenotype and stemness of GSCs.
    Cell death & disease, 2022, 07-23, Volume: 13, Issue:7

    Glioblastoma multiforme (GBM) is the most lethal type of craniocerebral gliomas. Glioma stem cells (GSCs) are fundamental reasons for the malignancy and recurrence of GBM. Revealing the critical mechanism within GSCs' self-renewal ability is essential. Our study found a novel circular RNA (circRPPH1) that was up-regulated in GSCs and correlated with poor survival. The effect of circRPPH1 on the malignant phenotype and self-renewal of GSCs was detected in vitro and in vivo. Mechanistically, UPF1 can bind to circRPPH1 and maintain its stability. Therefore, more existing circRPPH1 can interact with transcription factor ATF3 to further transcribe UPF1 and Nestin expression. It formed a feedback loop to keep a stable stream for stemness biomarker Nestin to strengthen tumorigenesis of GSCs continually. Besides, ATF3 can activate the TGF-β signaling to drive GSCs for tumorigenesis. Knocking down the expression of circRPPH1 significantly inhibited the proliferation and clonogenicity of GSCs both in vitro and in vivo. The overexpression of circRPPH1 enhanced the self-renewal of GSCs. Our findings suggest that UPF1/circRPPH1/ATF3 maintains the potential self-renewal of GSCs through interacting with RNA-binding protein and activating the TGF-β signal pathway. Breaking the feedback loop against self-renewing GSCs may represent a novel therapeutic target in GBM treatment.

    Topics: Activating Transcription Factor 3; Brain Neoplasms; Carcinogenesis; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Feedback; Glioblastoma; Glioma; Humans; Neoplastic Stem Cells; Nestin; Phenotype; RNA Helicases; Trans-Activators; Transforming Growth Factor beta

2022
Penetration Cascade of Size Switchable Nanosystem in Desmoplastic Stroma for Improved Pancreatic Cancer Therapy.
    ACS nano, 2021, 09-28, Volume: 15, Issue:9

    Pancreatic ductal adenocarcinoma (PDAC) cells are surrounded by a dense extracellular matrix (ECM), which greatly restricts the access of therapeutic agents, resulting in poor clinical response to chemotherapy. Transforming growth factor-β1 (TGF-β1) signaling plays a crucial role in construction of the desmoplastic stroma and provides potential targets for PDAC therapy. To surmount the pathological obstacle, we developed a size switchable nanosystem based on PEG-PLGA nanospheres encapsulated within liposomes for the combined delivery of vactosertib (VAC), a TGF-β1 receptor kinase inhibitor, and the cytotoxic drug paclitaxel (TAX). By surface modification of the liposomes with a peptide, APT

    Topics: Adenocarcinoma; Carcinoma, Pancreatic Ductal; Cell Transformation, Neoplastic; Humans; Pancreatic Neoplasms; Transforming Growth Factor beta

2021
DNA Methylation of TGFβ Target Genes: Epigenetic Control of TGFβ Functional Duality in Liver Cancer.
    Cells, 2021, 08-26, Volume: 10, Issue:9

    Transforming growth factor beta (TGFβ) plays a key role in liver carcinogenesis. However, its action is complex, since TGFβ exhibits tumor-suppressive or oncogenic properties, depending on the tumor stage. At an early stage TGFβ exhibits cytostatic features, but at a later stage it promotes cell growth and metastasis, as a potent inducer of epithelial to mesenchymal transition (EMT). Here, we evaluated DNA methylation as a possible molecular mechanism switching TGFβ activity toward tumor progression in hepatocellular carcinoma (HCC). We report that decitabine, a demethylating agent already used in the clinic for the treatment of several cancers, greatly impairs the transcriptional response of SNU449 HCC cells to TGFβ. Importantly, decitabine was shown to induce the expression of EMT-related transcription factors (e.g.,

    Topics: Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Decitabine; DNA Methylation; Epigenesis, Genetic; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Liver; Liver Neoplasms; Neoplasm Recurrence, Local; Signal Transduction; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2021
Positron Emission Tomography Imaging of Functional Transforming Growth Factor β (TGFβ) Activity and Benefit of TGFβ Inhibition in Irradiated Intracranial Tumors.
    International journal of radiation oncology, biology, physics, 2021, 02-01, Volume: 109, Issue:2

    Transforming growth factor β (TGFβ) promotes cell survival by endorsing DNA damage repair and mediates an immunosuppressive tumor microenvironment. Thus, TGFβ activation in response to radiation therapy is potentially targetable because it opposes therapeutic control. Strategies to assess this potential in the clinic are needed.. We evaluated positron emission tomography (PET) to image. This study demonstrates that

    Topics: Animals; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Brain Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Female; Humans; Kaplan-Meier Estimate; Male; Mice; Positron-Emission Tomography; Transforming Growth Factor beta

2021
ZNF224 is a mediator of TGF-β pro-oncogenic function in melanoma.
    Human molecular genetics, 2021, 11-01, Volume: 30, Issue:22

    The zinc finger protein ZNF224 plays a dual role in cancer, operating as both tumour suppressor and oncogenic factor depending on cellular and molecular partners. In this research we investigated the role of ZNF224 in melanoma, a highly invasive and metastatic cancer, and provided evidence for the involvement of ZNF224 in the TGF-β signalling as a mediator of the TGF-β pro-oncogenic function. Our results showed that ZNF224, whose expression increased in melanoma cell lines after TGF-β stimulation, potentiated the activation induced by TGF-β on its target genes involved in epithelial-mesenchymal transition (EMT). Accordingly, overexpression of ZNF224 enhanced the tumourigenic properties of melanoma cells, promoting cell proliferation and invasiveness, whereas ZNF224 knockdown had the opposite effect. Moreover, ZNF224 positively modulates the expression of TGF-β itself and its type 1 and 2 receptors (TβR1 and TβR2), thus highlighting a possible mechanism by which ZNF224 could enhance the endogenous TGFβ/Smad signalling. Our findings unveil a positive regulatory loop between TGF-β and ZNF224 to promote EMT, consequently increasing the tumour metastatic potential.

    Topics: Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Disease Susceptibility; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Repressor Proteins; Signal Transduction; Transforming Growth Factor beta

2021
TGFβ Signaling Activated by Cancer-Associated Fibroblasts Determines the Histological Signature of Lung Adenocarcinoma.
    Cancer research, 2021, 09-15, Volume: 81, Issue:18

    Invasive lung adenocarcinoma (LADC) can be classified histologically as lepidic, acinar, papillary, micropapillary, or solid. Most LADC tumors manifest several of these histological subtypes, with heterogeneity being related to therapeutic resistance. We report here that in immunodeficient mice, human LADC cells form tumors with distinct histological features, MUC5AC-expressing solid-type or cytokeratin 7 (CK7)-expressing acinar-type tumors, depending on the site of development, and that a solid-to-acinar transition (SAT) could be induced by the tumor microenvironment. The TGFβ-Smad signaling pathway was activated in both tumor and stromal cells of acinar-type tumors. Immortalized cancer-associated fibroblasts (CAF) derived from acinar-type tumors induced SAT in 3D cocultures with LADC cells. Exogenous TGFβ1 or overexpression of an active form of TGFβ1 increased CK7 expression and reduced MUC5AC expression in LADC cells, and knockdown of

    Topics: Adenocarcinoma of Lung; Animals; Cancer-Associated Fibroblasts; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Female; Fluorescent Antibody Technique; Heterografts; Humans; Immunohistochemistry; Interleukin-8; Mice; Models, Biological; Neoplasm Grading; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment

2021
Neutrophil extracellular traps (NETs) formation induced by TGF-β in oral lichen planus - Possible implications for the development of oral cancer.
    Immunobiology, 2020, Volume: 225, Issue:2

    Oral Potentially Malignant Disorders (OPMDs) including Oral Lichen Planus (OLP) are associated with risk of transformation to oral squamous cell carcinoma (OSCC). Available data show that innate immune cells involving polymorphonuclear neutrophils (PMNs) with their ability to neutrophil extracellular traps (NETs) formation are likely to be directly involved in development of cancer. Examination of NETs generation by TGF-β - induced neutrophils of OLP patients showed increased amounts of traps with MPO, H3Cit and cfDNA, known to be released with NETs. The presence of excessive amounts of NETs components may lead to numerous adverse consequences associated with potential transformation to OSCC. Bacterial-related infection may enhance the NETs formation and lead to consequences resulting from the excessive number of individual elements of these networks. It is likely that regulating NETs release by the flavonoids presented herein may be beneficial not only for inhibiting OLP development, but also in reducing risk of transformation to OSCC.

    Topics: Cell Transformation, Neoplastic; Extracellular Traps; Female; Humans; Lichen Planus, Oral; Male; Middle Aged; Mouth Neoplasms; Neutrophils; Squamous Cell Carcinoma of Head and Neck; Transforming Growth Factor beta

2020
Targeting the E3 Ubiquitin Ligase PJA1 Enhances Tumor-Suppressing TGFβ Signaling.
    Cancer research, 2020, 05-01, Volume: 80, Issue:9

    RING-finger E3 ligases are instrumental in the regulation of inflammatory cascades, apoptosis, and cancer. However, their roles are relatively unknown in TGFβ/SMAD signaling. SMAD3 and its adaptors, such as β2SP, are important mediators of TGFβ signaling and regulate gene expression to suppress stem cell-like phenotypes in diverse cancers, including hepatocellular carcinoma (HCC). Here, PJA1, an E3 ligase, promoted ubiquitination and degradation of phosphorylated SMAD3 and impaired a SMAD3/β2SP-dependent tumor-suppressing pathway in multiple HCC cell lines. In mice deficient for SMAD3 (

    Topics: Animals; Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Down-Regulation; Exome Sequencing; Gene Deletion; Gene Expression Regulation; Gene Knockdown Techniques; Gene Silencing; Heterografts; Humans; Liver Neoplasms; Mice; Mice, Nude; Neoplastic Stem Cells; Oleanolic Acid; Phosphorylation; RNA, Small Interfering; Smad Proteins; Smad2 Protein; Smad3 Protein; Spectrin; Stem Cells; Transforming Growth Factor beta; Transforming Growth Factor beta1; Ubiquitin-Protein Ligases; Ubiquitination; Up-Regulation

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
RGFP966, a histone deacetylase 3 inhibitor, promotes glioma stem cell differentiation by blocking TGF-β signaling via SMAD7.
    Biochemical pharmacology, 2020, Volume: 180

    Glioma stem cells (GSC) play a major role in drug resistance and tumor recurrence. Using a genetic screen with a set of shRNAs that can target chromatin regulators in a GSC model, we have HDAC3 as a major negative regulator of GSC differentiation. Inhibition of HDAC3 using a pharmacological inhibitor or a siRNA led to the induction of GSC differentiation into astrocytes. Consequently, HDAC3-inhibition also caused a strong reduction of tumor-promoting and self-renewal capabilities of GSCs. These phenotypes were highly associated with an increased acetylation of SMAD7, which protected its ubiquitination. SMAD7 inhibits a TGF-β signaling axis that is required for maintaining stemness. These results demonstrate that HDAC3 appears to be a proper target in anti-glioma therapy.

    Topics: Acrylamides; Animals; Brain Neoplasms; Cell Differentiation; Cell Line, Tumor; Cell Survival; Cell Transformation, Neoplastic; Glioma; Histone Deacetylase Inhibitors; Histone Deacetylases; Humans; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplastic Stem Cells; Phenylenediamines; RNA, Small Interfering; Signal Transduction; Smad7 Protein; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2020
LEFTY1 Is a Dual-SMAD Inhibitor that Promotes Mammary Progenitor Growth and Tumorigenesis.
    Cell stem cell, 2020, 08-06, Volume: 27, Issue:2

    SMAD pathways govern epithelial proliferation, and transforming growth factor β (TGF-β and BMP signaling through SMAD members has distinct effects on mammary development and homeostasis. Here, we show that LEFTY1, a secreted inhibitor of NODAL/SMAD2 signaling, is produced by mammary progenitor cells and, concomitantly, suppresses SMAD2 and SMAD5 signaling to promote long-term proliferation of normal and malignant mammary epithelial cells. In contrast, BMP7, a NODAL antagonist with context-dependent functions, is produced by basal cells and restrains progenitor cell proliferation. In normal mouse epithelium, LEFTY1 expression in a subset of luminal cells and rare basal cells opposes BMP7 to promote ductal branching. LEFTY1 binds BMPR2 to suppress BMP7-induced activation of SMAD5, and this LEFTY1-BMPR2 interaction is specific to tumor-initiating cells in triple-negative breast cancer xenografts that rely on LEFTY1 for growth. These results suggest that LEFTY1 is an endogenous dual-SMAD inhibitor and that suppressing its function may represent a therapeutic vulnerability in breast cancer.

    Topics: Animals; Carcinogenesis; Cell Transformation, Neoplastic; Mice; Signal Transduction; Transforming Growth Factor beta

2020
ARNTL2 promotes pancreatic ductal adenocarcinoma progression through TGF/BETA pathway and is regulated by miR-26a-5p.
    Cell death & disease, 2020, 08-10, Volume: 11, Issue:8

    Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies and the therapeutic outcomes remain undesirable. Increasing evidence shows that aryl hydrocarbon receptor nuclear translocator like 2 (ARNTL2) plays crucial roles in tumorigenesis of multiple tumors. However, the expression status and functions of ARNTL2 in PDAC remain elusive. Here we showed that ARNTL2 expression was markedly upregulated in PDAC tissues and cell lines. elevated expression of ARNTL2 was positively related to unfavorable prognosis. Knockdown of ARNTL2 could suppress motility and invasive ability of PDAC cells in vitro, as well as tumor development in vivo. In addition, microRNA-26a-5p (miR-26a-5p) was identified as the crucial specific arbitrator for ARNTL2 expression and the expression of miR-26a-5p was inversely correlated with ARNTL2 expression in PDAC tissues. Functionally, elevated expression of miR-26a-5p was found to inhibit the proliferation, migration, and invasion of PDAC cells in vitro, while ARNTL2 increased expression could partially abolish the suppressive effect of miR-26a-5p. Mechanism study indicated that elevated expression of miR-26a-5p suppressed TGF/BETA signaling pathway by targeting ARNTL2 in PDAC cells. In conclusion, our data suggested that ARNTL2 acted as an oncogene to regulate PDAC growth. MiR-26a-5p/ARNTL2 axis may be a novel therapeutic candidate target in PDAC treatment.

    Topics: Aged; Aged, 80 and over; Apoptosis; ARNTL Transcription Factors; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Female; Gene Expression Regulation, Neoplastic; Humans; Male; MicroRNAs; Middle Aged; Pancreatic Neoplasms; Prognosis; Transforming Growth Factor beta

2020
Hypoxia-induced HIF-1α and ZEB1 are critical for the malignant transformation of ameloblastoma via TGF-β-dependent EMT.
    Cancer medicine, 2019, Volume: 8, Issue:18

    Ameloblastic carcinoma (AC) is defined as a rare primary epithelial odontogenic malignant neoplasm and the malignant counterpart of benign epithelial odontogenic tumor of ameloblastoma (AB) by the WHO classification. AC develops pulmonary metastasis in about one third of the patients and reveals a poor prognosis. However, the mechanisms of AC oncogenesis remain unclear. In this report, we aimed to clarify the mechanisms of malignant transformation of AB or AC carcinogenesis. The relatively important genes in the malignant transformation of AB were screened by DNA microarray analysis, and the expression and localization of related proteins were examined by immunohistochemistry using samples of AB and secondary AC. Two genes of hypoxia-inducible factor 1 alpha subunit (HIF1A) and zinc finger E-box-binding homeobox 1 (ZEB1) were significantly and relatively upregulated in AC than in AB. Both genes were closely related in hypoxia and epithelial-mesenchymal transition (EMT). In addition, expressions of HIF-1α and ZEB1 proteins were significantly stronger in AC than in AB. In the cell assays using ameloblastoma cell line, AM-1, hypoxia condition upregulated the expression of transforming growth factor-β (TGF-β) and induced EMT. Furthermore, the hypoxia-induced morphological change and cell migration ability were inhibited by an antiallergic medicine tranilast. Finally, we concluded that hypoxia-induced HIF-1α and ZEB1 were critical for the malignant transformation of AB via TGF-β-dependent EMT. Then, both HIF-1α and ZEB1 could be potential biomarkers to predict the malignant transformation of AB.

    Topics: Adolescent; Adult; Aged; Ameloblastoma; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Female; Gene Expression Profiling; Humans; Hypoxia; Hypoxia-Inducible Factor 1, alpha Subunit; Immunohistochemistry; Male; Middle Aged; Transforming Growth Factor beta; Young Adult; Zinc Finger E-box-Binding Homeobox 1

2019
An Epithelial-to-Mesenchymal Transcriptional Switch Triggers Evolution of Pulmonary Sarcomatoid Carcinoma (PSC) and Identifies Dasatinib as New Therapeutic Option.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2019, 04-01, Volume: 25, Issue:7

    Pulmonary sarcomatoid carcinoma (PSC) is a rare and aggressive form of NSCLC. Rarity and poor characterization have limited the development of PSC-tailored treatment protocols, leaving patients with inadequate therapeutic options. In this study, we investigated the gene expression profile of PSCs, with the aim to characterize the molecular mechanisms responsible for their evolution and to identify new drugs for their treatment.. A training set of 17 biphasic PSCs was selected and tested for the expression of a large panel of 770 genes related to cancer progression using NanoString technology. Computational analyses were used to characterize a PSCs-gene specific signature from which pathways and drivers of PSC evolution were identified and validated using functional assays in vitro. This signature was validated in a separate set of 15 PSCs and 8 differentiated NSCLC and used to interrogate the cMAP database searching for FDA-approved small molecules able to counteract PSC phenotype.. We demonstrated that the transcriptional activation of an epithelial mesenchymal transition (EMT) program drives PSC phylogeny. Our data provide new insights into PSC evolution and provide the rationale for further clinical studies with dasatinib.

    Topics: Antineoplastic Agents; Cell Line, Tumor; Cell Transformation, Neoplastic; Computational Biology; Dasatinib; Drug Substitution; Epithelial-Mesenchymal Transition; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Humans; Immunohistochemistry; Lung Neoplasms; Models, Biological; Phenotype; Protein Kinase Inhibitors; Sarcoma; Transcription, Genetic; Transforming Growth Factor beta

2019
Chronic TGF-β exposure drives stabilized EMT, tumor stemness, and cancer drug resistance with vulnerability to bitopic mTOR inhibition.
    Science signaling, 2019, 02-26, Volume: 12, Issue:570

    Tumors comprise cancer stem cells (CSCs) and their heterogeneous progeny within a stromal microenvironment. In response to transforming growth factor-β (TGF-β), epithelial and carcinoma cells undergo a partial or complete epithelial-mesenchymal transition (EMT), which contributes to cancer progression. This process is seen as reversible because cells revert to an epithelial phenotype upon TGF-β removal. However, we found that prolonged TGF-β exposure, mimicking the state of in vivo carcinomas, promotes stable EMT in mammary epithelial and carcinoma cells, in contrast to the reversible EMT induced by a shorter exposure. The stabilized EMT was accompanied by stably enhanced stem cell generation and anticancer drug resistance. Furthermore, prolonged TGF-β exposure enhanced mammalian target of rapamycin (mTOR) signaling. A bitopic mTOR inhibitor repressed CSC generation, anchorage independence, cell survival, and chemoresistance and efficiently inhibited tumorigenesis in mice. These results reveal a role for mTOR in the stabilization of stemness and drug resistance of breast cancer cells and position mTOR inhibition as a treatment strategy to target CSCs.

    Topics: Animals; Antineoplastic Agents; Benzamides; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Dioxoles; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Female; Humans; Mice, Inbred NOD; Mice, Knockout; Mice, SCID; Neoplastic Stem Cells; Signal Transduction; TOR Serine-Threonine Kinases; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2019
Fibrillin-1, a novel TGF-beta-induced factor, is preferentially expressed in metaplastic carcinoma with spindle sarcomatous metaplasia.
    Pathology, 2019, Volume: 51, Issue:4

    TGF-β induces epithelial-mesenchymal transition (EMT), which is involved in tumour progression. This study aims to identify and characterise novel factors potentially related to TGF-β-mediated tumour aggression in breast cancer. We treated the human mammary epithelial cell line MCF10A with TGF-β and observed TGF-β-dependent upregulation of FBN1, involving demethylation of CpG sites, in MCF10A cells undergoing EMT. The biological importance of fibrillin-1, encoded by FBN1, was evaluated through immunohistochemistry on 225 breast cancer specimens of various subtypes. Fibrillin-1 expression was observed only in metaplastic carcinoma of the breast (MCB) (51.7%), and the expression was observed in spindle sarcomatous metaplasia (SSM), but not in other metaplasia, including matrix-producing, pleomorphic, and squamous metaplasia, and carcinomatous components of both MCB and non-MCB. Fibrillin-1 expression was also restricted to the SSM of non-mammary carcinosarcomas of various organs. Overall, fibrillin-1 expression was enriched in MCB and non-mammary carcinosarcoma with SSM (93.7% and 93.3%, respectively), but not in MCBs and non-mammary carcinosarcoma without SSM. FBN1 knockdown in MDA-MB-231 cells with high FBN1 expression did not compromise migration, invasion, and tumourigenesis, and did not alter the expression of other EMT-related markers. In conclusion, fibrillin-1 is a novel TGF-β-induced marker. Fibrillin-1 expression in SSM, but not in other metaplasia and carcinomatous components, in both MCBs and non-mammary carcinosarcomas, together with the inability of FBN1-knockdown to compromise migration and invasion, indicates that fibrillin-1 is a marker induced solely in spindle metaplasia during EMT and does not induce EMT nor lead to tumour aggressiveness.

    Topics: Breast; Breast Neoplasms; Carcinosarcoma; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Fibrillin-1; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Humans; Immunohistochemistry; Metaplasia; Sarcoma; Transforming Growth Factor beta; Up-Regulation

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
GLI2 Modulated by SUFU and SPOP Induces Intestinal Stem Cell Niche Signals in Development and Tumorigenesis.
    Cell reports, 2019, 06-04, Volume: 27, Issue:10

    Gut mesenchyme provides key stem cell niche signals such as Wnt ligands, but how these signals are regulated is unclear. Because Hedgehog (Hh) signaling is critical for gut mesenchymal development and tumorigenesis, we investigated Hh-mediated mechanisms by analyzing mice deleted for key negative regulators of Hh signaling, Sufu and/or Spop, in the gut mesenchyme, and demonstrated their dosage-dependent roles. Although these mutants exhibit abnormal mesenchymal cell growth and functionally defective muscle layers, villification is completed with proper mesenchymal clustering, implying a permissive role for Hh signaling. These mesenchymal defects are partially rescued by Gli2 reduction. Consistent with increased epithelial proliferation caused by abnormal Hh activation in development, Sufu reduction promotes intestinal tumorigenesis, whereas Gli2 heterozygosity suppresses it. Our analyses of chromatin and GLI2 binding genomic regions reveal its transcriptional regulation of stem cell niche signals through enhancers, providing mechanistic insight into the intestinal stem cell niche in development and tumorigenesis.

    Topics: Actins; Animals; Cell Proliferation; Cell Transformation, Neoplastic; Embryo, Mammalian; Fibroblasts; Hedgehog Proteins; Intestine, Small; Mice; Mice, Knockout; Microfilament Proteins; Muscle Contraction; Muscle Proteins; Muscles; Repressor Proteins; Signal Transduction; Stem Cell Niche; Transforming Growth Factor beta; Ubiquitin-Protein Ligase Complexes; Wnt Proteins; Zinc Finger Protein Gli2

2019
Breast Cancer Stem Cells with Tumor- versus Metastasis-Initiating Capacities Are Modulated by TGFBR1 Inhibition.
    Stem cell reports, 2019, 07-09, Volume: 13, Issue:1

    Topics: Animals; Biomarkers; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Epithelial-Mesenchymal Transition; Female; Fluorescent Antibody Technique; Humans; Immunophenotyping; Mice; Neoplasm Metastasis; Neoplastic Stem Cells; Receptor, Transforming Growth Factor-beta Type I; Signal Transduction; Transforming Growth Factor beta

2019
The transcription factor LEF1 promotes tumorigenicity and activates the TGF-β signaling pathway in esophageal squamous cell carcinoma.
    Journal of experimental & clinical cancer research : CR, 2019, Jul-11, Volume: 38, Issue:1

    Esophageal squamous cell carcinoma (ESCC) is the most difficult subtype of esophageal cancer to treat due to the paucity of effective targeted therapy. ESCC is believed to arise from cancer stem cells (CSCs) that contribute to metastasis and chemoresistance. Despite advances in diagnosis and treatment, the prognosis of ESCC patients remains poor.. In this study, we applied western blot, quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry, RNA-Seq analysis, luciferase reporter assay, Chip-qPCR, bioinformatics analysis, and a series of functional assays to show the potential role of LEF1 in regulating esophageal CSCs.. We found that the overexpression of LEF1 was associated with aberrant clinicopathological characteristics and the poor prognosis of ESCC patients. In addition, the elevated expression of LEF1 and OV6 was significantly associated with aberrant clinicopathological features, and poor patient prognosis. Moreover, the overexpression of LEF1 was observed in esophageal CSCs purified by the magnetic sorting of adherent and spheroidal ESCC cells. The increased level of LEF1 in CSCs facilitated the expression of CSC markers, stem cell-like properties, resistance to chemotherapy, and tumorigenicity and increased the percentage of CSCs in ESCC samples. Conversely, the knockdown of LEF1 significantly diminished the self-renewal properties of ESCC. We showed that LEF1 played an important mechanical role in activating the TGF-β signaling pathway by directly binding to the ID1 gene promoter. A positive association between LEF1 and ID1 expression was also observed in clinical ESCC samples.. Our results indicate that the overexpression of LEF1 promotes a CSC-like phenotype in and the tumorigenicity of ESCC by activating the TGF-β signaling pathway. The inhibition of LEF1 might therefore be a novel therapeutic target to inactivate CSCs and inhibit tumor progression.

    Topics: Aged; Aged, 80 and over; Animals; Antigens, Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Esophageal Squamous Cell Carcinoma; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Lymphoid Enhancer-Binding Factor 1; Male; Mice; Middle Aged; Neoplasm Grading; Neoplasm Metastasis; Neoplasm Staging; Neoplastic Stem Cells; Prognosis; Signal Transduction; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2019
Enhanced morphological and functional differences of pancreatic cancer with epithelial or mesenchymal characteristics in 3D culture.
    Scientific reports, 2019, 07-26, Volume: 9, Issue:1

    Pancreatic cancer, composed of heterogeneous cancer cells, alters epithelial to mesenchymal features during growth and metastasis. In this study, we aimed to characterize pancreatic ductal adenocarcinoma (PDAC) cells showing epithelial or mesenchymal features in 3D culture. In 3D culture, PK-1 cells had high E-cadherin and low vimentin expression and exhibited a round-like appearance encircled by flat cells. PANC-1 cells had high vimentin and low E-cadherin expression and formed grape-like spheres. PK-1 cells had secretary granules and many microvilli, desmosomes, and adherens junctions, while PANC-1 cells had few microvilli, adherens junction, and no desmosomes. Cytokeratin 7, trypsin, CA19-9, and E-cadherin were highly expressed in PK-1 cells but not in PANC-1 cells. Ki-67 was diffusely expressed in PANC-1 spheres but was restricted to the peripheral flat cells of PK-1 spheres. PANC-1 and PK-1 cells were positive for transforming growth factor (TGF) β receptor II and phosphorylated smad2/3, but PK-1 cells were smad4 negative. Taken together, 3D culture enhanced morphofunctional differences of PDAC cells showing epithelial or mesenchymal characteristics, and epithelial phenotype maintenance may be due to the ineffectiveness of the TGF- β pathway. Clarification of heterogeneity using 3D culture may be useful for development of individualized diagnostic and therapeutic methods in patients with PDAC.

    Topics: Antigens, Tumor-Associated, Carbohydrate; Cadherins; Carcinoma, Pancreatic Ductal; Cell Culture Techniques; Cell Line, Tumor; Cell Movement; Cell Shape; Cell Transformation, Neoplastic; Desmosomes; Epithelial Cells; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Keratin-7; Pancreatic Neoplasms; Signal Transduction; Transforming Growth Factor beta; Vimentin

2019
An increased CD25-positive intestinal regulatory T lymphocyte population is dependent upon Cox-2 activity in the Apc
    Clinical and experimental immunology, 2018, Volume: 191, Issue:1

    Only mismatch repair (MMR)-deficient colorectal cancer (CRC) appears to respond well to programmed death (PD)-1 inhibition at the present time. Emerging evidence suggests a role for micro-environmental factors such as CD25

    Topics: Animals; Biomarkers; Cell Transformation, Neoplastic; Colorectal Neoplasms; Cyclooxygenase 2; Cytokines; Disease Models, Animal; Forkhead Transcription Factors; Genes, APC; Immunohistochemistry; Immunophenotyping; Interleukin-2 Receptor alpha Subunit; Intestinal Mucosa; Mice; Mice, Knockout; Mucosal-Associated Invariant T Cells; T-Lymphocytes, Regulatory; Thymic Stromal Lymphopoietin; Transforming Growth Factor beta

2018
The truncated mutant HBsAg expression increases the tumorigenesis of hepatitis B virus by regulating TGF-β/Smad signaling pathway.
    Virology journal, 2018, 04-02, Volume: 15, Issue:1

    It has been reported that the emergence of HBV rtA181T/sW172* mutant could result in a dominant secretion defect of HBsAg and increase the risk of HCC development. This study was designed to reveal the role and possible pathogenic mechanism of truncated mutant HBsAg in tumorigenesis of HBV rtA181T/sW172* mutant.. As compared to wide type or substituted mutant HBsAg, the ratio of cell clones was significant higher in L02 cells stable expressing truncated mutant HBsAg. Injection of L02 cells stable expressing truncated mutant HBsAg into the dorsal skin fold of nude mice resulted in increased primary tumor growth compared to L02 cells stable expressing wide-type and substituted mutant HBsAg. In HBV replication L02 cell lines, the key molecular involved in TGF-β/Smad pathway was also investigated. We found that the mRNA and protein levels of Smad3/2, CREB and CyclinD1 were significantly higher and TGFBI level was significantly lower in cells stably expressing truncated mutant HBsAg as compared to cells stably expressing wide-type and substituted mutant HBsAg. Additionally, after administration of TGF-β1 (increasing TGFBI level), the volume of tumor is obviously reduced in nude mice with injection of L02 cells stable expressing truncated HBsAg.. The emergence of sW172* mutant may increase the tumorigenesis of HBV, and its mechanism may be associated with down-regulated expression of TGFBI in TGF-β/Smad signaling pathway.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Disease Models, Animal; Female; Gene Expression; Hepatitis B; Hepatitis B Surface Antigens; Hepatitis B virus; Humans; Mice; Mice, Nude; Mutation; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2018
Is matrix stiffness a cause for malignant transformation of oral submucous fibrosis?
    Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology, 2018, Volume: 47, Issue:6

    Topics: Biomechanical Phenomena; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Extracellular Matrix; Fibrosis; Humans; Matrix Metalloproteinases; Mechanotransduction, Cellular; Mouth Neoplasms; Oral Submucous Fibrosis; Signal Transduction; Transforming Growth Factor beta

2018
Tumor associated macrophages support the growth of FGF9-induced lung adenocarcinoma by multiple mechanisms.
    Lung cancer (Amsterdam, Netherlands), 2018, Volume: 119

    Tumor-associated macrophages (TAMs) are known to promote tumorigenesis but the mechanism(s) remain elusive. We have developed a mouse model of lung cancer that is initiated through an inducible overexpression of fibroblast growth factor 9 (FGF9) in type-2 pneumocytes. Expression of FGF9 in adult lungs resulted in a rapid development of multiple adenocarcinoma-like tumor nodules, and is associated with an intense immunological reaction. The purpose of this study is to characterize the immune response to the FGF9-induced lung adenocarcinoma and to determine the contribution of TAMs to growth and survival of these tumors.. We used flow cytometry, immunostaining, RT-PCR and in vitro culture system on various cell populations isolated from the FGF9-induced adenocarcinoma mouse lungs.. Immunostaining demonstrated that the majority of the inflammatory cells recruited to FGF9-induced lung tumors were macrophages. These TAMs were enriched for the alternatively activated (M2) macrophage subtype. TAMs performed a significantly high immune suppressive function on T-cells and displayed high levels of arginase-1 expression and activity. The growth and colony forming potential of tumor cells was induced by co-culture with TAMs. Additionally, TAMs were shown to promote fibroblast proliferation and angiogenesis. TAMs had high expression of Tgf-β, Vegf, Fgf2, Fgf10, Fgfr2 and several matrix metalloproteinases; factors that play multiple roles in supporting tumor growth, immune protection, fibroblast activation and angiogenesis.. Our results provide evidence that the Fgf9-induced lung adenocarcinoma is associated with recruitment and activation of M2-biased TAMs, which provided multiple means of support to the tumor. This model represents an excellent means to further study the complex interactions between TAMs, their related chemokines, and progression of lung adenocarcinoma, and adds further evidence to support the importance of TAMs in tumorigenesis.

    Topics: Adenocarcinoma; Animals; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Disease Models, Animal; Disease Progression; Fibroblast Growth Factor 9; Humans; Lung Neoplasms; Macrophages; Mice; Mice, Transgenic; Transforming Growth Factor beta

2018
Transforming growth factor superfamily ligands and links to tumorigenesis.
    Leukemia & lymphoma, 2018, Volume: 59, Issue:6

    Topics: Cell Transformation, Neoplastic; Humans; Ligands; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Transforming Growth Factors

2018
Inhibition of integrin α
    Cell communication and signaling : CCS, 2018, 07-02, Volume: 16, Issue:1

    Chemotherapeutic efficacy can be improved by targeting the structure and function of the extracellular matrix (ECM) in the carcinomal stroma. This can be accomplished by e.g. inhibiting TGF-β1 and -β3 or treating with Imatinib, which results in scarcer collagen fibril structure in xenografted human KAT-4/HT29 (KAT-4) colon adenocarcinoma.. The potential role of α. Both KAT-4 and Capan-2 cells expressed the α. Our data demonstrate that the α

    Topics: Animals; Antibodies, Monoclonal; Antigens, Neoplasm; Cell Line, Tumor; Cell Transformation, Neoplastic; Collagen; Extracellular Fluid; Female; Gene Expression Profiling; Humans; Integrins; Mice; Pressure; Transforming Growth Factor beta

2018
Transforming Growth Factor-β Promotes Morphomechanical Effects Involved in Epithelial to Mesenchymal Transition in Living Hepatocellular Carcinoma.
    International journal of molecular sciences, 2018, Dec-28, Volume: 20, Issue:1

    The epithelial mesenchymal transition (EMT) is a physiological multistep process involving epithelial cells acquiring a mesenchymal-like phenotype. It is widely demonstrated that EMT is linked to tumor progression and metastasis. The transforming growth factor (TGF)-β pathways have been widely investigated, but its role in the hepatocarcinoma EMT is still unclear. While the biochemical pathways have been extensively studied, the alteration of biomechanical behavior correlated to cellular phenotype and motility is not yet fully understood. To better define the involvement of TGF-β1 in the metastatic progression process in different hepatocarcinoma cell lines (HepG2, PLC/PRF/5, HLE), we applied a systematic morphomechanical approach in order to investigate the physical and the structural characteristics. In addition, we evaluated the antitumor effect of LY2157299, a TGF-βR1 kinase inhibitor, from a biomechanical point of view, using Atomic Force and Confocal Microscopy. Our approach allows for validation of biological data, therefore it may be used in the future as a diagnostic tool to be combined with conventional biomolecular techniques.

    Topics: Biomarkers; Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Transformation, Neoplastic; Cytoskeleton; Elastic Modulus; Epithelial-Mesenchymal Transition; Humans; Liver Neoplasms; Mechanotransduction, Cellular; Microscopy, Atomic Force; Neoplasm Grading; Transforming Growth Factor beta

2018
Aberrant TGF-β Signaling Drives Castration-Resistant Prostate Cancer in a Male Mouse Model of Prostate Tumorigenesis.
    Endocrinology, 2017, 06-01, Volume: 158, Issue:6

    The androgen receptor (AR) plays a critical role as a driver of castration-resistant prostate cancer (CRPC). Our previous studies demonstrated that disruption of transforming growth factor-β (TGF-β) signaling via introduction of dominant-negative transforming growth factor-β type II receptor (DNTGFβRII) in the prostate epithelium of transgenic adenocarcinoma of the prostate mice accelerated tumor. This study investigated the consequences of disrupted TGF-β signaling on prostate tumor growth under conditions of castration-induced androgen deprivation in the preclinical model of DNTGFβRII. Our results indicate that in response to androgen deprivation therapy (ADT) the proliferative index in prostate tumors from DNTGFβRII mice was higher compared with prostate tumors from TGFβRII wild-type (WT) mice, whereas there was a reduced incidence of apoptosis in tumors from DNTGFβRII. Protein and gene expression profiling revealed that tumors from DNTGFβRII mice exhibit a strong nuclear AR localization among the prostate tumor epithelial cells and increased AR messenger RNA after ADT. In contrast, TGFβRII WT mice exhibited a marked loss in nuclear AR in prostate tumor acini (20 weeks), followed by a downregulation of AR and transmembrane protease serine 2 messenger RNA. There was a significant increase in nuclear AR and activity in prostate tumors from castrate DNTGFβRII compared with TGFβRII WT mice. Consequential to aberrant TGF-β signaling, ADT enhanced expression and nuclear localization of Smad4 and β-catenin. Our findings support that under castrate conditions, aberrant TGF-β signaling leads to AR activation and β-catenin nuclear localization, an adaptation mechanism contributing to emergence of CRPC. The work defines a potentially significant new targeting platform for overcoming therapeutic resistance in CRPC.

    Topics: Animals; beta Catenin; Cell Transformation, Neoplastic; Disease Models, Animal; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; Prostatic Neoplasms, Castration-Resistant; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Receptors, Tumor Necrosis Factor, Member 25; Signal Transduction; Transforming Growth Factor beta

2017
Loss of MMP-8 in ductal carcinoma in situ (DCIS)-associated myoepithelial cells contributes to tumour promotion through altered adhesive and proteolytic function.
    Breast cancer research : BCR, 2017, 03-23, Volume: 19, Issue:1

    Normal myoepithelial cells (MECs) play an important tumour-suppressor role in the breast but display an altered phenotype in ductal carcinoma in situ (DCIS), gaining tumour-promoter functions. Matrix metalloproteinase-8 (MMP-8) is expressed by normal MECs but is lost in DCIS. This study investigated the function of MMP-8 in MECs and the impact of its loss in DCIS.. Primary normal and DCIS-associated MECs, and normal (N-1089) and DCIS-modified myoepithelial (β6-1089) cell lines, were used to assess MMP-8 expression and function. β6-1089 lacking MMP-8 were transfected with MMP-8 WT and catalytically inactive MMP-8 EA, and MMP-8 in N-1089 MEC was knocked down with siRNA. The effect on adhesion and migration to extracellular matrix (ECM), localisation of α6β4 integrin to hemidesmosomes (HD), TGF-β signalling and gelatinase activity was measured. The effect of altering MEC MMP-8 expression on tumour cell invasion was investigated in 2D and 3D organotypic models.. Assessment of primary cells and MEC lines confirmed expression of MMP-8 in normal MEC and its loss in DCIS-MEC. Over-expression of MMP-8 WT but not MMP-8 EA in β6-1089 cells increased adhesion to ECM proteins and reduced migration. Conversely, knock-down of MMP-8 in N-1089 reduced adhesion and increased migration. Expression of MMP-8 WT in β6-1089 led to greater localisation of α6β4 to HD and reduced retraction fibre formation, this being reversed by MMP-8 knock-down in N-1089. Over-expression of MMP-8 WT reduced TGF-β signalling and gelatinolytic activity. MMP-8 knock-down enhanced TGF-β signalling and gelatinolytic activity, which was reversed by blocking MMP-9 by knock-down or an inhibitor. MMP-8 WT but not MMP-8 EA over-expression in β6-1089 reduced breast cancer cell invasion in 2D and 3D invasion assays, while MMP-8 knock-down in N-1089 enhanced cancer cell invasion. Staining of breast cancer cases for MMP-8 revealed a statistically significant loss of MMP-8 expression in DCIS with invasion versus pure DCIS (p = 0.001).. These data indicate MMP-8 is a vital component of the myoepithelial tumour-suppressor function. It restores MEC interaction with the matrix, opposes TGF-β signalling and MMP-9 proteolysis, which contributes to inhibition of tumour cell invasion. Assessment of MMP-8 expression may help to determine risk of DCIS progression.

    Topics: Biomarkers, Tumor; Carcinoma, Ductal, Breast; Carcinoma, Intraductal, Noninfiltrating; Cell Adhesion; Cell Line, Transformed; Cell Line, Tumor; Cell Movement; Cell Survival; Cell Transformation, Neoplastic; Epithelial Cells; Female; Gene Expression; Gene Knockdown Techniques; Humans; Immunohistochemistry; Integrin alpha6beta4; Matrix Metalloproteinase 8; Matrix Metalloproteinase 9; Paracrine Communication; Protein Transport; Proteolysis; Signal Transduction; Transforming Growth Factor beta

2017
TGFβ engages MEK/ERK to differentially regulate benign and malignant pancreas cell function.
    Oncogene, 2017, 07-27, Volume: 36, Issue:30

    While TGFβ signals are anti-proliferative in benign and well-differentiated pancreatic cells, TGFβ appears to promote the progression of advanced cancers. To better understand dysregulation of the TGFβ pathway, we first generated mouse models of neoplastic disease with TGFβ receptor deficiencies. These models displayed reduced levels of pERK irrespective of KRAS mutation. Furthermore, exogenous TGFβ led to rapid and sustained TGFBR1-dependent ERK phosphorylation in benign pancreatic duct cells. Similar to results that our group has published in colon cancer cells, inhibition of ERK phosphorylation in duct cells mitigated TGFβ-induced upregulation of growth suppressive pSMAD2 and p21, prevented downregulation of the pro-growth signal CDK2 and ablated TGFβ-induced EMT. These observations suggest that ERK is a key factor in growth suppressive TGFβ signals, yet may also contribute to detrimental TGFβ signaling such as EMT. In neoplastic PanIN cells, pERK was not necessary for either TGFβ-induced pSMAD2 phosphorylation or CDK2 repression, but was required for upregulation of p21 and EMT indicating a partial divergence between TGFβ and MEK/ERK in early carcinogenesis. In cancer cells, pERK had no effect on TGFβ-induced upregulation of pSMAD2 and p21, suggesting the two pathways have completely diverged with respect to the cell cycle. Furthermore, inhibition of pERK both reduced levels of CDK2 and prevented EMT independent of exogenous TGFβ, consistent with most observations identifying pERK as a tumor promoter. Combined, these data suggest that during carcinogenesis pERK initially facilitates and later antagonizes TGFβ-mediated cell cycle arrest, yet remains critical for the pathological, EMT-inducing arm of TGFβ signaling.

    Topics: Animals; Blotting, Western; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Epithelial-Mesenchymal Transition; Extracellular Signal-Regulated MAP Kinases; Female; Fluorescent Antibody Technique; Humans; Immunohistochemistry; Immunoprecipitation; Male; MAP Kinase Kinase Kinases; MAP Kinase Signaling System; Mice; Mice, Inbred C57BL; Mice, Mutant Strains; Pancreas; Pancreatic Neoplasms; Transforming Growth Factor beta

2017
The size-speed-force relationship governs migratory cell response to tumorigenic factors.
    Molecular biology of the cell, 2017, Jun-15, Volume: 28, Issue:12

    Tumor development progresses through a complex path of biomechanical changes leading first to cell growth and contraction and then cell deadhesion, scattering, and invasion. Tumorigenic factors may act specifically on one of these steps or have a wider spectrum of actions, leading to a variety of effects and thus sometimes to apparent contradictory outcomes. Here we used micropatterned lines of collagen type I/fibronectin on deformable surfaces to standardize cell behavior and measure simultaneously cell size, speed of motion and magnitude of the associated traction forces at the level of a single cell. We analyzed and compared the normal human breast cell line MCF10A in control conditions and in response to various tumorigenic factors. In all conditions, a wide range of biomechanical properties was identified. Despite this heterogeneity, normal and transformed motile cells followed a common trend whereby size and contractile forces were negatively correlated with cell speed. Some tumorigenic factors, such as activation of ErbB2 or loss of the βsubunit of casein kinase 2, shifted the whole population toward a faster speed and lower contractility state. Treatment with transforming growth factor β induced some cells to adopt opposing behaviors such as extremely high versus extremely low contractility. Thus tumor transformation amplified preexisting population heterogeneity and led some cells to exhibit biomechanical properties that were more extreme than those observed with normal cells.

    Topics: Biomechanical Phenomena; Breast Neoplasms; Carcinogenesis; Cell Adhesion; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Size; Cell Transformation, Neoplastic; Collagen Type I; Contractile Proteins; Female; Humans; Receptor, ErbB-2; Transforming Growth Factor beta

2017
Dynamics of chromatin accessibility during TGF-β-induced EMT of Ras-transformed mammary gland epithelial cells.
    Scientific reports, 2017, 04-26, Volume: 7, Issue:1

    Epithelial-mesenchymal transition (EMT) is induced by transforming growth factor (TGF)-β and facilitates tumor progression. We here performed global mapping of accessible chromatin in the mouse mammary gland epithelial EpH4 cell line and its Ras-transformed derivative (EpRas) using formaldehyde-assisted isolation of regulatory element (FAIRE)-sequencing. TGF-β and Ras altered chromatin accessibility either cooperatively or independently, and AP1, ETS, and RUNX binding motifs were enriched in the accessible chromatin regions of EpH4 and EpRas cells. Etv4, an ETS family oncogenic transcription factor, was strongly expressed and bound to more than one-third of the accessible chromatin regions in EpRas cells treated with TGF-β. While knockdown of Etv4 and another ETS family member Etv5 showed limited effects on the decrease in the E-cadherin abundance and stress fiber formation by TGF-β, gene ontology analysis showed that genes encoding extracellular proteins were most strongly down-regulated by Etv4 and Etv5 siRNAs. Accordingly, TGF-β-induced expression of Mmp13 and cell invasiveness were suppressed by Etv4 and Etv5 siRNAs, which were accompanied by the reduced chromatin accessibility at an enhancer region of Mmp13 gene. These findings suggest a mechanism of transcriptional regulation during Ras- and TGF-β-induced EMT that involves alterations of accessible chromatin, which are partly regulated by Etv4 and Etv5.

    Topics: Animals; Binding Sites; Cell Line; Cell Transformation, Neoplastic; Chromatin; DNA; Epithelial Cells; Epithelial-Mesenchymal Transition; Gene Expression Regulation; Mammary Glands, Animal; Mice; Protein Binding; Proto-Oncogene Proteins p21(ras); Transforming Growth Factor beta

2017
Integrin β1 activation induces an anti-melanoma host response.
    PloS one, 2017, Volume: 12, Issue:4

    TGF-β is a cytokine thought to function as a tumor promoter in advanced malignancies. In this setting, TGF-β increases cancer cell proliferation, survival, and migration, and orchestrates complex, pro-tumorigenic changes in the tumor microenvironment. Here, we find that in melanoma, integrin β1-mediated TGF-β activation may also produce tumor suppression via an altered host response. In the A375 human melanoma cell nu/nu xenograft model, we demonstrate that cell surface integrin β1-activation increases TGF-β activity, resulting in stromal activation, neo-angiogenesis and, unexpectedly for this nude mouse model, increase in the number of intra-tumoral CD8+ T lymphocytes within the tumor microenvironment. This is associated with attenuation of tumor growth and long-term survival benefit. Correspondingly, in human melanomas, TGF-β1 correlates with integrin β1/TGF-β1 activation and the expression of markers for vasculature and stromal activation. Surprisingly, this integrin β1/TGF-β1 transcriptional footprint also correlates with the expression of markers for tumor-infiltrating lymphocytes, multiple immune checkpoints and regulatory pathways, and, importantly, better long-term survival of patients. These correlations are unique to melanoma, in that we do not observe similar associations between β1 integrin/TGF-β1 activation and better long-term survival in other human tumor types. These results suggest that activation of TGF-β1 in melanoma may be associated with the generation of an anti-tumor host response that warrants further study.

    Topics: Animals; Antibodies; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Extracellular Space; Female; Integrin beta1; Melanoma; Mice; Mice, Nude; Neovascularization, Pathologic; Signal Transduction; Survival Analysis; T-Lymphocytes; Transforming Growth Factor beta; Tumor Microenvironment

2017
Mesenchymal stem cell infiltration during neoplastic transformation of the human prostate.
    Oncotarget, 2017, Jul-18, Volume: 8, Issue:29

    Mesenchymal Stem Cells (MSCs) have been identified in prostate cancer, raising the critical question of their physical and temporal source. Therefore, MSCs were quantified and characterized in benign and malignant prostate tissue representing different disease states and a wide range of age groups from fetal development through adult death using analytical and functional methodologies. In contrast to lineage-restricted Mesenchymal Progenitor Cells (MPCs) found in normal prostate tissue, MSCs with tri-lineage differentiation potential (adipogenesis, osteogenesis, and chondrogenesis) are identified in prostate tissue from a subset of men with prostate cancer, consistent with an influx of more stem-like progenitors (i.e. MSCs) from the bone marrow. Additionally, prostate tissue from a subset of these patients is highly enriched in MSCs, suggesting their enumeration may have prognostic value for identifying men with aggressive disease. This influx is an ongoing process continuing throughout disease progression as documented by the presence of MSCs in metastatic lesions from multiple organ sites harvested at the time of death in metastatic castration-resistant prostate cancer (mCRPC) patients. This infiltration of MSCs from systemic circulation provides the rationale for their use as a cell-based vector to deliver therapeutic agents.

    Topics: Aged; Aged, 80 and over; Atrophy; Biomarkers; Cell Transformation, Neoplastic; Cells, Cultured; Cellular Microenvironment; Humans; Immunophenotyping; Male; Mesenchymal Stem Cells; Middle Aged; Muscle, Smooth; Neoplasm Grading; Organogenesis; Paracrine Communication; Prostate; Prostatic Hyperplasia; Prostatic Neoplasms; Receptors, Androgen; Transforming Growth Factor beta

2017
TGFβ pathway limits dedifferentiation following WNT and MAPK pathway activation to suppress intestinal tumourigenesis.
    Cell death and differentiation, 2017, Volume: 24, Issue:10

    Recent studies have suggested increased plasticity of differentiated cells within the intestine to act both as intestinal stem cells (ISCs) and tumour-initiating cells. However, little is known of the processes that regulate this plasticity. Our previous work has shown that activating mutations of Kras or the NF-κB pathway can drive dedifferentiation of intestinal cells lacking Apc. To investigate this process further, we profiled both cells undergoing dedifferentiation in vitro and tumours generated from these cells in vivo by gene expression analysis. Remarkably, no clear differences were observed in the tumours; however, during dedifferentiation in vitro we found a marked upregulation of TGFβ signalling, a pathway commonly mutated in colorectal cancer (CRC). Genetic inactivation of TGFβ type 1 receptor (Tgfbr1/Alk5) enhanced the ability of Kras

    Topics: Animals; Carcinogenesis; Cell Proliferation; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Genes, ras; MAP Kinase Signaling System; Mice, Inbred C57BL; Mice, Transgenic; Mutation; NF-kappa B; Transforming Growth Factor beta; Wnt Signaling Pathway

2017
Transforming Growth Factor-β Promotes Liver Tumorigenesis in Mice via Up-regulation of Snail.
    Gastroenterology, 2017, Volume: 153, Issue:5

    Transforming growth factor beta (TGF-β) suppresses early stages of tumorigenesis, but also contributes to migration and metastasis of cancer cells. A large number of human tumors contain mutations that inactivate its receptors, or downstream proteins such as Smad transcription factors, indicating that the TGF-β signaling pathway prevents tumor growth. We investigated the effects of TGF-β inhibition on liver tumorigenesis in mice.. C57BL/6 mice received hydrodynamic tail-vein injections of transposons encoding HRAS. TGF-β inhibition via overexpression of SMAD7 (or knockdown of SMAD2, SMAD3, or SMAD4) consistently reduced formation and growth of liver tumors in mice that expressed activated RAS plus shRNA against p53, or in mice that expressed activated RAS and TAZ. TGF-β signaling activated transcription of the Snail gene in liver tumors induced by HRAS. In analyses of transgenic mice, we found TGF-β signaling to be required for formation of liver tumors upon expression of activated RAS and shRNA down-regulating p53, and upon expression of activated RAS and TAZ. Snail is the TGF-β target that is required for hepatic tumorigenesis in these models.

    Topics: Animals; Carcinoma, Hepatocellular; Cell Proliferation; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Genes, myc; Genes, ras; Genetic Predisposition to Disease; Hep G2 Cells; Humans; Liver; Liver Neoplasms, Experimental; Male; Mice, Inbred C57BL; Mice, Knockout; Phenotype; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA Interference; Signal Transduction; Smad Proteins; Snail Family Transcription Factors; Time Factors; Transfection; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Up-Regulation

2017
Overexpression of immunomodulatory mediators in oral precancerous lesions.
    Human immunology, 2017, Volume: 78, Issue:11-12

    Topics: Adult; B7-H1 Antigen; Carcinoma, Squamous Cell; Cell Proliferation; Cell Transformation, Neoplastic; Epithelial Cells; Female; Gene Expression Regulation, Neoplastic; Histocompatibility Antigens Class I; HLA-E Antigens; HLA-G Antigens; Humans; Immunologic Factors; Interleukin-10; Leukoplakia, Oral; Male; Middle Aged; Mouth Mucosa; Mouth Neoplasms; Precancerous Conditions; Transforming Growth Factor beta

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
CD51 correlates with the TGF-beta pathway and is a functional marker for colorectal cancer stem cells.
    Oncogene, 2017, Volume: 36, Issue:10

    Topics: Animals; Antineoplastic Agents; Biomarkers; Cell Line, Tumor; Cell Transformation, Neoplastic; Colorectal Neoplasms; Disease Models, Animal; Drug Resistance, Neoplasm; Gene Expression; Heterografts; Humans; Immunohistochemistry; Integrin alphaV; Mice; Neoplastic Stem Cells; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2017
Histone demethylase PHF8 promotes epithelial to mesenchymal transition and breast tumorigenesis.
    Nucleic acids research, 2017, 02-28, Volume: 45, Issue:4

    Histone demethylase PHF8 is upregulated and plays oncogenic roles in various cancers; however, the mechanisms underlying its dysregulation and functions in carcinogenesis remain obscure. Here, we report the novel functions of PHF8 in EMT (epithelial to mesenchymal transition) and breast cancer development. Genome-wide gene expression analysis revealed that PHF8 overexpression induces an EMT-like process, including the upregulation of SNAI1 and ZEB1. PHF8 demethylates H3K9me1, H3K9me2 and sustains H3K4me3 to prime the transcriptional activation of SNAI1 by TGF-β signaling. We show that PHF8 is upregulated and positively correlated with MYC at protein levels in breast cancer. MYC post-transcriptionally regulates the expression of PHF8 via the repression of microRNAs. Specifically, miR-22 directly targets and inhibits PHF8 expression, and mediates the regulation of PHF8 by MYC and TGF-β signaling. This novel MYC/microRNAs/PHF8 regulatory axis thus places PHF8 as an important downstream effector of MYC. Indeed, PHF8 contributes to MYC-induced cell proliferation and the expression of EMT-related genes. We also report that PHF8 plays important roles in breast cancer cell migration and tumor growth. These oncogenic functions of PHF8 in breast cancer confer its candidacy as a promising therapeutic target for this disease.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Disease Models, Animal; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Heterografts; Histone Demethylases; Histones; Humans; Mice; MicroRNAs; Proto-Oncogene Proteins c-myc; Signal Transduction; Transcription Factors; Transcriptional Activation; Transforming Growth Factor beta

2017
Targeting MUC1 and JNK by RNA interference and inhibitor inhibit the development of hepatocellular carcinoma.
    Cancer science, 2017, Volume: 108, Issue:3

    Mucin 1 (MUC1), as an oncogene, is overexpressed in hepatocellular carcinoma (HCC) cells and promotes the progression and tumorigenesis of HCC through JNK/TGF-β signaling pathway. In the present study, RNA interference (RNAi) and JNK inhibitor SP600125, which target MUC1 and/or JNK, were used to treat HCC cells in vitro, and the results showed that both silencing the expression of MUC1 and blocking the activity of JNK inhibited the proliferation of HCC cells. In addition, MUC1-stable-knockdown and SP600125 significantly inhibited the growth of tumors in the subcutaneous transplant tumor models that established in BALB/c nude mice rather than MUC1 or JNK siRNAs transiently transfection. Furthermore, the results from immunohistochemical staining assays showed that the inhibitory effects of MUC1 gene silencing and SP600125 on the proliferation of HCC cells in vivo were through the JNK/TGF-β signaling pathway. These results indicate that MUC1 and JNK are attractive targets for HCC therapy and may provide new therapeutic strategies for the treatment of HCC.

    Topics: Animals; Anthracenes; Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Humans; JNK Mitogen-Activated Protein Kinases; Liver Neoplasms; MAP Kinase Signaling System; Mice; Mice, Inbred BALB C; Mice, Nude; Mucin-1; RNA Interference; RNA, Small Interfering; Transforming Growth Factor beta

2017
Cancer-associated fibroblasts enact field cancerization by promoting extratumoral oxidative stress.
    Cell death & disease, 2017, 01-19, Volume: 8, Issue:1

    Histological inspection of visually normal tissue adjacent to neoplastic lesions often reveals multiple foci of cellular abnormalities. This suggests the presence of a regional carcinogenic signal that spreads oncogenic transformation and field cancerization. We observed an abundance of mutagenic reactive oxygen species in the stroma of cryosectioned patient tumor biopsies, indicative of extratumoral oxidative stress. Diffusible hydrogen peroxide (H

    Topics: Breast Neoplasms; Cancer-Associated Fibroblasts; Carcinoma, Squamous Cell; Cell Line, Tumor; Cell Transformation, Neoplastic; Female; Gene Expression Regulation, Neoplastic; Humans; Hydrogen Peroxide; Oxidative Stress; PTEN Phosphohydrolase; Reactive Oxygen Species; Signal Transduction; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2017
MEN1 and microRNAs: The link between sporadic pituitary, parathyroid and adrenocortical tumors?
    Medical hypotheses, 2017, Volume: 99

    Sporadic tumors of the pituitary, parathyroids and adrenal cortex are unique, as their benign forms are very common, but malignant forms are exceptionally rare. Hereditary forms of these tumors occur in multiple endocrine neoplasia syndrome type 1 (MEN1). We hypothesize that the pathogenic link among the sporadic tumors of these organs of different germ layers might be represented by common molecular pathways involving the MEN1 gene and microRNAs (miR). miR-24 might be a microRNA linking the three tumor entities, but other candidates such as miR-142-3p and microRNAs forming the DLK1-MEG3 miRNA cluster might also be of importance.

    Topics: Adrenal Cortex Neoplasms; Calcium-Binding Proteins; Cell Transformation, Neoplastic; Computational Biology; Gene Expression Profiling; Humans; Intercellular Signaling Peptides and Proteins; Membrane Proteins; MicroRNAs; Models, Theoretical; Multigene Family; Multiple Endocrine Neoplasia Type 1; Mutation; Parathyroid Neoplasms; Pituitary Gland; Pituitary Neoplasms; Proto-Oncogene Proteins; RNA, Long Noncoding; Smad3 Protein; 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
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
The role of mesenchymal stem cells in promoting the transformation of androgen-dependent human prostate cancer cells into androgen-independent manner.
    Scientific reports, 2016, Jan-20, Volume: 6

    Mesenchymal stem cells (MSCs) play an important role in the development of human prostate cancer (PCa). However, the role of MSCs in the transformation of androgen-dependent human PCa cells into androgen-independent manner has been poorly understood. In this study, we investigated the underlying mechanism of MSCs in promoting PCa cells from androgen-dependent into androgen-independent manner. Firstly, we demonstrated that MSCs could affect the transformation of androgen-dependent human PCa cells into androgen-independent manner in vivo and in vitro. Then we found a substantial expression of TGF-β in MSCs. TGF-β blockade could significantly inhibit the promotive function of MSCs in PCa cells. Besides that, we also demonstrated androgen might inhibit the expression of TGF-β in MSCs. Furthermore, we found that either overexpression of SSEA-4 or the number of SSEA-4 positive MSCs in PCa tissues was associated with a shorter cancer-free survival interval (CFSI) and a worse overall survival (OS). Our results suggest that androgen blockade treatment in clinical PCa therapy may elicit the expression of TGF-β in MSCs, which will result in the transformation of androgen-dependent human PCa cells into androgen-independent manner.

    Topics: Aged; Aged, 80 and over; Androgens; Animals; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Disease Models, Animal; Gene Expression Regulation, Neoplastic; Heterografts; Humans; Insulin-Like Growth Factor I; Interleukin-6; Male; Mesenchymal Stem Cells; Mice; Middle Aged; Neoplasm Grading; Neoplasm Staging; Prostatic Neoplasms; Stage-Specific Embryonic Antigens; Transforming Growth Factor beta

2016
Interferon-microRNA signalling drives liver precancerous lesion formation and hepatocarcinogenesis.
    Gut, 2016, Volume: 65, Issue:7

    Precancerous lesion, a well-established histopathologically premalignant tissue with the highest risk for tumourigenesis, develops preferentially from activation of DNA damage checkpoint and persistent inflammation. However, little is known about the mechanisms by which precancerous lesions are initiated and their physiological significance.. Laser capture microdissection was used to acquire matched normal liver, precancerous lesion and tumour tissues. miR-484(-/-), Ifnar1(-/-) and Tgfbr2(△hep) mice were employed to determine the critical role of the interferon (IFN)-microRNA pathway in precancerous lesion formation and tumourigenesis. RNA immunoprecipitation (RIP), pull-down and chromatin immunoprecipitation (ChIP) assays were applied to explore the underlying mechanisms.. miR-484 is highly expressed in over 88% liver samples clinically. DEN-induced precancerous lesions and hepatocellular carcinoma were dramatically impaired in miR-484(-/-) mice. Mechanistically, ectopic expression of miR-484 initiates tumourigenesis and cell malignant transformation through synergistic activation of the transforming growth factor-β/Gli and nuclear factor-κB/type I IFN pathways. Specific acetylation of H3K27 is indispensable for basal IFN-induced continuous transcription of miR-484 and cell transformation. Convincingly, formation of precancerous lesions were significantly attenuated in both Tgfbr2(△hep) and Ifnar1(-/-) mice.. These findings demonstrate a new protumourigenic axis involving type I IFN-microRNA signalling, providing a potential therapeutic strategy to manipulate or reverse liver precancerous lesions and tumourigenesis.

    Topics: Acetylation; Animals; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Hepatocytes; Humans; Interferon Type I; Liver; Liver Neoplasms; Mice; Mice, Knockout; MicroRNAs; NF-kappa B; NIH 3T3 Cells; Pentanones; Precancerous Conditions; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; Receptor, Interferon alpha-beta; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta; Zinc Finger Protein GLI1

2016
Overexpression of PIK3CA in murine head and neck epithelium drives tumor invasion and metastasis through PDK1 and enhanced TGFβ signaling.
    Oncogene, 2016, 09-01, Volume: 35, Issue:35

    Head and neck squamous cell carcinoma (HNSCC) patients have a poor prognosis, with invasion and metastasis as major causes of mortality. The phosphatidylinositol 3-kinase (PI3K) pathway regulates a wide range of cellular processes crucial for tumorigenesis, and PIK3CA amplification and mutation are among the most common genetic alterations in human HNSCC. Compared with the well-documented roles of the PI3K pathway in cell growth and survival, the roles of the PI3K pathway in tumor invasion and metastasis have not been well delineated. We generated a PIK3CA genetically engineered mouse model (PIK3CA-GEMM) in which wild-type PIK3CA is overexpressed in head and neck epithelium. Although PIK3CA overexpression alone was not sufficient to initiate HNSCC formation, it significantly increased tumor susceptibility in an oral carcinogenesis mouse model. PIK3CA overexpression in mouse oral epithelium increased tumor invasiveness and metastasis by increasing epithelial-mesenchymal transition and by enriching a cancer stem cell phenotype in tumor epithelial cells. In addition to these epithelial alterations, we also observed marked inflammation in tumor stroma. AKT is a central signaling mediator of the PI3K pathway. However, molecular analysis suggested that progression of PIK3CA-driven HNSCC is facilitated by 3-phosphoinositide-dependent protein kinase (PDK1) and enhanced transforming growth factor β (TGFβ) signaling rather than by AKT. Examination of human HNSCC clinical samples revealed that both PIK3CA and PDK1 protein levels correlated with tumor progression, highlighting the significance of this pathway. In summary, our results offer significant insight into how PIK3CA overexpression drives HNSCC invasion and metastasis, providing a rationale for targeting PI3K/PDK1 and TGFβ signaling in advanced HNSCC patients with PIK3CA amplification.

    Topics: Animals; Animals, Genetically Modified; Cell Proliferation; Cell Transformation, Neoplastic; Class I Phosphatidylinositol 3-Kinases; Disease Models, Animal; Epithelial-Mesenchymal Transition; Epithelium; Female; Gene Expression Regulation, Neoplastic; Head and Neck Neoplasms; Humans; Lymphatic Metastasis; Male; Mice; Mutation; Neoplasm Invasiveness; Neoplastic Stem Cells; Phosphatidylinositol 3-Kinases; Protein Serine-Threonine Kinases; Pyruvate Dehydrogenase Acetyl-Transferring Kinase; Signal Transduction; Transforming Growth Factor beta

2016
Surgical resection and radiofrequency ablation initiate cancer in cytokeratin-19+- liver cells deficient for p53 and Rb.
    Oncotarget, 2016, Aug-23, Volume: 7, Issue:34

    The long term prognosis of liver cancer patients remains unsatisfactory because of cancer recurrence after surgical interventions, particularly in patients with viral infections. Since hepatitis B and C viral proteins lead to inactivation of the tumor suppressors p53 and Retinoblastoma (Rb), we hypothesize that surgery in the context of p53/Rb inactivation initiate de novo tumorigenesis.We, therefore, generated transgenic mice with hepatocyte and cholangiocyte/liver progenitor cell (LPC)-specific deletion of p53 and Rb, by interbreeding conditional p53/Rb knockout mice with either Albumin-cre or Cytokeratin-19-cre transgenic mice.We show that liver cancer develops at the necrotic injury site after surgical resection or radiofrequency ablation in p53/Rb deficient livers. Cancer initiation occurs as a result of specific migration, expansion and transformation of cytokeratin-19+-liver (CK-19+) cells. At the injury site migrating CK-19+ cells formed small bile ducts and adjacent cells strongly expressed the transforming growth factor β (TGFβ). Isolated cytokeratin-19+ cells deficient for p53/Rb were resistant against hypoxia and TGFβ-mediated growth inhibition. CK-19+ specific deletion of p53/Rb verified that carcinomas at the injury site originates from cholangiocytes or liver progenitor cells.These findings suggest that human liver patients with hepatitis B and C viral infection or with mutations for p53 and Rb are at high risk to develop tumors at the surgical intervention site.

    Topics: Animals; Catheter Ablation; Cell Proliferation; Cell Transformation, Neoplastic; Hepatectomy; Hepatocytes; Humans; Keratin-19; Liver; Liver Neoplasms; Mice; Mice, Knockout; Mice, Transgenic; Retinoblastoma Protein; Tissue Culture Techniques; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2016
Altered TGF-α/β signaling drives cooperation between breast cancer cell populations.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2016, Volume: 30, Issue:10

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Epithelium; Humans; Mice, SCID; Signal Transduction; Transforming Growth Factor alpha; Transforming Growth Factor beta

2016
ZEB1 drives epithelial-to-mesenchymal transition in lung cancer.
    The Journal of clinical investigation, 2016, 09-01, Volume: 126, Issue:9

    Increased expression of zinc finger E-box binding homeobox 1 (ZEB1) is associated with tumor grade and metastasis in lung cancer, likely due to its role as a transcription factor in epithelial-to-mesenchymal transition (EMT). Here, we modeled malignant transformation in human bronchial epithelial cells (HBECs) and determined that EMT and ZEB1 expression are early, critical events in lung cancer pathogenesis. Specific oncogenic mutations in TP53 and KRAS were required for HBECs to engage EMT machinery in response to microenvironmental (serum/TGF-β) or oncogenetic (MYC) factors. Both TGF-β- and MYC-induced EMT required ZEB1, but engaged distinct TGF-β-dependent and vitamin D receptor-dependent (VDR-dependent) pathways, respectively. Functionally, we found that ZEB1 causally promotes malignant progression of HBECs and tumorigenicity, invasion, and metastases in non-small cell lung cancer (NSCLC) lines. Mechanistically, ZEB1 expression in HBECs directly repressed epithelial splicing regulatory protein 1 (ESRP1), leading to increased expression of a mesenchymal splice variant of CD44 and a more invasive phenotype. In addition, ZEB1 expression in early stage IB primary NSCLC correlated with tumor-node-metastasis stage. These findings indicate that ZEB1-induced EMT and associated molecular changes in ESRP1 and CD44 contribute to early pathogenesis and metastatic potential in established lung cancer. Moreover, TGF-β and VDR signaling and CD44 splicing pathways associated with ZEB1 are potential EMT chemoprevention and therapeutic targets in NSCLC.

    Topics: Animals; Carcinoma, Non-Small-Cell Lung; Carcinoma, Small Cell; Cell Line; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Female; Humans; Hyaluronan Receptors; Lung Neoplasms; Mice; Mice, Inbred NOD; Mice, SCID; Microcirculation; Neoplasm Invasiveness; Neoplasm Metastasis; Phenotype; Proto-Oncogene Proteins c-myc; Receptors, Calcitriol; RNA-Binding Proteins; Transforming Growth Factor beta; Zinc Finger E-box-Binding Homeobox 1

2016
c-Abl inhibits breast cancer tumorigenesis through reactivation of p53-mediated p21 expression.
    Oncotarget, 2016, Nov-08, Volume: 7, Issue:45

    We previously reported that constitutive c-Abl activity (CST-Abl) abrogates the tumorigenicity of triple-negative breast cancer cells through the combined actions of two cellular events: downregulated matrix metalloproteinase (MMP) and upregulated p21Waf1/Cip1 expression. We now find decreased c-Abl expression to be significantly associated with diminished relapse-fee survival in breast cancer patients, particularly those exhibiting invasive and basal phenotypes. Moreover, CST-Abl expression enabled 4T1 cells to persist innocuously in the mammary glands of mice, doing so by exhausting their supply of cancer stem cells. Restoring MMP-9 expression and activity in CST-Abl-expressing 4T1 cells failed to rescue their malignant phenotypes; however, rendering these same cells deficient in p21 expression not only delayed their acquisition of senescent phenotypes, but also partially restored their tumorigenicity in mice. Although 4T1 cells lacked detectable expression of p53, those engineered to express CST-Abl exhibited robust production and secretion of TGF-β1 that engendered the reactivated expression of p53. Mechanistically, TGF-β-mediated p53 expression transpired through the combined actions of Smad1/5/8 and Smad2, leading to the dramatic upregulation of p21 and its stimulation of TNBC senescence. Collectively, we identified a novel c-Abl:p53:p21 signaling axis that functions as a powerful suppressor of mammary tumorigenesis and metastatic progression.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; Disease Models, Animal; Disease Progression; Female; Gene Expression Regulation, Neoplastic; Humans; Mammary Neoplasms, Animal; Matrix Metalloproteinase 9; Mice; Models, Biological; Proto-Oncogene Proteins c-abl; Smad Proteins, Receptor-Regulated; Transcriptional Activation; Transforming Growth Factor beta; Triple Negative Breast Neoplasms; Tumor Suppressor Protein p53

2016
Infantile Hemangioma Originates From A Dysregulated But Not Fully Transformed Multipotent Stem Cell.
    Scientific reports, 2016, 10-27, Volume: 6

    Infantile hemangioma (IH) is the most common tumor of infancy. Its cellular origin and biological signals for uncontrolled growth are poorly understood, and specific pharmacological treatment is unavailable. To understand the process of hemangioma-genesis we characterized the progenitor hemangioma-derived stem cell (HemSC) and its lineage and non-lineage derivatives. For this purpose we performed a high-throughput (HT) phenotypic and gene expression analysis of HemSCs, and analyzed HemSC-derived tumorspheres. We found that IH is characterized by high expression of genes involved in vasculogenesis, angiogenesis, tumorigenesis and associated signaling pathways. These results show that IH derives from a dysregulated stem cell that remains in an immature, arrested stage of development. The potential biomarkers we identified can afford the development of diagnostic tools and precision-medicine therapies to "rewire" or redirect cellular transitions at an early stage, such as signaling pathways or immune response modifiers.

    Topics: Biomarkers, Tumor; Cell Differentiation; Cell Proliferation; Cell Transformation, Neoplastic; Endoglin; Gene Expression Regulation, Neoplastic; Hemangioma; Human Umbilical Vein Endothelial Cells; Humans; Infant; Multipotent Stem Cells; Neoplastic Stem Cells; Neovascularization, Pathologic; Transforming Growth Factor beta; Tumor Cells, Cultured

2016
Decreased TIP30 promotes Snail-mediated epithelial-mesenchymal transition and tumor-initiating properties in hepatocellular carcinoma.
    Oncogene, 2015, Mar-12, Volume: 34, Issue:11

    The poor prognosis of hepatocellular carcinoma (HCC) is mainly due to tumor recurrence and metastases. Recently, epithelial-mesenchymal transition (EMT) has been implicated in tumor invasion and metastasis. However, the underlying molecular mechanisms are yet to be elucidated. Here, we show that 30-kDa Tat-interacting protein (TIP30), also called CC3, is significantly downregulated during transforming growth factor-β-induced EMT. In our in vitro and in vivo studies, we show that decreased TIP30 expression leads to EMT, as well as enhanced motility and invasion of HCC cells. Also, increased self-renewal ability and chemotherapeutic resistance are observed with TIP30 depletion. Moreover, Snail is one of the key transcription factors promoting EMT, and overexpression of TIP30 greatly decreased nucleic accumulation in Snail through the regulation of intracellular localization. Small interfering RNAs targeting Snail attenuated EMT and tumor-initiating properties induced by TIP30 deficiency. We further confirmed that TIP30 competitively interrupted the interaction of Snail with importin-β2 to block the nuclear import of Snail. Consistently, TIP30 expression significantly correlates with E-cadherin expression in HCC patients. TIP30 or combination of E-cadherin is a powerful marker in predicting the prognosis of HCC. Taken together, our results suggest a novel and critical role of TIP30 involved in HCC progression and aggressiveness.

    Topics: Acetyltransferases; Animals; beta Karyopherins; Cadherins; Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Down-Regulation; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Hep G2 Cells; Humans; Liver Neoplasms; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Middle Aged; Neoplasm Invasiveness; Prognosis; RNA Interference; RNA, Small Interfering; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2015
Phosphorylation of eIF4E promotes EMT and metastasis via translational control of SNAIL and MMP-3.
    Oncogene, 2015, Apr-16, Volume: 34, Issue:16

    The progression of cancers from primary tumors to invasive and metastatic stages accounts for the overwhelming majority of cancer deaths. Understanding the molecular events which promote metastasis is thus critical in the clinic. Translational control is emerging as an important factor in tumorigenesis. The messenger RNA (mRNA) cap-binding protein eIF4E is an oncoprotein that has an important role in cancer initiation and progression. eIF4E must be phosphorylated to promote tumor development. However, the role of eIF4E phosphorylation in metastasis is not known. Here, we show that mice in which eukaryotic translation initiation factor 4E (eIF4E) cannot be phosphorylated are resistant to lung metastases in a mammary tumor model, and that cells isolated from these mice exhibit impaired invasion. We also demonstrate that transforming growth factor-beta (TGFβ) induces eIF4E phosphorylation to promote the translation of Snail and Mmp-3 mRNAs, and the induction of epithelial-to-mesenchymal transition (EMT). Furthermore, we describe a new model wherein EMT induced by TGFβ requires translational activation via the non-canonical TGFβ signaling branch acting through eIF4E phosphorylation.

    Topics: Animals; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Eukaryotic Initiation Factor-4E; Female; Lung Neoplasms; Mammary Neoplasms, Experimental; Matrix Metalloproteinase 3; Mice; Phosphorylation; Protein Biosynthesis; RNA, Messenger; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2015
SETBP1 mutations drive leukemic transformation in ASXL1-mutated MDS.
    Leukemia, 2015, Volume: 29, Issue:4

    Mutations in ASXL1 are frequent in patients with myelodysplastic syndrome (MDS) and are associated with adverse survival, yet the molecular pathogenesis of ASXL1 mutations (ASXL1-MT) is not fully understood. Recently, it has been found that deletion of Asxl1 or expression of C-terminal-truncating ASXL1-MTs inhibit myeloid differentiation and induce MDS-like disease in mice. Here, we find that SET-binding protein 1 (SETBP1) mutations (SETBP1-MT) are enriched among ASXL1-mutated MDS patients and associated with increased incidence of leukemic transformation, as well as shorter survival, suggesting that SETBP1-MT play a critical role in leukemic transformation of MDS. We identify that SETBP1-MT inhibit ubiquitination and subsequent degradation of SETBP1, resulting in increased expression. Expression of SETBP1-MT, in turn, inhibited protein phosphatase 2A activity, leading to Akt activation and enhanced expression of posterior Hoxa genes in ASXL1-mutant cells. Biologically, SETBP1-MT augmented ASXL1-MT-induced differentiation block, inhibited apoptosis and enhanced myeloid colony output. SETBP1-MT collaborated with ASXL1-MT in inducing acute myeloid leukemia in vivo. The combination of ASXL1-MT and SETBP1-MT activated a stem cell signature and repressed the tumor growth factor-β signaling pathway, in contrast to the ASXL1-MT-induced MDS model. These data reveal that SETBP1-MT are critical drivers of ASXL1-mutated MDS and identify several deregulated pathways as potential therapeutic targets in high-risk MDS.

    Topics: Adult; Animals; Apoptosis; Carrier Proteins; Cell Differentiation; Cell Transformation, Neoplastic; Gene Expression Regulation, Leukemic; HEK293 Cells; HL-60 Cells; Homeodomain Proteins; Humans; Leukemia, Myeloid, Acute; Mice; Mice, Inbred C57BL; Mutation; Myelodysplastic Syndromes; Nuclear Proteins; Protein Phosphatase 2; Proteolysis; Proto-Oncogene Proteins c-akt; Repressor Proteins; Signal Transduction; Survival Analysis; Transforming Growth Factor beta; Ubiquitination

2015
EDB Fibronectin Specific Peptide for Prostate Cancer Targeting.
    Bioconjugate chemistry, 2015, May-20, Volume: 26, Issue:5

    Extradomain-B fibronectin (EDB-FN), one of the oncofetal fibronectin (onfFN) isoforms, is a high-molecular-weight glycoprotein that mediates cell adhesion and migration. The expression of EDB-FN is associated with a number of cancer-related biological processes such as tumorigenesis, angiogenesis, and epithelial-to-mesenchymal transition (EMT). Here, we report the development of a small peptide specific to EDB-FN for targeting prostate cancer. A cyclic nonapeptide, CTVRTSADC (ZD2), was identified using peptide phage display. A ZD2-Cy5 conjugate was synthesized to accomplish molecular imaging of prostate cancer in vitro and in vivo. ZD2-Cy5 demonstrated effective binding to up-regulated EDB-FN secreted by TGF-β-induced PC3 cancer cells following EMT. Following intravenous injections, the targeted fluorescent probe specifically bound to and delineated PC3-GFP prostate tumors in nude mice bearing the tumor xenografts. ZD2-Cy5 also showed stronger binding to human prostate tumor specimens with a higher Gleason score (GS9) compared to those with a lower score (GS 7), with no binding in benign prostatic hyperplasia (BPH). Thus, the ZD2 peptide is a promising strategy for molecular imaging and targeted therapy of prostate cancer.

    Topics: Amino Acid Sequence; Animals; Carbocyanines; Cell Line, Tumor; Cell Transformation, Neoplastic; Coloring Agents; Fibronectins; Humans; Male; Mice; Mice, Nude; Molecular Targeted Therapy; Oligopeptides; Optical Imaging; Prostatic Neoplasms; Substrate Specificity; Transforming Growth Factor beta

2015
Inhibition of iNOS as a novel effective targeted therapy against triple-negative breast cancer.
    Breast cancer research : BCR, 2015, Feb-22, Volume: 17

    Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer with no effective targeted therapy. Inducible nitric oxide synthase (iNOS) is associated with poor survival in patients with breast cancer by increasing tumor aggressiveness. This work aimed to investigate the potential of iNOS inhibitors as a targeted therapy for TNBC. We hypothesized that inhibition of endogenous iNOS would decrease TNBC aggressiveness by reducing tumor initiation and metastasis through modulation of epithelial-mesenchymal transition (EMT)-inducing factors.. iNOS protein levels were determined in 83 human TNBC tissues and correlated with clinical outcome. Proliferation, mammosphere-forming efficiency, migration, and EMT transcription factors were assessed in vitro after iNOS inhibition. Endogenous iNOS targeting was evaluated as a potential therapy in TNBC mouse models.. High endogenous iNOS expression was associated with worse prognosis in patients with TNBC by gene expression as well as immunohistochemical analysis. Selective iNOS (1400 W) and pan-NOS (L-NMMA and L-NAME) inhibitors diminished cell proliferation, cancer stem cell self-renewal, and cell migration in vitro, together with inhibition of EMT transcription factors (Snail, Slug, Twist1, and Zeb1). Impairment of hypoxia-inducible factor 1α, endoplasmic reticulum stress (IRE1α/XBP1), and the crosstalk between activating transcription factor 3/activating transcription factor 4 and transforming growth factor β was observed. iNOS inhibition significantly reduced tumor growth, the number of lung metastases, tumor initiation, and self-renewal.. Considering the effectiveness of L-NMMA in decreasing tumor growth and enhancing survival rate in TNBC, we propose a targeted therapeutic clinical trial by re-purposing the pan-NOS inhibitor L-NMMA, which has been extensively investigated for cardiogenic shock as an anti-cancer therapeutic.

    Topics: Activating Transcription Factor 3; Animals; Antineoplastic Agents; Antineoplastic Combined Chemotherapy Protocols; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Disease Models, Animal; Endoplasmic Reticulum Stress; Enzyme Inhibitors; Epithelial-Mesenchymal Transition; Female; Gene Expression; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Lung Neoplasms; Mice; Molecular Targeted Therapy; Neoplasm Invasiveness; Nitric Oxide Synthase Type II; Prognosis; Transforming Growth Factor beta; Triple Negative Breast Neoplasms; Tumor Burden; Xenograft Model Antitumor Assays

2015
NPC1L1 knockout protects against colitis-associated tumorigenesis in mice.
    BMC cancer, 2015, Mar-27, Volume: 15

    Colorectal cancer is strongly associated with lipid metabolism. NPC1L1, a sterol transporter, plays a key role in modulating lipid homeostasis in vivo. Its inhibitor, ezetimibe, began to be used clinically to lower cholesterol and this caused the great debate on its role in causing carcinogenesis. Here we explored the role of NPC1L1 in colorectal tumorigenesis.. Wild-type mice and NPC1L1(-/-) (NPC1L1 knockout) mice were treated with azoxymethane (AOM)-dextran sodium sulfate (DSS) to induce colitis-associated colorectal tumorigenesis. Mice were sacrificed 10, 15, 18 or 20 weeks after AOM treatment, respectively. Colorectal tumors were counted and analyzed. Plasma lipid concentrations were measured using enzymatic reagent kit. Protein expression level was assayed by western blot.. NPC1L1(-/-) mice significantly had fewer tumors than wild-type. The ratio of malignant/tumor in NPC1L1(-/-) mice was significantly lower than in wild-type 20 weeks after AOM-DSS treatment. NPC1L1 was highly expressed in the small intestine of wild-type mice but its expression was undetectable in colorectal mucous membranes or tumors in either group. NPC1L1 knockout decreased plasma total cholesterol and phospholipid. NPC1L1(-/-) mice had significant lower intestinal inflammation scores and expressed inflammatory markers p-c-Jun, p-ERK and Caspase-1 p20 lower than wild-type. NPC1L1 knockout also reduced lymphadenectasis what may be caused by inflammation. NPC1L1 knockout in mice decreased β-catenin in tumors and regulated TGF-β and p-gp in adjacent colons or tumors. There was not detectable change of p53 by NPC1L1 knockout.. Our results provide the first evidence that NPC1L1 knockout protects against colitis-associated tumorigenesis. NPC1L1 knockout decreasing plasma lipid, especially cholesterol, to reduce inflammation and decreasing β-catenin, p-c-Jun and p-ERK may be involved in the mechanism.

    Topics: Animals; ATP Binding Cassette Transporter, Subfamily B, Member 1; Azoxymethane; beta Catenin; Cell Transformation, Neoplastic; Colitis; Colonic Neoplasms; Dextran Sulfate; Disease Models, Animal; Homozygote; Intestinal Mucosa; Lipids; Membrane Transport Proteins; Mice; Mice, Knockout; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2015
Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration.
    Integrative biology : quantitative biosciences from nano to macro, 2015, Volume: 7, Issue:10

    Tumors are stiff and data suggest that the extracellular matrix stiffening that correlates with experimental mammary malignancy drives tumor invasion and metastasis. Nevertheless, the relationship between tissue and extracellular matrix stiffness and human breast cancer progression and aggression remains unclear. We undertook a biophysical and biochemical assessment of stromal-epithelial interactions in noninvasive, invasive and normal adjacent human breast tissue and in breast cancers of increasingly aggressive subtype. Our analysis revealed that human breast cancer transformation is accompanied by an incremental increase in collagen deposition and a progressive linearization and thickening of interstitial collagen. The linearization of collagen was visualized as an overall increase in tissue birefringence and was most striking at the invasive front of the tumor where the stiffness of the stroma and cellular mechanosignaling were the highest. Amongst breast cancer subtypes we found that the stroma at the invasive region of the more aggressive Basal-like and Her2 tumor subtypes was the most heterogeneous and the stiffest when compared to the less aggressive luminal A and B subtypes. Intriguingly, we quantified the greatest number of infiltrating macrophages and the highest level of TGF beta signaling within the cells at the invasive front. We also established that stroma stiffness and the level of cellular TGF beta signaling positively correlated with each other and with the number of infiltrating tumor-activated macrophages, which was highest in the more aggressive tumor subtypes. These findings indicate that human breast cancer progression and aggression, collagen linearization and stromal stiffening are linked and implicate tissue inflammation and TGF beta.

    Topics: Biomechanical Phenomena; Biophysical Phenomena; Birefringence; Breast Neoplasms; Carcinoma, Intraductal, Noninfiltrating; Cell Transformation, Neoplastic; Collagen; Disease Progression; Extracellular Matrix; Female; Humans; Macrophages; Microscopy, Atomic Force; Microscopy, Fluorescence, Multiphoton; Neoplasm Invasiveness; Signal Transduction; Transforming Growth Factor beta

2015
Eukaryotic Translation Initiation Factor 4E Is a Feed-Forward Translational Coactivator of Transforming Growth Factor β Early Protransforming Events in Breast Epithelial Cells.
    Molecular and cellular biology, 2015, Volume: 35, Issue:15

    Eukaryotic translation initiation factor 4E (eIF4E) is overexpressed early in breast cancers in association with disease progression and reduced survival. Much remains to be understood regarding the role of eIF4E in human cancer. We determined, using immortalized human breast epithelial cells, that elevated expression of eIF4E translationally activates the transforming growth factor β (TGF-β) pathway, promoting cell invasion, a loss of cell polarity, increased cell survival, and other hallmarks of early neoplasia. Overexpression of eIF4E is shown to facilitate the selective translation of integrin β1 mRNA, which drives the translationally controlled assembly of a TGF-β receptor signaling complex containing α3β1 integrins, β-catenin, TGF-β receptor I, E-cadherin, and phosphorylated Smad2/3. This receptor complex acutely sensitizes nonmalignant breast epithelial cells to activation by typically substimulatory levels of activated TGF-β. TGF-β can promote cellular differentiation or invasion and transformation. As a translational coactivator of TGF-β, eIF4E confers selective mRNA translation, reprogramming nonmalignant cells to an invasive phenotype by reducing the set point for stimulation by activated TGF-β. Overexpression of eIF4E may be a proinvasive facilitator of TGF-β activity.

    Topics: beta Catenin; Breast Neoplasms; Cadherins; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Epithelial Cells; Eukaryotic Initiation Factor-4E; Female; Humans; Integrin alpha3beta1; Integrin beta1; Neoplasm Invasiveness; Phosphorylation; Receptors, Transforming Growth Factor beta; RNA Interference; RNA, Small Interfering; Signal Transduction; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta

2015
miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer-associated fibroblasts.
    Carcinogenesis, 2015, Volume: 36, Issue:8

    There is increasing evidence that the expression of microRNA (miRNA) in cancer is associated with chemosensitivity but the mechanism of miRNA-induced chemoresistance has not been fully elucidated. The aim of this study was to examine the role of extracellular miRNA in the response to chemotherapy in esophageal cancer. First, serum expression of miRNAs selected by miRNA array was measured by quantitative reverse transcription-polymerase chain reaction in 68 patients with esophageal cancer who received cisplatin-based chemotherapy to examine the relationship between miRNA expression and response to chemotherapy. The serum expression levels of 18 miRNAs were different between responders and non-responders by miRNA array. Of these, high expression levels of miR-27a/b correlated with poor response to chemotherapy in patients with esophageal cancer. Next, in vitro assays were conducted to investigate the mechanism of miRNA-induced chemoresistance. Although transfection of miR-27a/b to cancer cells had no significant impact on chemosensitivity, esophageal cancer cells cultured in supernatant of miR-27a/b-transfected normal fibroblast showed reduced chemosensitivity to cisplatin, compared with cancer cells cultured in supernatant of normal fibroblast. MiR-27a/b-transfected normal fibroblast showed α-smooth muscle actin (α-SMA) expression, a marker of cancer-associated fibroblasts (CAF) and increased production of transforming growth factor-β (TGF-β). Chemosensitivity recovered after administration of neutralizing antibody of TGF-β to the supernatant transfer experiments. Our results indicated that miR-27a/b is involved in resistance to chemotherapy in esophageal cancer, through miR-27a/b-induced transformation of normal fibroblast into CAF.

    Topics: Actins; Aged; Aged, 80 and over; Apoptosis; Cell Transformation, Neoplastic; Cells, Cultured; Cisplatin; Drug Resistance, Neoplasm; Esophageal Neoplasms; Fibroblasts; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Middle Aged; Reference Values; Transforming Growth Factor beta

2015
CDKN2B Loss Promotes Progression from Benign Melanocytic Nevus to Melanoma.
    Cancer discovery, 2015, Volume: 5, Issue:10

    Deletion of the entire CDKN2B-CDKN2A gene cluster is among the most common genetic events in cancer. The tumor-promoting effects are generally attributed to loss of CDKN2A-encoded p16 and p14ARF tumor suppressors. The degree to which the associated CDKN2B-encoded p15 loss contributes to human tumorigenesis is unclear. Here, we show that CDKN2B is highly upregulated in benign melanocytic nevi, contributes to maintaining nevus melanocytes in a growth-arrested premalignant state, and is commonly lost in melanoma. Using primary melanocytes isolated directly from freshly excised human nevi naturally expressing the common BRAF(V600E)-activating mutation, nevi progressing to melanoma, and normal melanocytes engineered to inducibly express BRAF(V600E), we show that BRAF activation results in reversible, TGFβ-dependent, p15 induction that halts proliferation. Furthermore, we engineer human skin grafts containing nevus-derived melanocytes to establish a new, architecturally faithful, in vivo melanoma model, and demonstrate that p15 loss promotes the transition from benign nevus to melanoma.. Although BRAF(V600E) mutations cause melanocytes to initially proliferate into benign moles, mechanisms responsible for their eventual growth arrest are unknown. Using melanocytes from human moles, we show that BRAF activation leads to a CDKN2B induction that is critical for restraining BRAF oncogenic effects, and when lost, contributes to melanoma.

    Topics: Animals; Cell Cycle Checkpoints; Cell Transformation, Neoplastic; Chromatin; Cyclin-Dependent Kinase Inhibitor p15; Disease Models, Animal; Disease Progression; DNA Mutational Analysis; Epigenesis, Genetic; Gene Expression Regulation, Neoplastic; Heterografts; Humans; Immunohistochemistry; Melanocytes; Melanoma; Mice; Mutation; Nevus; Proto-Oncogene Proteins B-raf; Signal Transduction; Transcriptional Activation; Transforming Growth Factor beta

2015
Crosstalk between epithelial-mesenchymal transition and castration resistance mediated by Twist1/AR signaling in prostate cancer.
    Endocrine-related cancer, 2015, Volume: 22, Issue:6

    Although invasive and metastatic progression via the epithelial-mesenchymal transition (EMT) and acquisition of resistance to castration are both critical steps in prostate cancer, the molecular mechanism of this interaction remains unclear. In this study, we aimed to elucidate the interaction of signaling between castration resistance and EMT, and to apply this information to the development of a novel therapeutic concept using transforming growth factor-β (TGF-β) inhibitor SB525334 combined with androgen-deprivation therapy against prostate cancer using an in vivo model. This study revealed that an EMT inducer (TGF-β) induced full-length androgen receptor (AR) and AR variant expression. In addition, a highly invasive clone showed augmented full-length AR and AR variant expression as well as acquisition of castration resistance. Conversely, full-length AR and AR as well as Twist1 and mesenchymal molecules variant expression were up-regulated in castration-resistant LNCaP xenograft. Finally, TGF-β inhibitor suppressed Twist1 and AR expression as well as prostate cancer growth combined with castration. Taken together, these results demonstrate that Twist1/AR signaling was augmented in castration resistant as well as mesenchymal-phenotype prostate cancer, indicating the molecular mechanism of mutual and functional crosstalk between EMT and castration resistance, which may play a crucial role in prostate carcinogenesis and progression.

    Topics: Adenocarcinoma; Androgens; Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Combined Modality Therapy; Epithelial-Mesenchymal Transition; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Imidazoles; Male; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Proteins; Neoplasms, Hormone-Dependent; Nuclear Proteins; Orchiectomy; Prostatic Neoplasms; Quinoxalines; Random Allocation; Receptors, Androgen; RNA Interference; RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta; Twist-Related Protein 1; Xenograft Model Antitumor Assays

2015
Mitogen-activated protein kinase activator with WD40 repeats (MAWD) and MAWD-binding protein induce cell differentiation in gastric cancer.
    BMC cancer, 2015, Sep-15, Volume: 15

    Our previous proteomic analysis revealed that mitogen-activated protein kinase activator with WD40 repeats (MAWD) and MAWD-binding protein (MAWBP) were downregulated in gastric cancer (GC) tissues. These proteins interacted and formed complexes in GC cells. To investigate the role of MAWD and MAWBP in GC differentiation, we analyzed the relationship between MAWD/MAWBP and clinicopathologic characteristics of GC tissues and examined the expression of E-cadherin and pepsinogen C (PGC)-used as gastric mucosa differentiation markers-in MAWD/MAWBP-overexpressing GC cells and xenografts.. We measured MAWD, MAWBP, transforming growth factor-beta (TGF-beta), E-cadherin, and PGC expression in 223 GC tissues and matched-adjacent normal tissues using tissue microarray and immunohistochemistry (IHC) analyses, and correlated these expression levels with clinicopathologic features. MAWD and MAWBP were overexpressed alone or together in SGC7901 cells and then E-cadherin, N-cadherin, PGC, Snail, and p-Smad2 levels were determined using western blotting, semiquantitative RT-PCR, and immunofluorescence analysis. Alkaline phosphatase (AKP) activity was measured to investigate the differentiation level of various transfected cells, and the transfected cells were used in tumorigenicity assays and for IHC analysis of protein expression in xenografts.. MAWD/MAWBP positive staining was significantly lower in GC tissues than in normal samples (P < 0.001), and the expression of these proteins was closely correlated with GC differentiation grade. Kaplan-Meier survival curves indicated that low MAWD and MAWBP expression was associated with poor patient survival (P < 0.05). The differentiation-related proteins E-cadherin and PGC were expressed in GC tissues at a lower level than in normal tissues (P < 0.001), but were upregulated in MAWD/MAWBP-overexpressing cells. N-cadherin and Snail expression was strongr in vector-expressing cells and comparatively weaker in MAWD/MAWBP co-overexpressing cells. MAWD/MAWBP co-overexpression inhibited Smad2 phosphorylation and nuclear translocation (P < 0.05), and AKP activity was lowest in MAWD/MAWBP coexpressing cells and highest in vector-expressing cells (P < 0.001). TGF-beta, E-cadherin, and PGC expression in xenograft tumors derived from MAWD/MAWBP coexpressing cells was higher than that in control.. MAWD and MAWBP were downregulated and associated with the differentiation grade in GC tissues. MAWD and MAWBP might induce the expression of differentiation-related proteins by modulating TGF-beta signaling in GC cells.

    Topics: Animals; Biomarkers; Cadherins; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Female; Gene Expression Regulation, Neoplastic; Heterografts; Humans; Mice; Neoplasm Grading; Neoplasm Proteins; Prognosis; Protein Interaction Maps; Proteins; RNA-Binding Proteins; Signal Transduction; Stomach Neoplasms; Transforming Growth Factor beta

2015
Modulation of thyroidal radioiodide uptake by oncological pipeline inhibitors and Apigenin.
    Oncotarget, 2015, Oct-13, Volume: 6, Issue:31

    Targeted radioiodine therapy for thyroid cancer is based on selective stimulation of Na+/I- Symporter (NIS)-mediated radioactive iodide uptake (RAIU) in thyroid cells by thyrotropin. Patients with advanced thyroid cancer do not benefit from radioiodine therapy due to reduced or absent NIS expression. To identify inhibitors that can be readily translated into clinical care, we examined oncological pipeline inhibitors targeting Akt, MEK, PI3K, Hsp90 or BRAF in their ability to increase RAIU in thyroid cells expressing BRAFV600E or RET/PTC3 oncogene. Our data showed that (1) PI3K inhibitor GDC-0941 outperformed other inhibitors in RAIU increase mainly by decreasing iodide efflux rate to a great extent; (2) RAIU increase by all inhibitors was extensively reduced by TGF-β, a cytokine secreted in the invasive fronts of thyroid cancers; (3) RAIU reduction by TGF-β was mainly mediated by NIS reduction and could be reversed by Apigenin, a plant-derived flavonoid; and (4) In the presence of TGF-β, GDC-0941 with Apigenin co-treatment had the highest RAIU level in both BRAFV600E expressing cells and RET/PTC3 expressing cells. Taken together, Apigenin may serve as a dietary supplement along with small molecule inhibitors to improve radioiodine therapeutic efficacy on invasive tumor margins thereby minimizing future metastatic events.

    Topics: Animals; Apigenin; Blotting, Western; Cell Transformation, Neoplastic; Cells, Cultured; Immunoenzyme Techniques; Iodine Radioisotopes; Proto-Oncogene Proteins c-ret; Radionuclide Imaging; Rats; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Small Molecule Libraries; Thyroid Gland; Tissue Distribution; Transforming Growth Factor beta

2015
Dual Roles of RNF2 in Melanoma Progression.
    Cancer discovery, 2015, Volume: 5, Issue:12

    Epigenetic regulators have emerged as critical factors governing the biology of cancer. Here, in the context of melanoma, we show that RNF2 is prognostic, exhibiting progression-correlated expression in human melanocytic neoplasms. Through a series of complementary gain-of-function and loss-of-function studies in mouse and human systems, we establish that RNF2 is oncogenic and prometastatic. Mechanistically, RNF2-mediated invasive behavior is dependent on its ability to monoubiquitinate H2AK119 at the promoter of LTBP2, resulting in silencing of this negative regulator of TGFβ signaling. In contrast, RNF2's oncogenic activity does not require its catalytic activity nor does it derive from its canonical gene repression function. Instead, RNF2 drives proliferation through direct transcriptional upregulation of the cell-cycle regulator CCND2. We further show that MEK1-mediated phosphorylation of RNF2 promotes recruitment of activating histone modifiers UTX and p300 to a subset of poised promoters, which activates gene expression. In summary, RNF2 regulates distinct biologic processes in the genesis and progression of melanoma via different molecular mechanisms.. The role of epigenetic regulators in cancer progression is being increasingly appreciated. We show novel roles for RNF2 in melanoma tumorigenesis and metastasis, albeit via different mechanisms. Our findings support the notion that epigenetic regulators, such as RNF2, directly and functionally control powerful gene networks that are vital in multiple cancer processes.

    Topics: Animals; Catalysis; Cell Transformation, Neoplastic; Cyclin D2; Disease Progression; E1A-Associated p300 Protein; Gene Expression; Gene Expression Regulation, Neoplastic; Histone Demethylases; Humans; Latent TGF-beta Binding Proteins; MAP Kinase Signaling System; Melanoma; Mice; Neoplasm Metastasis; Nuclear Proteins; Oncogenes; Phosphorylation; Polycomb Repressive Complex 1; Prognosis; Promoter Regions, Genetic; Transforming Growth Factor beta

2015
Knockdown of Cul4A increases chemosensitivity to gemcitabine through upregulation of TGFBI in lung cancer cells.
    Oncology reports, 2015, Volume: 34, Issue:6

    Cullin 4A (Cul4A) promotes oncogenesis through overexpression and then ubiquitination‑mediated proteolysis of tumor suppressors in various types of cancers. Transforming growth factor β‑induced (TGFBI) has been implicated as a tumor suppressor, which enhances gemcitabine chemosensitivity in lung cancer cells. The present study aimed to investigate the association of TGFBI and Cul4A and the mechanism by which Cul4A regulates TGFBI. In addition, we also evaluated the therapeutic value of Cul4A RNAi using adenoviral transfection of Cul4A RNAi in nude mouse xenograft models. We observed that knockdown of Cul4A was associated with increased sensitivity to gemcitabine through upregulation of TGFBI in lung cancer cells. Cul4A regulated TGFBI through direct interaction and then ubiquitin‑mediated protein degradation. In the nude mouse xenograft models, adenoviral transfection of Cul4A RNAi in combination with gemcitabine chemotherapy inhibited lung cancer tumor growth. As the result, combination of Cul4A RNAi with chemotherapy may provide a new approach to lung cancer treatment.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Cullin Proteins; Deoxycytidine; Drug Resistance, Neoplasm; Extracellular Matrix Proteins; Gemcitabine; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Humans; Lung Neoplasms; Mice; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2015
Intercellular HOCl-mediated Apoptosis Induction in Malignant Cells: Interplay Between NOX1-Dependent Superoxide Anion Generation and DUOX-related HOCl-generating Peroxidase Activity.
    Anticancer research, 2015, Volume: 35, Issue:11

    Intercellular apoptosis-inducing HOCl signaling is discussed as a control step during oncogenesis. It is defined as a sophisticated interplay between transformed target cells and non-transformed or transformed effector cells. In this study, transformed target cells were seeded as clumps of high local cell density, but low total cell number. They were surrounded by large numbers of effector cells, seeded at low local density. This spatially defined experimental arrangement allowed study of the impact of siRNA-mediated knockdown of NADPH oxidase 1 (NOX1) or dual oxidase 1 (DUOX1) on intercellular HOCl signaling. Our data show that the target function of transformed cells is defined as expression of NOX1 and subsequent extracellular superoxide anion generation. The NOX domain of DUOX1 does not contribute to the target function. The peroxidase domain of DUOX1 is released from transforming growth factor β1-treated non-transformed and transformed cells and acts in trans as HOCl-synthesizing peroxidase. These findings clarify the biochemical source of HOCl during HOCl-mediated signaling.

    Topics: Animals; Apoptosis; Blotting, Western; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Dual Oxidases; Female; Fibroblasts; Flow Cytometry; Humans; Hypochlorous Acid; NADH, NADPH Oxidoreductases; NADPH Oxidase 1; NADPH Oxidases; Ovary; Oxidants; Peroxidase; Rats; RNA, Small Interfering; Signal Transduction; Stomach Neoplasms; Superoxide Dismutase; Superoxides; Transforming Growth Factor beta

2015
Site-specific Effects of DUOX1-Related Peroxidase on Intercellular Apoptosis Signaling.
    Anticancer research, 2015, Volume: 35, Issue:11

    Intercellular apoptosis-inducing HOCl signaling is known as an interplay between superoxide anions/H₂O₂ of transformed target cells and dual oxidase 1 (DUOX1)-related peroxidase that is released from neighboring non-transformed or transformed effector cells. Effector cells are dispensable when the release of the peroxidase domain of DUOX1 from target cells is prevented through inhibition of matrix metalloproteinase (MMP) activity. Membrane-associated peroxidase is then co-localized to NADPH oxidase 1 (NOX1) and establishes HOCl signaling specifically in transformed cells, using the same biochemical pathways as classical intercellular HOCl signaling. Membrane-associated peroxidase protects against exogenous HOCl through reversal of the peroxidase reaction. In addition, membrane-associated peroxidase protects against NO/peroxynitrite signaling as it oxidates NO and decomposes peroxynitrite. The protective function of membrane-associated peroxidase (in the absence of MMP) is analogous to that of catalase, whereas the destructive effect of the enzyme, i.e. the synthesis of HOCl, is independent of its localization and of MMP activity.

    Topics: Animals; Apoptosis; Blotting, Western; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Dipeptides; Dual Oxidases; Fibroblasts; Flow Cytometry; Humans; Hypochlorous Acid; Matrix Metalloproteinase 2; Matrix Metalloproteinase Inhibitors; NADH, NADPH Oxidoreductases; NADPH Oxidase 1; NADPH Oxidases; Nitric Oxide; Oxidants; Peroxidase; Peroxynitrous Acid; Rats; RNA, Small Interfering; Signal Transduction; Stomach Neoplasms; Superoxide Dismutase; Superoxides; Transforming Growth Factor beta

2015
Cancer-associated fibroblasts might sustain the stemness of scirrhous gastric cancer cells via transforming growth factor-β signaling.
    International journal of cancer, 2014, Apr-15, Volume: 134, Issue:8

    Cancer-associated fibroblasts (CAFs) have recently been implicated in tumor growth and metastasis in gastric cancer. Cancer stem cells (CSCs) have been proposed to have an important role in cancer progression. The aim of this study was to clarify the effect of CAFs on CSCs characteristics in gastric carcinoma. Scirrhous gastric cancer cell lines, OCUM-12 and OCUM-2MD3, and non-scirrhous gastric cancer cell lines, MKN-45 and MKN-74, were used. OCUM-12/side population (SP) cells and OCUM-2MD3/SP cells were sorted by flow cytometry as CSC-rich cells from the parent cells. CaF-37 was established from the tumoral gastric specimens as CAFs. Flow cytometric analysis of SP fraction, spheroid colony assay, and RT-PCR analysis of CSC markers were performed to identify CSCs properties. Effect of CAFs on the tumorigenicity by OCUM-12/SP cells was examined using nude mice. CAF CM significantly increased the percentages of the SP fraction of OCUM-12/SP and OCUM-2MD3/SP cells, but not that of MKN-45/SP and MKN-74/SP cells. Taken together, CM from CaF-37 significantly increased the number of spheroid colonies and the expression level of CSC markers of OCUM-12/SP and OCUM-2MD3/SP cells. These stimulating-activities by CM were significantly decreased by TGFβ inhibitors, but not FGFR and cMet inhibitor. Tumorigenicity by subcutaneous coinoculation of OCUM-12/SP cells with CAFs was significantly high in comparison with that by OCUM-12/SP cells alone. Phospho-Smad2 expression level was significantly increased by co-inoculation with CAFs. These findings suggested that CAFs might regulate the stemness of CSCs in scirrhous gastric cancer by TGFβ signaling.

    Topics: Actins; Adenocarcinoma, Scirrhous; Animals; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Female; Fibroblasts; Humans; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Middle Aged; Neoplastic Stem Cells; Proto-Oncogene Proteins c-met; Receptor, Fibroblast Growth Factor, Type 1; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad2 Protein; Spheroids, Cellular; Stomach Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment

2014
Gastritis promotes an activated bone marrow-derived mesenchymal stem cell with a phenotype reminiscent of a cancer-promoting cell.
    Digestive diseases and sciences, 2014, Volume: 59, Issue:3

    Bone marrow-derived mesenchymal stem cells (BM-MSCs) promote gastric cancer in response to gastritis. In culture, BM-MSCs are prone to mutation with continued passage but it is unknown whether a similar process occurs in vivo in response to gastritis.. The purpose of this study was to identify the role of chronic gastritis in the transformation of BM-MSCs leading to an activated cancer-promoting phenotype.. Age matched C57BL/6 (BL/6) and gastrin deficient (GKO) mice were used for isolation of stomach, serum and mesenchymal stem cells (MSCs) at 3 and 6 months of age. MSC activation was assessed by growth curve analysis, fluorescence-activated cell sorting and xenograft assays. To allow for the isolation of bone marrow-derived stromal cells and assay in response to chronic gastritis, IRG/Vav-1(Cre) mice that expressed both enhanced green fluorescent protein-expressing hematopoietic cells and red fluorescent protein-expressing stromal cells were generated. In a parabiosis experiment, IRG/Vav-1(Cre) mice were paired to either an uninfected Vav-1(Cre) littermate or a BL/6 mouse inoculated with Helicobacter pylori.. GKO mice displayed severe atrophic gastritis accompanied by elevated gastric tissue and circulating transforming growth factor beta (TGFβ) by 3 months of age. Compared to BM-MSCs isolated from uninflamed BL/6 mice, BM-MSCs isolated from GKO mice displayed an increased proliferative rate and elevated phosphorylated-Smad3 suggesting active TGFβ signaling. In xenograft assays, mice injected with BM-MSCs from 6-month-old GKO animals displayed tumor growth. RFP+ stromal cells were rapidly recruited to the gastric mucosa of H. pylori parabionts and exhibited changes in gene expression.. Gastritis promotes the in vivo activation of BM-MSCs to a phenotype reminiscent of a cancer-promoting cell.

    Topics: Animals; Biomarkers; Cell Proliferation; Cell Transformation, Neoplastic; Gastric Mucosa; Gastrins; Gastritis, Atrophic; Hedgehog Proteins; Helicobacter Infections; Helicobacter pylori; Immunoblotting; Mesenchymal Stem Cells; Mice; Mice, Inbred C57BL; Mice, Transgenic; Parabiosis; Phenotype; Real-Time Polymerase Chain Reaction; Smad3 Protein; Transforming Growth Factor beta

2014
FGFR1-WNT-TGF-β signaling in prostate cancer mouse models recapitulates human reactive stroma.
    Cancer research, 2014, Jan-15, Volume: 74, Issue:2

    The reactive stroma surrounding tumor lesions performs critical roles ranging from supporting tumor cell proliferation to inducing tumorigenesis and metastasis. Therefore, it is critical to understand the cellular components and signaling control mechanisms that underlie the etiology of reactive stroma. Previous studies have individually implicated fibroblast growth factor receptor 1 (FGFR1) and canonical WNT/β-catenin signaling in prostate cancer progression and the initiation and maintenance of a reactive stroma; however, both pathways are frequently found to be coactivated in cancer tissue. Using autochthonous transgenic mouse models for inducible FGFR1 (JOCK1) and prostate-specific and ubiquitously expressed inducible β-catenin (Pro-Cat and Ubi-Cat, respectively) and bigenic crosses between these lines (Pro-Cat × JOCK1 and Ubi-Cat × JOCK1), we describe WNT-induced synergistic acceleration of FGFR1-driven adenocarcinoma, associated with a pronounced fibroblastic reactive stroma activation surrounding prostatic intraepithelial neoplasia (mPIN) lesions found both in in situ and reconstitution assays. Both mouse and human reactive stroma exhibited increased transforming growth factor-β (TGF-β) signaling adjacent to pathologic lesions likely contributing to invasion. Furthermore, elevated stromal TGF-β signaling was associated with higher Gleason scores in archived human biopsies, mirroring murine patterns. Our findings establish the importance of the FGFR1-WNT-TGF-β signaling axes as driving forces behind reactive stroma in aggressive prostate adenocarcinomas, deepening their relevance as therapeutic targets.

    Topics: Adenocarcinoma; Animals; Biopsy; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Disease Models, Animal; Fibroblasts; Humans; Inflammation; Male; Mice; Mice, Nude; Mice, Transgenic; Prostatic Neoplasms; Receptor, Fibroblast Growth Factor, Type 1; Signal Transduction; Species Specificity; Stromal Cells; Transforming Growth Factor beta; Wnt Proteins

2014
Six1 promotes epithelial-mesenchymal transition and malignant conversion in human papillomavirus type 16-immortalized human keratinocytes.
    Carcinogenesis, 2014, Volume: 35, Issue:6

    Six1, a member of the Six family of homeodomain transcription factors, is overexpressed in various human cancers, and SIX1 overexpression is associated with tumor progression and metastasis. Six1 messenger RNA levels increase during in vitro progression of human papillomavirus type 16 (HPV16)-immortalized human keratinocytes (HKc/HPV16) toward a differentiation-resistant (HKc/DR) phenotype. In this study, we show that HKc/DR-overexpressing Six1 exhibited a more mesenchymal phenotype, as characterized by a fibroblastic appearance and increased invasion. We utilized Whole Human Genome Microarrays to explore the gene expression changes associated with Six1 overexpression in HKc/DR. We found that overexpression of Six1 downregulated epithelial-related genes and upregulated mesenchymal-related genes, which suggests that Six1 overexpression induces epithelial-mesenchymal transition (EMT). Pathway analysis of the microarray data showed alterations in the transforming growth factor-beta (TGF-β) pathway, including enhanced expression of the TGF-β receptor type II (TβRII), and activation of the mitogen-activated protein kinase (MAPK) pathway in HKc/DR-overexpressing Six1, suggesting that Smad-independent pathways of TGF-β signaling may be involved in Six1-mediated EMT. p38 MAPK activation was required for sustained Six1-induced EMT and TβRII overexpression. Finally, we determined that Six1 overexpression in HKc/DR resulted in malignant conversion and increased the cancer stem cell (CSC)-like population. Thus, Six1 overexpression promotes EMT, CSCs properties and malignant conversion in HKc/DR through MAPK activation, which supports the possible use of p38-TβRII inhibitors for the treatment of cancers overexpressing Six1.

    Topics: Animals; Cell Line, Transformed; Cell Line, Tumor; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cluster Analysis; Disease Models, Animal; Epithelial-Mesenchymal Transition; Female; Gene Expression; Gene Expression Profiling; HeLa Cells; Heterografts; Homeodomain Proteins; Human papillomavirus 16; Humans; Keratinocytes; MAP Kinase Signaling System; Mice; Neoplastic Stem Cells; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Burden

2014
C-kit induces epithelial-mesenchymal transition and contributes to salivary adenoid cystic cancer progression.
    Oncotarget, 2014, Mar-30, Volume: 5, Issue:6

    Epithelial-mesenchymal transition (EMT) is associated with salivary adenoid cystic cancer (ACC) progression and metastasis. Here, we report that ectopic overexpression of c-kit in ACC cell lines is sufficient for acquisition of mesenchymal traits, enhanced cell invasion, along with stem cell properties defined by the presence of a CD133+/CD44+ cell subpopulation. c-kit positively regulated expression of known EMT inducers, also activating TGF-β to contribute to EMT. c-kit itself was induced by TGF-β in ACC cell lines and required for TGF-β-induced EMT. Xenograft experiments showed that c-kit cooperated with oncogenic Ras to promote tumorigenesis in vivo. Finally, in human specimens of ACC, we found that c-kit was abnormally overexpressed and correlated with the prognosis of ACC. Our findings define an important function for c-kit in ACC progression by orchestrating EMT, and they implicate this gene product as a marker of poor prognosis in this disease.

    Topics: Apoptosis; Blotting, Western; Carcinoma, Adenoid Cystic; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Disease Progression; Epithelial-Mesenchymal Transition; Flow Cytometry; Fluorescent Antibody Technique; Gene Expression Regulation, Neoplastic; Genes, ras; Humans; Immunoenzyme Techniques; Neoplasm Invasiveness; Neoplastic Stem Cells; Prognosis; Proto-Oncogene Proteins c-kit; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Salivary Gland Neoplasms; Signal Transduction; Survival Rate; Transforming Growth Factor beta; Tumor Cells, Cultured

2014
Impact of Mucin1 knockdown on the phenotypic characteristics of the human hepatocellular carcinoma cell line SMMC-7721.
    Oncology reports, 2014, Volume: 31, Issue:6

    Mucin1 (MUC1) is a transmembrane glycoprotein that plays a key role as an oncogene in the tumorigenesis of many human adenocarcinomas. However, the role of MUC1 in human hepatocellular carcinoma (HCC) progression remains unclear. In the present study, we silenced MUC1 to investigate its effect on the human HCC cell line SMMC-7721 and found that knockdown of MUC1 significantly inhibited cell proliferation, enhanced cell-cell aggregation and induced apoptosis. No significant differences were found in in vitro migration or invasion. We also observed that knockdown of MUC1 decreased the translocation of β‑catenin to the nucleus, reduced the activity of T cell factor and blocked the expression of cyclin D1 and c-Myc. In addition, MUC1 knockdown enhanced the expression of E-cadherin, a molecular chaperone of β‑catenin that plays an important role in cell-cell aggregation. In vivo assays demonstrated that there was no tumor growth in mice injected with MUC1-silenced cells. Global gene expression analysis showed that a series of genes encoding molecules in the Wnt/β‑catenin, nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), insulin, transforming growth factor β (TGF-β) and vascular endothelial growth factor (VEGF) signaling pathways were all influenced by the knockdown of MUC1, and these may contribute to the phenotypic alterations observed. Collectively, our results indicate that MUC1 plays a key role in HCC tumorigenesis.

    Topics: Animals; Apoptosis; beta Catenin; Cadherins; Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Gene Knockdown Techniques; Humans; Liver Neoplasms; Mice; Mitogen-Activated Protein Kinases; Mucin-1; NF-kappa B; Signal Transduction; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2014
Reprogramming tumor-infiltrating dendritic cells for CD103+ CD8+ mucosal T-cell differentiation and breast cancer rejection.
    Cancer immunology research, 2014, Volume: 2, Issue:5

    Our studies showed that tumor-infiltrating dendritic cells (DC) in breast cancer drive inflammatory Th2 (iTh2) cells and protumor inflammation. Here, we show that intratumoral delivery of the β-glucan curdlan, a ligand of dectin-1, blocks the generation of iTh2 cells and prevents breast cancer progression in vivo. Curdlan reprograms tumor-infiltrating DCs via the ligation of dectin-1, enabling the DCs to become resistant to cancer-derived thymic stromal lymphopoietin (TSLP), to produce IL-12p70, and to favor the generation of Th1 cells. DCs activated via dectin-1, but not those activated with TLR-7/8 ligand or poly I:C, induce CD8+ T cells to express CD103 (αE integrin), a ligand for cancer cells, E-cadherin. Generation of these mucosal CD8+ T cells is regulated by DC-derived integrin αvβ8 and TGF-β activation in a dectin-1-dependent fashion. These CD103+ CD8+ mucosal T cells accumulate in the tumors, thereby increasing cancer necrosis and inhibiting cancer progression in vivo in a humanized mouse model of breast cancer. Importantly, CD103+ CD8+ mucosal T cells elicited by reprogrammed DCs can reject established cancer. Thus, reprogramming tumor-infiltrating DCs represents a new strategy for cancer rejection.

    Topics: Animals; beta-Glucans; Breast Neoplasms; CD8-Positive T-Lymphocytes; Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Dendritic Cells; Disease Models, Animal; Female; Humans; Lectins, C-Type; Mice; Mucous Membrane; Signal Transduction; T-Lymphocyte Subsets; Th2 Cells; Transforming Growth Factor beta

2014
Brief reports: A distinct DNA methylation signature defines breast cancer stem cells and predicts cancer outcome.
    Stem cells (Dayton, Ohio), 2014, Volume: 32, Issue:11

    Self-renewal and differentiation are two epigenetic programs that regulate stem cells fate. Dysregulation of these two programs leads to the development of cancer stem cells (CSCs). Recent evidence suggests that CSCs are relatively resistant to conventional therapies and responsible for metastasis formation. Deciphering these processes will help understand oncogenesis and allow the development of new targeted therapies. Here, we have used a whole genome promoter microarray to establish the DNA methylation portraits of breast cancer stem cells (bCSCs) and non-bCSCs. A total of 68 differentially methylated regions (DMRs) were more hypomethylated in bCSCs than in non-bCSCs. Using a differentiation assay we demonstrated that DMRs are rapidly hypermethylated within the first 6 hours following induction of CSC differentiation whereas the cells reached the steady-state within 6 days, suggesting that these DMRs are linked to early CSC epigenetic regulation. These DMRs were significantly enriched in genes coding for TGF-β signaling-related proteins. Interestingly, DMRs hypomethylation was correlated to an overexpression of TGF-β signaling genes in a series of 109 breast tumors. Moreover, patients with tumors harboring the bCSC DMRs signature had a worse prognosis than those with non-bCSC DMRs signature. Our results show that bCSCs have a distinct DNA methylation landscape with TGF-β signaling as a key epigenetic regulator of bCSCs differentiation.

    Topics: Breast Neoplasms; Cell Differentiation; Cell Transformation, Neoplastic; DNA Methylation; Embryonic Stem Cells; Epigenesis, Genetic; Female; Humans; Neoplastic Stem Cells; Transforming Growth Factor beta

2014
Definition of smad3 phosphorylation events that affect malignant and metastatic behaviors in breast cancer cells.
    Cancer research, 2014, Nov-01, Volume: 74, Issue:21

    Smad3, a major intracellular mediator of TGFβ signaling, functions as both a positive and negative regulator in carcinogenesis. In response to TGFβ, the TGFβ receptor phosphorylates serine residues at the Smad3 C-tail. Cancer cells often contain high levels of the MAPK and CDK activities, which can lead to the Smad3 linker region becoming highly phosphorylated. Here, we report, for the first time, that mutation of the Smad3 linker phosphorylation sites markedly inhibited primary tumor growth, but significantly increased lung metastasis of breast cancer cell lines. In contrast, mutation of the Smad3 C-tail phosphorylation sites had the opposite effect. We show that mutation of the Smad3 linker phosphorylation sites greatly intensifies all TGFβ-induced responses, including growth arrest, apoptosis, reduction in the size of putative cancer stem cell population, epithelial-mesenchymal transition, and invasive activity. Moreover, all TGFβ responses were completely lost on mutation of the Smad3 C-tail phosphorylation sites. Our results demonstrate a critical role of the counterbalance between the Smad3 C-tail and linker phosphorylation in tumorigenesis and metastasis. Our findings have important implications for therapeutic intervention of breast cancer.

    Topics: Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Humans; Mutation; Neoplasm Metastasis; Phosphorylation; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad3 Protein; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2014
The MEKK1 PHD ubiquitinates TAB1 to activate MAPKs in response to cytokines.
    The EMBO journal, 2014, Nov-03, Volume: 33, Issue:21

    Unlike the other MAP3Ks, MEKK1 (encoded by Map3k1) contains a PHD motif. To understand the role of this motif, we have created a knockin mutant of mouse Map3k1 (Map3k1(m) (PHD)) with an inactive PHD motif. Map3k1(m) (PHD) ES cells demonstrate that the MEKK1 PHD controls p38 and JNK activation during TGF-β, EGF and microtubule disruption signalling, but does not affect MAPK responses to hyperosmotic stress. Protein microarray profiling identified the adaptor TAB1 as a PHD substrate, and TGF-β- or EGF-stimulated Map3k1(m) (PHD) ES cells exhibit defective non-canonical ubiquitination of MEKK1 and TAB1. The MEKK1 PHD binds and mediates the transfer of Lys63-linked poly-Ub, using the conjugating enzyme UBE2N, onto TAB1 to regulate TAK1 and MAPK activation by TGF-β and EGF. Both the MEKK1 PHD and TAB1 are critical for ES-cell differentiation and tumourigenesis. Map3k1(m) (PHD) (/+) mice exhibit aberrant cardiac tissue, B-cell development, testis and T-cell signalling.

    Topics: Adaptor Proteins, Signal Transducing; Amino Acid Motifs; Animals; Cell Differentiation; Cell Transformation, Neoplastic; Embryonic Stem Cells; Epidermal Growth Factor; MAP Kinase Kinase 4; MAP Kinase Kinase Kinase 1; MAP Kinase Signaling System; Mice; Mice, Knockout; p38 Mitogen-Activated Protein Kinases; Polyubiquitin; Protein Binding; Transforming Growth Factor beta; Ubiquitination

2014
NF-κB functions in tumor initiation by suppressing the surveillance of both innate and adaptive immune cells.
    Cell reports, 2014, Oct-09, Volume: 9, Issue:1

    NF-κB is considered a major contributor to tumor development, but how this factor functions in the initial stages of oncogenesis is not clear. In a model of Ras-induced transformation, we probed NF-κB function as preneoplastic cells formed tumors in mice. As previously shown, the p65 subunit of NF-κB acts as a tumor suppressor in normal cells by sustaining senescence following DNA damage. Our current data reveal that, following immortalization, p65 switches to an oncogene by counteracting the surveillance properties of immune cells. NF-κB exerts this effect by protecting transformed cells against macrophage-derived proapoptotic factors, tumor necrosis factor, and nitric oxide. Additionally, NF-κB acts through transforming growth factor beta (TGF-β) to mitigate T cell cytotoxicity and other factors to expand myeloid-derived suppressor cells. Together, these data suggest that NF-κB functions in the early stages of transformation by suppressing immune surveillance of both innate and adaptive immune cells, information that may be useful for targeted immunotherapies.

    Topics: Adaptive Immunity; Animals; Cell Transformation, Neoplastic; Cytoskeletal Proteins; Immunity, Innate; Macrophages; Mice; Mice, Inbred C57BL; Mice, SCID; Mice, Transgenic; Microfilament Proteins; NF-kappa B; Phosphoproteins; Precancerous Conditions; ras Proteins; Risk Factors; Signal Transduction; Transforming Growth Factor beta

2014
Suppression of Lefty expression in induced pluripotent cancer cells.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2013, Volume: 27, Issue:6

    Cancer and stem cells share the ability to silence tumor suppressors. We focused on Lefty, which encodes one of the most abundant tumor suppressors in embryonic stem (ES) cells and is not expressed in somatic cancer cells. We found that transforming growth factor β (TGF-β) induced demethylation of the Lefty B cytosine-phosphate-guanine (CpG) island and increased Lefty expression (10-200 times) in human pancreatic cancer cells and human liver cancer cells (PLC/PRF/5 and HLF). Expression of Cripto, another important factor in Nodal-Lefty signaling, was not increased after adding TGF-β. We generated reprogrammed cancer cells that revealed high expression of immature marker proteins, high proliferation, and the potential to express morphological patterns of ectoderm, mesoderm, and endoderm, suggesting that these cells may have cancer stem cell-like phenotypes. We investigated Lefty and found that reprogrammed human liver cancer cells (induced pluripotent cancer cells) displayed a much lower ability to express Lefty, although less Lefty B CpG methylation was also observed. We also found that a MEK inhibitor dramatically enhanced Lefty expression in human pancreatic cancers with mutated ras, whereas Lefty B CpG methylation was not decreased. These observations indicate that despite the demethylation of DNA strands in promoter regions of pluripotency-associated genes, including Lefty gene, Lefty expression was not induced well in reprogrammed cells. Of note was the fact that Lefty is abundantly expressed in human ES cells but not in induced pluripotent stem (iPS) cells. We thus think that reprogrammed cancer cells share the mechanism for expression of Lefty with iPS cells. This shared mechanism may contribute to the cancerous transformation of iPS cells.

    Topics: Cell Line, Tumor; Cell Transdifferentiation; Cell Transformation, Neoplastic; CpG Islands; DNA Methylation; Down-Regulation; Genes, Tumor Suppressor; Humans; Induced Pluripotent Stem Cells; Left-Right Determination Factors; Promoter Regions, Genetic; Suppression, Genetic; Transforming Growth Factor beta

2013
Inflammation-mediated genetic and epigenetic alterations drive cancer development in the neighboring epithelium upon stromal abrogation of TGF-β signaling.
    PLoS genetics, 2013, Volume: 9, Issue:2

    Deletion of tumor suppressor genes in stromal fibroblasts induces epithelial cancer development, suggesting an important role of stroma in epithelial homoeostasis. However, the underlying mechanisms remain to be elucidated. Here we report that deletion of the gene encoding TGFβ receptor 2 (Tgfbr2) in the stromal fibroblasts (Tgfbr2(fspKO)) induces inflammation and significant DNA damage in the neighboring epithelia of the forestomach. This results in loss or down-regulation of cyclin-dependent kinase inhibitors p15, p16, and p21, which contribute to the development of invasive squamous cell carcinoma (SCC). Anti-inflammation treatment restored p21 expression, delayed tumorigenesis, and increased survival of Tgfbr2(fspKO) mice. Our data demonstrate for the first time that inflammation is a critical player in the epigenetic silencing of p21 in tumor progression. Examination of human esophageal SCC showed a down-regulation of TGFβ receptor 2 (TβRII) in the stromal fibroblasts, as well as increased inflammation, DNA damage, and loss or decreased p15/p16 expression. Our study suggests anti-inflammation may be a new therapeutic option in treating human SCCs with down-regulation of TβRII in the stroma.

    Topics: Animals; Apoptosis; Breast Neoplasms; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor Proteins; Down-Regulation; Epigenesis, Genetic; Epithelial Cells; Esophageal Neoplasms; Esophageal Squamous Cell Carcinoma; Female; Fibroblasts; Humans; Inflammation; Mice; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2013
Expression profiles of SnoN in normal and cancerous human tissues support its tumor suppressor role in human cancer.
    PloS one, 2013, Volume: 8, Issue:2

    SnoN is a negative regulator of TGF-β signaling and also an activator of the tumor suppressor p53 in response to cellular stress. Its role in human cancer is complex and controversial with both pro-oncogenic and anti-oncogenic activities reported. To clarify its role in human cancer and provide clinical relevance to its signaling activities, we examined SnoN expression in normal and cancerous human esophageal, ovarian, pancreatic and breast tissues. In normal tissues, SnoN is expressed in both the epithelium and the surrounding stroma at a moderate level and is predominantly cytoplasmic. SnoN levels in all tumor epithelia examined are lower than or similar to that in the matched normal samples, consistent with its anti-tumorigenic activity in epithelial cells. In contrast, SnoN expression in the stroma is highly upregulated in the infiltrating inflammatory cells in high-grade esophageal and ovarian tumor samples, suggesting that SnoN may potentially promote malignant progression through modulating the tumor microenvironment in these tumor types. The overall levels of SnoN expression in these cancer tissues do not correlate with the p53 status. However, in human cancer cell lines with amplification of the snoN gene, a strong correlation between increased SnoN copy number and inactivation of p53 was detected, suggesting that the tumor suppressor SnoN-p53 pathway must be inactivated, either through downregulation of SnoN or inactivation of p53, in order to allow cancer cell to proliferate and survive. These data strongly suggest that SnoN can function as a tumor suppressor at early stages of tumorigenesis in human cancer tissues.

    Topics: Adult; Breast; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Progression; Esophageal Neoplasms; Esophagus; Female; Gene Expression Regulation, Neoplastic; Humans; Intracellular Signaling Peptides and Proteins; Middle Aged; Ovarian Neoplasms; Ovary; Pancreas; Pancreatic Neoplasms; Proto-Oncogene Proteins; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2013
Breast fibroblasts modulate early dissemination, tumorigenesis, and metastasis through alteration of extracellular matrix characteristics.
    Neoplasia (New York, N.Y.), 2013, Volume: 15, Issue:3

    A wealth of evidence has now demonstrated that the microenvironment in which a tumorigenic cell evolves is as critical to its evolution as the genetic mutations it accrues. However, there is still relatively little known about how signals from the microenvironment contribute to the early events in the progression to malignancy. To address this question, we used a premalignant mammary model to examine how fibroblasts, and the extracellular matrix (ECM) proteins they secrete, influence progression to malignancy. Their effect on metastatic malignant cells was also assessed for comparison. We found that carcinoma-associated fibroblasts, and the distinct aligned ECM they deposit, can cause both premalignant and malignant mammary epithelial cells to assume a mesenchymal morphology that is associated with increased dissemination and metastasis, while benign reduction mammoplasty fibroblasts favor the maintenance of an epithelial morphology and constrain early dissemination, tumor growth, and metastasis. Our results suggest that normalizing the organization of the ECM could be effective in limiting systemic dissemination and tumor growth.

    Topics: Animals; Breast; Cell Line, Tumor; Cell Transformation, Neoplastic; Coculture Techniques; Epithelial Cells; Extracellular Matrix; Extracellular Signal-Regulated MAP Kinases; Female; Fibroblasts; Humans; Lung Neoplasms; Mammary Neoplasms, Experimental; Neoplasm Metastasis; Phenotype; Proto-Oncogene Proteins c-jun; rho GTP-Binding Proteins; Signal Transduction; Transforming Growth Factor beta

2013
Dual effects of Ral-activated pathways on p27 localization and TGF-β signaling.
    Molecular biology of the cell, 2013, Volume: 24, Issue:11

    Constitutive activation or overactivation of Ras signaling pathways contributes to epithelial tumorigenesis in several ways, one of which is cytoplasmic mislocalization of the cyclin-dependent kinase inhibitor p27(Kip1) (p27). We previously showed that such an effect can be mediated by activation of the Ral-GEF pathway by oncogenic N-Ras. However, the mechanism(s) leading to p27 cytoplasmic accumulation downstream of activated Ral remained unknown. Here, we report a dual regulation of p27 cellular localization by Ral downstream pathways, based on opposing effects via the Ral effectors RalBP1 and phospholipase D1 (PLD1). Because RalA and RalB are equally effective in mislocalizing both murine and human p27, we focus on RalA and murine p27, which lacks the Thr-157 phosphorylation site of human p27. In experiments based on specific RalA and p27 mutants, complemented with short hairpin RNA-mediated knockdown of Ral downstream signaling components, we show that activation of RalBP1 induces cytoplasmic accumulation of p27 and that this event requires p27 Ser-10 phosphorylation by protein kinase B/Akt. Of note, activation of PLD1 counteracts this effect in a Ser-10-independent manner. The physiological relevance of the modulation of p27 localization by Ral is demonstrated by the ability of Ral-mediated activation of the RalBP1 pathway to abrogate transforming growth factor-β-mediated growth arrest in epithelial cells.

    Topics: Animals; ATP-Binding Cassette Transporters; Cell Transformation, Neoplastic; Chlorocebus aethiops; COS Cells; Cyclin-Dependent Kinase Inhibitor p27; Cytoplasm; Epithelial Cells; Gene Expression Regulation; GTPase-Activating Proteins; HEK293 Cells; Humans; Mice; Mink; Phospholipase D; Phosphorylation; Protein Transport; Proto-Oncogene Proteins c-akt; ral GTP-Binding Proteins; RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta

2013
Effects of the Smad4 C324Y mutation on thyroid cell proliferation.
    International journal of oncology, 2013, Volume: 42, Issue:6

    Smad4 is a key mediator of the transforming growth factor-β (TGF-β) superfamily that is involved in the control of cell proliferation and differentiation. We recently demonstrated that a Smad4 mutation, Smad4 C324Y, isolated from nodal metastases of papillary thyroid carcinoma, causes an increase of TGF-β signaling, responsible for the acquisition of transformed phenotype and invasive behaviour in thyroid cells stably expressing this mutation. In this paper, we demonstrate that the stable expression of Smad4 C324Y mutation in FRTL-5 cells is responsible for TSH-independent growth ability. Our data show that the Smad4 C324Y mutation interacts with P-Smad3 more strongly than Smad4 wt, already in basal condition; this interaction is responsible for TGF-β signaling and PKA activation that, in turn, determines an increased phosphorylation of CREB, necessary for the mitogenic actions of TSH. The expression of cyclin D1 also increases in all cells overexpressing the Smad4 C324Y mutation. All together, these data demonstrate that Smad4 C324Y mutation, interacting with the PKA pathway, gives cells the ability to proliferate independently from TSH.

    Topics: Carcinoma; Carcinoma, Papillary; Cell Differentiation; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Cyclic AMP Response Element-Binding Protein; Cyclic AMP-Dependent Protein Kinases; Cyclin D1; Humans; Mutation; Phosphorylation; Smad3 Protein; Smad4 Protein; Thyroglobulin; Thyroid Cancer, Papillary; Thyroid Gland; Thyroid Neoplasms; Thyrotropin; Transforming Growth Factor beta

2013
L1-ORF1p, a Smad4 interaction protein, promotes proliferation of HepG2 cells and tumorigenesis in mice.
    DNA and cell biology, 2013, Volume: 32, Issue:9

    Long interspersed nucleotide element (LINE-1; L1) as an autonomous retrotransposon is localized usually in AT-rich, low-recombined, and gene-poor regions of genome. It is transiently activated in embryonic development and continuously activated in all tumor cells tested so far. Full-length L1 gene contains 5' untranslated region, two open reading frames (ORFs) encoded L1ORF1p and L1ORF2p, and a 3' terminal polyadenylation site. Compared with L1ORF2p, a protein encompassing reverse transcriptase and endonuclease activities, L1ORF1p remains to be elucidated. With liver cancer cells and tissues, the expression and sub-localization of L1ORF1p were investigated and shown that L1-ORF1p expresses just in liver cancer cells and tissues but not in normal liver cells and almost not in adjacent tissues. To characterize L1ORF1p, the strategies for over-expression and down-regulation of L1ORF1p in transfected cells were implemented. The phenomenon of promoting cell proliferation and colony formation was observed in transfected cells with L1ORF1p over-expression and vice versa. Down-regulation of L1ORF1p suppresses tumorigenesis in vitro and in vivo. Smad4 as an interaction protein of L1ORF1p is identified for the first time, while L1ORF1p is responsible for Smad4 sequestration in the cytoplasm. Thus, L1ORF1p contributed to tumorigenesis and may attribute to, at least partly, its participation in Smad4-signaling regulation.

    Topics: 5' Untranslated Regions; Animals; Cell Proliferation; Cell Transformation, Neoplastic; Cytoplasm; Gene Expression Regulation, Neoplastic; Genome, Human; HEK293 Cells; Humans; Liver Neoplasms; Liver Neoplasms, Experimental; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Open Reading Frames; Protein Interaction Mapping; Ribonucleoproteins; Smad4 Protein; Transfection; Transforming Growth Factor beta

2013
Tumor microenvironmental signaling elicits epithelial-mesenchymal plasticity through cooperation with transforming genetic events.
    Neoplasia (New York, N.Y.), 2013, Volume: 15, Issue:9

    Epithelial-to-mesenchymal transition (EMT) facilitates the escape of epithelial cancer cells from the primary tumor site, which is a key event early in metastasis. Here, we explore how extrinsic, tumor microenvironmental cytokines cooperate with intrinsic, genetic changes to promote EMT in human mammary epithelial cells (HMECs). Viral transduction of transforming genetic events into HMECs routinely generated two distinct cell populations. One population retained epithelial characteristics, while an emergent population spontaneously acquired a mesenchymal morphology and properties associated with cancer stem cells (CSCs). Interestingly, the spontaneous mesenchymal/CSCs were unable to differentiate and lacked epithelial-mesenchymal plasticity. In contrast, exposure of the transformed HMECs retaining epithelial characteristics to exogenous transforming growth factor-β (TGF-β) generated a mesenchymal/CSC population with remarkable plasticity. The TGF-β-induced mesenchymal/CSC population was dependent on the continued presence of TGF-β. Removal of TGF-β or pharmacologic or genetic inhibition of TGF-β/SMAD signaling led to the reversion of mesenchymal/CSC to epithelial/non-CSC. Our results demonstrate that targeting exogenous cytokine signaling disrupts epithelial-mesenchymal plasticity and may be an effective strategy to inhibit the emergence of circulating tumor cells. The model of epithelial-mesenchymal plasticity we describe here can be used to identify novel tumor microenvironmental factors and downstream signaling that cooperate with intrinsic genetic changes to drive metastasis. Understanding the interaction between extrinsic and intrinsic factors that regulate epithelial-mesenchymal plasticity will allow the development of new therapies that target tumor microenvironmental signals to reduce metastasis.

    Topics: Animals; Breast Neoplasms; Cell Proliferation; Cell Transformation, Neoplastic; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Humans; Mammary Glands, Human; Mesenchymal Stem Cells; Mice; Mice, Nude; Neoplasm Transplantation; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Microenvironment; Xenograft Model Antitumor Assays

2013
Intestinal GUCY2C prevents TGF-β secretion coordinating desmoplasia and hyperproliferation in colorectal cancer.
    Cancer research, 2013, Nov-15, Volume: 73, Issue:22

    Tumorigenesis is a multistep process that reflects intimate reciprocal interactions between epithelia and underlying stroma. However, tumor-initiating mechanisms coordinating transformation of both epithelial and stromal components are not defined. In humans and mice, initiation of colorectal cancer is universally associated with loss of guanylin and uroguanylin, the endogenous ligands for the tumor suppressor guanylyl cyclase C (GUCY2C), disrupting a network of homeostatic mechanisms along the crypt-surface axis. Here, we reveal that silencing GUCY2C in human colon cancer cells increases Akt-dependent TGF-β secretion, activating fibroblasts through TGF-β type I receptors and Smad3 phosphorylation. In turn, activating TGF-β signaling induces fibroblasts to secrete hepatocyte growth factor (HGF), reciprocally driving colon cancer cell proliferation through cMET-dependent signaling. Elimination of GUCY2C signaling in mice (Gucy2c(-/-)) produces intestinal desmoplasia, with increased reactive myofibroblasts, which is suppressed by anti-TGF-β antibodies or genetic silencing of Akt. Thus, GUCY2C coordinates intestinal epithelial-mesenchymal homeostasis through reciprocal paracrine circuits mediated by TGF-β and HGF. In that context, GUCY2C signaling constitutes a direct link between the initiation of colorectal cancer and the induction of its associated desmoplastic stromal niche. The recent regulatory approval of oral GUCY2C ligands to treat chronic gastrointestinal disorders underscores the potential therapeutic opportunity for oral GUCY2C hormone replacement to prevent remodeling of the microenvironment essential for colorectal tumorigenesis.

    Topics: Animals; Caco-2 Cells; Cell Proliferation; Cell Transformation, Neoplastic; Colorectal Neoplasms; Down-Regulation; Epithelial-Mesenchymal Transition; Fibrosis; HCT116 Cells; Humans; Intestinal Mucosa; Intestines; Mice; Mice, Inbred C57BL; Mice, Knockout; Receptors, Enterotoxin; Receptors, Guanylate Cyclase-Coupled; Receptors, Peptide; Stem Cell Niche; Transforming Growth Factor beta

2013
Loss of canonical Smad4 signaling promotes KRAS driven malignant transformation of human pancreatic duct epithelial cells and metastasis.
    PloS one, 2013, Volume: 8, Issue:12

    Pancreatic ductal adenocarcinoma (PDAC) is the fourth most common cause of cancer death in North America. Activating KRAS mutations and Smad4 loss occur in approximately 90% and 55% of PDAC, respectively. While their roles in the early stages of PDAC development have been confirmed in genetically modified mouse models, their roles in the multistep malignant transformation of human pancreatic duct cells have not been directly demonstrated. Here, we report that Smad4 represents a barrier in KRAS-mediated malignant transformation of the near normal immortalized human pancreatic duct epithelial (HPDE) cell line model. Marked Smad4 downregulation by shRNA in KRAS (G12V) expressing HPDE cells failed to cause tumorigenic transformation. However, KRAS-mediated malignant transformation occurred in a new HPDE-TGF-β resistant (TβR) cell line that completely lacks Smad4 protein expression and is resistant to the mito-inhibitory activity of TGF-β. This transformation resulted in tumor formation and development of metastatic phenotype when the cells were implanted orthotopically into the mouse pancreas. Smad4 restoration re-established TGF-β sensitivity, markedly increased tumor latency by promoting apoptosis, and decreased metastatic potential. These results directly establish the critical combination of the KRAS oncogene and complete Smad4 inactivation in the multi-stage malignant transformation and metastatic progression of normal human HPDE cells.

    Topics: Animals; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial Cells; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Gene Silencing; Humans; Mice; Neoplasm Invasiveness; Neoplasm Metastasis; Pancreatic Ducts; Proto-Oncogene Proteins; Proto-Oncogene Proteins p21(ras); ras Proteins; RNA, Small Interfering; Signal Transduction; Smad4 Protein; Survival Analysis; Transforming Growth Factor beta

2013
Small molecule inhibitors promote efficient generation of induced pluripotent stem cells from human skeletal myoblasts.
    Stem cells and development, 2013, Jan-01, Volume: 22, Issue:1

    Human somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs) by ectopic expression of key transcription factors. iPSCs have been generated from a variety of cell types. However, iPSC induction from human myoblasts has not yet been reported. Human primary skeletal myoblasts can be cultured from diagnostic muscle biopsy specimens, and thousands of lines are frozen and stored in biobanks, and are a valuable source for iPSC-based etiological and pathogenic studies. Our aim was to generate iPSCs from human skeletal myoblasts enriched from muscle biopsy samples. We used retro- or Sendai virus vector-mediated reprogramming of enriched human myoblasts from 7 donors. We show that stable iPSC lines can be generated from human myoblasts at efficiency similar to that of fibroblasts when appropriate media is used, and the efficiency of the feeder-free iPSC generation can be significantly improved by inhibitors of histone deacetylase (sodium butyrate) and TGF-β signaling (SB431542).

    Topics: Adult; Animals; Antigens, Differentiation; Benzamides; Butyric Acid; Cell Culture Techniques; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Dioxoles; Female; Gene Silencing; Histone Deacetylase Inhibitors; Humans; Induced Pluripotent Stem Cells; Infant; Infant, Newborn; Male; Mice; Mice, Nude; Middle Aged; Muscle, Skeletal; Myoblasts, Skeletal; Retroviridae; Sendai virus; Signal Transduction; Teratoma; Transduction, Genetic; Transforming Growth Factor beta; Young Adult

2013
TGF-β promotes colorectal cancer cell growth in vitro and in vivo.
    Hepato-gastroenterology, 2013, Volume: 60, Issue:124

    TGF-β has dual functions as a tumor promoter or a tumor suppressor. Recent studies suggest the roles of TGF-β might change from a tumor suppressor to a tumor promoter, when lacking SMAD4 expression in CRC (colorectal cancer). However, the precise role of TGF-β as tumor promoter in CRC is still unclear.. We evaluated the role of TGF-β in SMAD4-null CRC SW620 cells. In vitro, we measured cell growth and cell cycle distribution by flow cytometry. In vivo, we implanted SW620 cells into mice and measured tumor weight after TGF-β treatment. We also determined cell proliferation ability in tumor by immunohistochemistry of proliferating cell nuclear antigen (PCNA).. Cell growth and DNA synthesis in S phase was remarkably enhanced by TGF-β in vitro. Besides, TGF-β-activated ERK1/2 MAPK signal was also observed. In vivo, TGF-β enhanced tumor growth and cell proliferation in tumor.. TGF-β serves as a tumor promoter, which promotes CRC cell growth in vitro and in vivo. These suggest that TGF-β might be developed as an effective therapeutic target for CRC patients.

    Topics: Animals; Cell Cycle; Cell Line, Tumor; Cell Transformation, Neoplastic; Colorectal Neoplasms; Flow Cytometry; Immunohistochemistry; In Vitro Techniques; Mice; Mitogen-Activated Protein Kinase 3; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta

2013
COUP-TFII inhibits TGF-β-induced growth barrier to promote prostate tumorigenesis.
    Nature, 2013, Jan-10, Volume: 493, Issue:7431

    Mutations in phosphatase and tensin homologue (PTEN) or genomic alterations in the phosphatidylinositol-3-OH kinase-signalling pathway are the most common genetic alterations reported in human prostate cancer. However, the precise mechanism underlying how indolent tumours with PTEN alterations acquire metastatic potential remains poorly understood. Recent studies suggest that upregulation of transforming growth factor (TGF)-β signalling triggered by PTEN loss will form a growth barrier as a defence mechanism to constrain prostate cancer progression, underscoring that TGF-β signalling might represent a pre-invasive checkpoint to prevent PTEN-mediated prostate tumorigenesis. Here we show that COUP transcription factor II (COUP-TFII, also known as NR2F2), a member of the nuclear receptor superfamily, serves as a key regulator to inhibit SMAD4-dependent transcription, and consequently overrides the TGF-β-dependent checkpoint for PTEN-null indolent tumours. Overexpression of COUP-TFII in the mouse prostate epithelium cooperates with PTEN deletion to augment malignant progression and produce an aggressive metastasis-prone tumour. The functional counteraction between COUP-TFII and SMAD4 is reinforced by genetically engineered mouse models in which conditional loss of SMAD4 diminishes the inhibitory effects elicited by COUP-TFII ablation. The biological significance of COUP-TFII in prostate carcinogenesis is substantiated by patient sample analysis, in which COUP-TFII expression or activity is tightly correlated with tumour recurrence and disease progression, whereas it is inversely associated with TGF-β signalling. These findings reveal that the destruction of the TGF-β-dependent barrier by COUP-TFII is crucial for the progression of PTEN-mutant prostate cancer into a life-threatening disease, and supports COUP-TFII as a potential drug target for the intervention of metastatic human prostate cancer.

    Topics: Animals; Cell Cycle Checkpoints; Cell Line, Tumor; Cell Transformation, Neoplastic; COUP Transcription Factor II; Disease Models, Animal; Disease Progression; Gene Deletion; Humans; Male; Mice; Neoplasm Metastasis; Proportional Hazards Models; Prostate; Prostatic Neoplasms; PTEN Phosphohydrolase; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta

2013
The chick embryo as an experimental system for melanoma cell invasion.
    PloS one, 2013, Volume: 8, Issue:1

    A primary cutaneous melanoma will not kill the patient, but its metastases. Since in vitro studies on melanoma cells in 2-D cultures do often not reflect reality, 3-D models might come closer to the physiological situation in the patient during cancer initiation and progression.. Here, we describe the chick embryo model for in vivo studies of melanoma cell migration and invasion. After transplantation of neural crest-derived melanoma cells into the neural tube, the melanoma cells resume neural crest cell migration along the medial and lateral pathways and finally undergo apoptosis in the target areas. Upon transplantation into ectopic areas such as the hindbrain or the optic cup malignant invasion and local tissue destruction occurs. In contrast, melanocytes are not able to spontaneously resume neural crest cell migration. However, malignant invasion can be induced in melanocytes by pre-treatment with the TGF-beta family members bone morphegenetic protein-2 or nodal. Transplantation of MCF7 breast cancer cells yields a different growth pattern in the rhombencephalon than melanoma cells.. The chick embryo model is a feasible, cost-effective in vivo system to study invasion by cancer cells in an embryonic environment. It may be useful to study invasive behavior induced by embryonic oncogenes and for targeted manipulation of melanoma or breast cancer cells aiming at ablation of invasive properties.

    Topics: Animals; Bone Morphogenetic Protein 2; Brain Neoplasms; Cell Movement; Cell Transformation, Neoplastic; Chick Embryo; Epithelial-Mesenchymal Transition; Humans; MCF-7 Cells; Melanocytes; Melanoma; Neoplasm Invasiveness; Neural Crest; Rhombencephalon; Tissue Culture Techniques; Transforming Growth Factor beta

2013
MicroRNA miR-146b-5p regulates signal transduction of TGF-β by repressing SMAD4 in thyroid cancer.
    Oncogene, 2012, Apr-12, Volume: 31, Issue:15

    MicroRNAs (miRNA) are small non-coding RNAs involved in post-transcriptional gene regulation that have crucial roles in several types of tumors, including papillary thyroid carcinoma (PTC). miR-146b-5p is overexpressed in PTCs and is regarded as a relevant diagnostic marker for this type of cancer. A computational search revealed that miR-146b-5p putatively binds to the 3' untranslated region (UTR) of SMAD4, an important member of the transforming growth factor β (TGF-β) signaling pathway. The TGF-β pathway is a negative regulator of thyroid follicular cell growth, and the mechanism by which thyroid cancer cells evade its inhibitory signal remains unclear. We questioned whether the modulation of the TGF-β pathway by miR-146b-5p can contribute to thyroid tumorigenesis. Luciferase reporter assay confirmed the direct binding of miR-146b-5p on the SMAD4 3'UTR. Specific inhibition of miR-146b-5p with a locked nucleic acid-modified anti-miR-146b oligonucleotide significantly increased SMAD4 levels in the human papillary carcinoma cell lines, TPC-1 and BCPAP. Moreover, suppression of miR-146b-5p increased the cellular response to the TGF-β anti-proliferative signal, significantly decreasing the proliferation rate. The overexpression of miR-146b-5p in normal rat follicular PCCL3 cells decreased SMAD4 levels and disrupted TGF-β signal transduction. MiR-146b-5p overexpression in PCCL3 cells also significantly increased cell proliferation in the absence of thyroid-stimulating hormone and conferred resistance to TGF-β-mediated cell-cycle arrest. Additionally, the activation of thyroid most common oncogenes RET/PTC3 and BRAF in PCCL3 cells upregulated miR-146b-5p expression. Our results confirm the oncogenic role of miR-146b-5p in thyroid follicular cells and contribute to knowledge regarding the modulation of TGF-β signal transduction by miRNAs in PTCs.

    Topics: 3' Untranslated Regions; Animals; Carcinoma, Papillary; Cell Line, Tumor; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Oncogenes; Rats; Signal Transduction; Smad4 Protein; Thyroid Gland; Thyroid Neoplasms; Transforming Growth Factor beta

2012
Heterogeneity in MYC-induced mammary tumors contributes to escape from oncogene dependence.
    Oncogene, 2012, May-17, Volume: 31, Issue:20

    A hallmark of human cancer is heterogeneity, reflecting the complex series of changes resulting in the activation of oncogenes coupled with inactivation of tumor suppressor genes. Breast cancer is no exception and indeed, many studies have revealed considerable complexity and heterogeneity in the population of primary breast tumors and substantial changes in a recurrent breast tumor that has acquired metastatic properties and drug resistance. We have made use of a Myc-inducible transgenic mouse model of breast cancer in which elimination of Myc activity following tumor development initially leads to a regression of a subset of tumors generally followed by de novo Myc-independent growth. We have observed that tumors that grow independent of Myc expression have gene profiles that are distinct from the primary tumors with characteristics indicative of an epithelial-mesenchymal transition (EMT) phenotype. Phenotypic analyses of Myc-independent tumors confirm the acquisition of an EMT phenotype suggested to be associated with invasive and migratory properties in human cancer cells. Further genomic analyses reveal mouse mammary tumors growing independent of myc have a higher probability of exhibiting a gene signature similar to that observed for human 'tumor-initiating' cells. Collectively, the data reveal genetic alterations that underlie tumor progression and an escape from Myc-dependent growth in a transgenic mouse model that can provide insights to what occurs in human cancers as they acquire drug resistance and metastatic properties.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Disease Models, Animal; Epithelial-Mesenchymal Transition; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Genes, ras; Humans; Mammary Neoplasms, Experimental; Mice; Mice, Transgenic; Neoplastic Stem Cells; Proto-Oncogene Proteins c-myc; Transforming Growth Factor alpha; Transforming Growth Factor beta

2012
CD49f and CD61 identify Her2/neu-induced mammary tumor-initiating cells that are potentially derived from luminal progenitors and maintained by the integrin-TGFβ signaling.
    Oncogene, 2012, May-24, Volume: 31, Issue:21

    Human epidermal growth factor receptor 2 (HER2)/Neu is overexpressed in 20-30% of breast cancers and associated with aggressive phenotypes and poor prognosis. For deciphering the role of HER2/Neu in breast cancer, mouse mammary tumor virus (MMTV)-Her2/neu transgenic mice that develop mammary tumors resembling human HER2-subtype breast cancer have been established. Several recent studies have revealed that HER2/Neu is overexpressed in and regulates self renewal of breast tumor-initiating cells (TICs). However, in the MMTV-Her2/neu transgenic mouse model, the identity of TICs remains elusive, despite previous studies showing supportive evidence for existence of TICs in Her2/neu-induced mammary tumors. Through systematic screening and characterization, we identified that surface markers CD49f, CD61 and ESA were aberrantly overexpressed in Her2-overexpressing mammary tumor cells. Analysis of these markers and CD24 detected anomalous expansion of the luminal progenitor population in preneoplastic mammary glands of Her2/neu transgenic mice, indicating that aberrant luminal progenitors originated in Her2-induced mammary tumors. The combined markers, CD49f and CD61, further delineated the CD49f(high)CD61(high)-sorted fraction as a TIC-enriched population, which displayed increased tumorsphere formation ability, enhanced tumorigenicity both in vitro and in vivo and drug resistance to pacitaxel and doxorubicin. Moreover, the TIC-enriched population manifested increased transforming growth factor-β (TGFβ) signaling and exhibited gene expression signatures of stemness, TGFβ signaling and epithelial-to-mesenchymal transition. Our findings that self-renewal and clonogenicity of TICs were suppressed by pharmacologically inhibiting the TGFβ signaling further indicate that the TGFβ pathway is vital for maintenance of the TIC population. Finally, we showed that the integrin-β3 (CD61) signaling pathway was required for sustaining active TGFβ signaling and self-renewal of TICs. We for the first time developed a technique to highly enrich TICs from mammary tumors of Her2/neu transgenic mice, unraveled their properties and identified the cooperative integrin-β3-TGFβ signaling axis as a potential therapeutic target for HER2-induced TICs.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Female; Humans; Integrin alpha6; Integrin beta3; Mammary Neoplasms, Animal; Mice; Mice, Transgenic; Neoplastic Stem Cells; Receptor, ErbB-2; Signal Transduction; Transforming Growth Factor beta

2012
2-Amino-3-methylimidazo[4,5-f]quinoline (IQ) promotes mouse hepatocarcinogenesis by activating transforming growth factor-β and Wnt/β-catenin signaling pathways.
    Toxicological sciences : an official journal of the Society of Toxicology, 2012, Volume: 125, Issue:2

    The purposes of the present study were to investigate the modifying effects of 2-amino-3-methylimidazo[4,5-f]quinoline (IQ), a genotoxic carcinogen produced during cooking of protein-rich foods, and elucidate underlying mechanisms in a two-stage hepatocarcinogenesis mice model. Six-week-old B6C3F1 mice were subjected to two-thirds partial hepatectomy at the beginning of the study, followed by an intraperitoneal injection of diethylnitrosamine on day 1. Starting 1 week later, they were fed diets containing IQ at doses of 30, 100, or 300 ppm for 39 weeks. A dose-dependent trend for increase in eosinophilic altered foci as well as eosinophilic hepatocellular adenomas was observed, along with significant elevation in the incidence of hepatocellular carcinomas in the 100- and 300-ppm IQ groups as compared with initiation control group. Furthermore, IQ elevated the protein expression levels of Wnt1, transforming growth factor-β (TGF-β), TGF-β receptors 1 and 2 (TβR1 and TβR2), and phosphorylated c-Jun (p-c-Jun), while suppressing those of E-cadherin and p21(WAF1/Cip1). Moreover, translocation of β-catenin to the nuclei as well as upregulated nuclear expression of c-Myc and cyclin D1, which are downstream targets of β-catenin and p-c-Jun, were detected at 100 and 300 ppm. These findings suggest that IQ exerts dose-dependent promoting effects on mice hepatocarcinogenesis by activating TGF-β and Wnt/β-catenin signaling pathways and inhibiting cell adhesion.

    Topics: Animals; beta Catenin; Blotting, Western; Body Weight; Carcinoma, Hepatocellular; Cell Adhesion; Cell Transformation, Neoplastic; Diethylnitrosamine; Dose-Response Relationship, Drug; Drinking; Eating; Hepatectomy; Immunohistochemistry; Liver; Liver Neoplasms, Experimental; Male; Mice; Organ Size; Quinolines; Time Factors; Transforming Growth Factor beta; Wnt Proteins; Wnt Signaling Pathway

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
Bone morphogenetic protein (BMP) signaling regulates mitotic checkpoint protein levels in human breast cancer cells.
    Cellular signalling, 2012, Volume: 24, Issue:4

    Aberrant expression of mitotic checkpoint genes compromises mitotic checkpoint, leads to chromosome instability and tumorigenesis. However, the cell signals that control mitotic checkpoint gene expression have not been reported so far. In the present study we show that, in human breast cancer cells, chemical inhibition of Bone morphogenetic proteins (BMPs), but not Transforming Growth Factor-β (TGF-β), abrogates the mitotic arrest induced by nocodazole. Protein expression analysis reveals that inhibition of BMP signaling dramatically down regulates protein levels of mitotic checkpoint components BUB3, Hec1, TTK and MAD2, but inhibition of TGF-β has relatively minor effect on the expression of these proteins. Activation of BMP signaling specifically up regulates BUB3, and activation of Activin A signaling globally down regulates these proteins level. Furthermore, overexpressing MAD2, TTK, BUB3 or Hec1 significantly rescues the mitotic arrest defect caused by BMP inhibition. Our results demonstrated for the first time that TGF-β family cytokines are cellular signals regulating mitotic checkpoint and perturbations in intrinsic BMP signaling could lead to suppression of mitotic checkpoint signaling by downregulating key checkpoint proteins. The results suggest a possible mechanism by which dysregulation of TGF-β signaling causes mitotic checkpoint defects and drives tumorigenesis. The finding also provides a potential and more specific strategy for cancer prevention by targeting BMP and mitotic checkpoint connection.

    Topics: Activins; Benzamides; Bone Morphogenetic Proteins; Breast Neoplasms; Calcium-Binding Proteins; Cell Cycle Checkpoints; Cell Cycle Proteins; Cell Line, Tumor; Cell Transformation, Neoplastic; Cytoskeletal Proteins; Dioxoles; Female; Gene Expression Regulation, Neoplastic; HEK293 Cells; Humans; Mad2 Proteins; Nocodazole; Nuclear Proteins; Plasmids; Poly-ADP-Ribose Binding Proteins; Protein Kinase Inhibitors; Protein Serine-Threonine Kinases; Protein-Tyrosine Kinases; Pyrazoles; Pyrimidines; Repressor Proteins; Signal Transduction; Transfection; Transforming Growth Factor beta

2012
The miR-106b-25 cluster targets Smad7, activates TGF-β signaling, and induces EMT and tumor initiating cell characteristics downstream of Six1 in human breast cancer.
    Oncogene, 2012, Dec-13, Volume: 31, Issue:50

    The role of TGF-β signaling in tumorigenesis is paradoxical: it can be tumor suppressive or tumor promotional, depending on context. The metastatic regulator, Six1, was recently shown to mediate this switch, providing a novel means to explain this elusive 'TGF-β paradox'. Herein, we identify a mechanism by which Six1 activates the tumor promotional arm of TGF-β signaling, via its ability to upregulate the miR-106b-25 microRNA cluster, and further identify a novel function for this cluster of microRNAs. Although expression of the miR-106b-25 cluster is known to overcome TGF-β-mediated growth suppression via targeting p21 and BIM, we demonstrate for the first time that this same cluster can additionally target the inhibitory Smad7 protein, resulting in increased levels of the TGF-β type I receptor and downstream activation of TGF-β signaling. We further show that the miR-106b-25 cluster is sufficient to induce an epithelial-to-mesenchymal transition and a tumor initiating cell phenotype, and that it is required downstream of Six1 to induce these phenotypes. Finally, we demonstrate a significant correlation between miR-106b, Six1, and activated TGF-β signaling in human breast cancers, and further show that high levels of miR-106b and miR-93 in breast tumors significantly predicts shortened time to relapse. These findings expand the spectrum of oncogenic functions of miR-106b-25, and may provide a novel molecular explanation, through the Six1 regulated miR-106b-25 cluster, by which TGF-β signaling shifts from tumor suppressive to tumor promoting.

    Topics: Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; MCF-7 Cells; MicroRNAs; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad7 Protein; Transforming Growth Factor beta; Up-Regulation

2012
Loss of TGF-β responsiveness in prostate stromal cells alters chemokine levels and facilitates the development of mixed osteoblastic/osteolytic bone lesions.
    Molecular cancer research : MCR, 2012, Volume: 10, Issue:4

    Loss of TGF-β type II receptor (TβRII, encoded by Tgfbr2) expression in the prostate stroma contributes to prostate cancer initiation, progression, and invasion. We evaluated whether TβRII loss also affected prostate cancer bone metastatic growth. Immunohistologic analysis revealed that TβRII expression was lost in cancer-associated fibroblasts in human prostate cancer bone metastatic tissues. We recapitulated the human situation with a conditional stromal Tgfbr2 knockout (Tgfbr2-KO) mouse model. Conditioned media from primary cultured Tgfbr2-KO or control Tgfbr2-flox prostatic fibroblasts (koPFCM or wtPFCM, respectively) were applied to C4-2B prostate cancer cells before grafting the cells tibially. We found that koPFCM promoted prostate cancer cell growth in the bone and development of early mixed osteoblastic/osteolytic bone lesions. Furthermore, the koPFCM promoted greater C4-2B adhesion to type-I collagen, the major component of bone matrix, compared to wtPFCM-treated C4-2B. Cytokine antibody array analysis revealed that koPFCM had more than two-fold elevation in granulocyte colony-stimulating factor and CXCL1, CXCL16, and CXCL5 expression relative to wtPFCM. Interestingly, neutralizing antibodies of CXCL16 or CXCL1 were able to reduce koPFCM-associated C4-2B type-I collagen adhesion to that comparable with wtPFCM-mediated adhesion. Collectively, our data indicate that loss of TGF-β responsiveness in prostatic fibroblasts results in upregulation of CXCL16 and CXCL1 and that these paracrine signals increase prostate cancer cell adhesion in the bone matrix. These microenvironment changes at the primary tumor site can mediate early establishment of prostate cancer cells in the bone and support subsequent tumor development at the metastatic site.

    Topics: Animals; Bone Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Chemokines; Fibroblasts; Humans; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, SCID; Osteoblasts; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Stromal Cells; Transforming Growth Factor beta

2012
CBL enhances breast tumor formation by inhibiting tumor suppressive activity of TGF-β signaling.
    Oncogene, 2012, Dec-13, Volume: 31, Issue:50

    Casitas B-lineage lymphoma (CBL) protein family functions as multifunctional adaptor proteins and E3 ubiquitin ligases that are implicated as regulators of signaling in various cell types. Recent discovery revealed mutations of proto-oncogenic CBL in the linker region and RING finger domain in human acute myeloid neoplasm, and these transforming mutations induced carcinogenesis. However, the adaptor function of CBL mediated signaling pathway during tumorigenesis has not been well characterized. Here, we show that CBL is highly expressed in breast cancer cells and significantly inhibits transforming growth factor-β (TGF-β) tumor suppressive activity. Knockdown of CBL expression resulted in the increased expression of TGF-β target genes, PAI-I and CDK inhibitors such as p15(INK4b) and p21(Cip1). Furthermore, we demonstrate that CBL is frequently overexpressed in human breast cancer tissues, and the loss of CBL decreases the tumorigenic activity of breast cancer cells in vivo. CBL directly binds to Smad3 through its proline-rich motif, thereby preventing Smad3 from interacting with Smad4 and blocking nuclear translocation of Smad3. CBL-b, one of CBL protein family, also interacted with Smad3 and knockdown of both CBL and CBL-b further enhanced TGF-β transcriptional activity. Our findings provide evidence for a previously undescribed mechanism by which oncogenic CBL can block TGF-β tumor suppressor activity.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Cyclin-Dependent Kinases; Female; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Genes, Tumor Suppressor; Humans; Mice; Mice, Nude; Plasminogen Activator Inhibitor 1; Protein Binding; Protein Transport; Proto-Oncogene Proteins c-cbl; Signal Transduction; Smad3 Protein; Transcription, Genetic; Transforming Growth Factor beta; Transplantation, Heterologous

2012
Interleukin-1β and transforming growth factor-β cooperate to induce neurosphere formation and increase tumorigenicity of adherent LN-229 glioma cells.
    Stem cell research & therapy, 2012, Feb-10, Volume: 3, Issue:1

    Glioma stem cells (GSCs) have the property of self-renewal and appear to be a driving force for the initiation and recurrence of gliomas. We recently found that the human tumorigenic LN-229 glioma cell line failed to form neurospheres in serum-free conditions and generated mostly small tumors in vivo, suggesting that either LN-229 GSCs are not active in these conditions or GSCs are absent in the LN-229 cell line.. Using self-renewal assay, soft-agar colony assay, cell proliferation assay, invasion assay, real time PCR analysis, ELISA and in vivo tumorigenic assay, we investigated the effects of interleukin (IL)-1β and transforming growth factor (TGF)-β on the development of GSCs from LN-229 cells.. Here, we demonstrate that the combination of IL-1β and TGF-β can induce LN-229 cells to form neurospheres in serum-free medium. IL-1β/TGF-β-induced neurospheres display up-regulated expression of stemness factor genes (nestin, Bmi-1, Notch-2 and LIF), and increased invasiveness, drug resistance and tumor growth in vivo: hallmarks of GSCs. These results indicate that IL-1β and TGF-β cooperate to induce a GSC phenotype in the LN-229 cell line. Induction of nestin, LIF and Notch-2 by IL-1β/TGF-β can be reverted after cytokine withdrawal. Remarkably, however, up-regulated Bmi-1 levels remained unchanged after cytokine withdrawal; and the cytokine-withdrawn cells maintained strong clonogenicity, suggesting that Bmi-1 may play a crucial role in tumorigenesis.. Our finding indicates that glioma cells without self-renewal capability in standard conditions could also contribute to glioma malignancy when cytokines, such as IL-1β and TGF-β, are present in the tumor environment. Targeting GSC-promoting cytokines that are highly expressed in glioblastomas may contribute to the development of more effective glioma therapies.

    Topics: Animals; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Glioma; Humans; Interleukin-1beta; Intermediate Filament Proteins; Leukemia Inhibitory Factor; Mice; Mice, Nude; Neoplastic Stem Cells; Nerve Tissue Proteins; Nestin; Nuclear Proteins; Polycomb Repressive Complex 1; Proto-Oncogene Proteins; Receptor, Notch2; Repressor Proteins; Transforming Growth Factor beta

2012
USP15 stabilizes TGF-β receptor I and promotes oncogenesis through the activation of TGF-β signaling in glioblastoma.
    Nature medicine, 2012, Feb-19, Volume: 18, Issue:3

    In advanced cancer, including glioblastoma, the transforming growth factor β (TGF-β) pathway acts as an oncogenic factor and is considered to be a therapeutic target. Using a functional RNAi screen, we identified the deubiquitinating enzyme ubiquitin-specific peptidase 15 (USP15) as a key component of the TGF-β signaling pathway. USP15 binds to the SMAD7-SMAD specific E3 ubiquitin protein ligase 2 (SMURF2) complex and deubiquitinates and stabilizes type I TGF-β receptor (TβR-I), leading to an enhanced TGF-β signal. High expression of USP15 correlates with high TGF-β activity, and the USP15 gene is found amplified in glioblastoma, breast and ovarian cancer. USP15 amplification confers poor prognosis in individuals with glioblastoma. Downregulation or inhibition of USP15 in a patient-derived orthotopic mouse model of glioblastoma decreases TGF-β activity. Moreover, depletion of USP15 decreases the oncogenic capacity of patient-derived glioma-initiating cells due to the repression of TGF-β signaling. Our results show that USP15 regulates the TGF-β pathway and is a key factor in glioblastoma pathogenesis.

    Topics: Animals; Brain Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Endopeptidases; Gene Expression Regulation, Neoplastic; Glioblastoma; HEK293 Cells; Humans; Magnetic Resonance Imaging; Mice; Phosphorylation; Prognosis; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; RNA Interference; Signal Transduction; Smad2 Protein; Smad7 Protein; Transforming Growth Factor beta; Ubiquitin; Ubiquitin-Protein Ligases; Ubiquitin-Specific Proteases

2012
Attenuation of TGF-β signaling suppresses premature senescence in a p21-dependent manner and promotes oncogenic Ras-mediated metastatic transformation in human mammary epithelial cells.
    Molecular biology of the cell, 2012, Volume: 23, Issue:8

    The molecular mechanisms that drive triple-negative, basal-like breast cancer progression are elusive. Few molecular targets have been identified for the prevention or treatment of this disease. Here we developed a series of isogenic basal-like human mammary epithelial cells (HMECs) with altered transforming growth factor-β (TGF-β) sensitivity and different malignancy, resembling a full spectrum of basal-like breast carcinogenesis, and determined the molecular mechanisms that contribute to oncogene-induced transformation of basal-like HMECs when TGF-β signaling is attenuated. We found that expression of a dominant-negative type II receptor (DNRII) of TGF-β abrogated autocrine TGF-β signaling in telomerase-immortalized HMECs and suppressed H-Ras-V12-induced senescence-like growth arrest (SLGA). Furthermore, coexpression of DNRII and H-Ras-V12 rendered HMECs highly tumorigenic and metastatic in vivo in comparison with H-Ras-V12-transformed HMECs that spontaneously escaped H-Ras-V12-induced SLGA. Microarray analysis revealed that p21 was the major player mediating Ras-induced SLGA, and attenuated or loss of p21 expression contributed to the escape from SLGA when autocrine TGF-β signaling was blocked in HMECs. Furthermore, knockdown of p21 also suppressed H-Ras-V12-induced SLGA. Our results identify that autocrine TGF-β signaling is an integral part of the cellular anti-transformation network by suppressing the expression of a host of genes, including p21-regulated genes, that mediate oncogene-induced transformation in basal-like breast cancer.

    Topics: Animals; Breast Neoplasms; Cell Line; Cell Transformation, Neoplastic; Cellular Senescence; Epithelial Cells; ErbB Receptors; Female; Humans; Mammary Glands, Human; Mice; Mice, Nude; Oncogene Protein p21(ras); ras Proteins; Receptors, Estrogen; Receptors, Progesterone; RNA Interference; RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta

2012
CLIC4 is a tumor suppressor for cutaneous squamous cell cancer.
    Carcinogenesis, 2012, Volume: 33, Issue:5

    Chloride intracellular channel (CLIC) 4 is a member of a redox-regulated, metamorphic multifunctional protein family, first characterized as intracellular chloride channels. Current knowledge indicates that CLICs participate in signaling, cytoskeleton integrity and differentiation functions of multiple tissues. In metabolically stressed skin keratinocytes, cytoplasmic CLIC4 is S-nitrosylated and translocates to the nucleus where it enhances transforming growth factor-β (TGF-β) signaling by protecting phospho-Smad 2 and 3 from dephosphorylation. CLIC4 expression is diminished in multiple human epithelial cancers, and the protein is excluded from the nucleus. We now show that CLIC4 expression is reduced in chemically induced mouse skin papillomas, mouse and human squamous carcinomas and squamous cancer cell lines, and the protein is excluded from the nucleus. The extent of reduction in CLIC4 coincides with progression of squamous tumors from benign to malignant. Inhibiting antioxidant defense in tumor cells increases S-nitrosylation and nuclear translocation of CLIC4. Adenoviral-mediated reconstitution of nuclear CLIC4 in squamous cancer cells enhances TGF-β-dependent transcriptional activity and inhibits growth. Adenoviral targeting of CLIC4 to the nucleus of tumor cells in orthografts inhibits tumor growth, whereas elevation of CLIC4 in transgenic epidermis reduces de novo chemically induced skin tumor formation. In parallel, overexpression of exogenous CLIC4 in squamous tumor orthografts suppresses tumor growth and enhances TGF-β signaling. These results indicate that CLIC4 suppresses the growth of squamous cancers, that reduced CLIC4 expression and nuclear residence detected in cancer cells is associated with the altered redox state of tumor cells and the absence of detectable nuclear CLIC4 in cancers contributes to TGF-β resistance and enhances tumor development.

    Topics: Animals; Cell Line, Tumor; Cell Nucleus; Cell Transformation, Neoplastic; Chloride Channels; Fibroblasts; Humans; Keratinocytes; Male; Mice; Mice, Inbred BALB C; Mice, Inbred SENCAR; Mitochondrial Proteins; Neoplasms, Squamous Cell; Oxidation-Reduction; Papilloma; Protein Transport; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta; Tumor Suppressor Proteins

2012
22 : 6n-3 DHA inhibits differentiation of prostate fibroblasts into myofibroblasts and tumorigenesis.
    The British journal of nutrition, 2012, Dec-28, Volume: 108, Issue:12

    Prostate cancer is one of the most common malignancies in men. Epidemiological and experimental studies have revealed that stromal cells of the tumour microenvironment contribute to the development of prostate cancers, while long-chain n-3 PUFA-enriched diets reduce the risk of this tumour histotype. These findings prompted us to investigate whether DHA, an n-3 PUFA, may abrogate differentiation of prostate fibroblasts into myofibroblasts, the activated form of fibroblasts generally involved in prostate cancer progression. We used the human prostate carcinoma cell line (PC3) as a prostate adenocarcinoma model and found that DHA (1) inhibits α-smooth muscle actin (α-SMA) expression, a typical marker of myofibroblast differentiation, in prostate fibroblasts stimulated in vitro with transforming growth factor-β (TGF-β), (2) blocks the matrix metalloproteinase-2-dependent enhanced invasiveness of PC3 prostate adenocarcinoma cells migrated in a medium conditioned by TGF-β-stimulated prostate fibroblasts, (3) prevents epithelial-mesenchymal transition (EMT) and invasiveness of PC3 cells promoted by a medium conditioned by TGF-β-stimulated prostate fibroblasts, and (4) reduces the growth rate of tumours obtained in immunodeficient animals injected with PC3 cells plus TGF-β-stimulated prostate fibroblasts. Moreover, DHA was found to revert α-SMA expression and the invasiveness-promoting activity exerted in PC3 cells by tumoral-activated fibroblasts. Thus, DHA represents a suitable agent to inhibit prostate myofibroblast differentiation, invasiveness and EMT, two most important tumour-promoting activities involved in the progression of prostate cancer cells.

    Topics: Actins; Adenocarcinoma; Animals; Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Docosahexaenoic Acids; Epithelial-Mesenchymal Transition; Fatty Acids, Omega-3; Fibroblasts; Humans; Male; Mice; Mice, SCID; Myofibroblasts; Prostate; Prostatic Neoplasms; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2012
Coordinated expression of REG4 and aldehyde dehydrogenase 1 regulating tumourigenic capacity of diffuse-type gastric carcinoma-initiating cells is inhibited by TGF-β.
    The Journal of pathology, 2012, Volume: 228, Issue:3

    Aldehyde dehydrogenase 1 (ALDH1) has been shown to serve as a marker for cancer-initiating cells (CICs), but little is known about the regulation of the CIC functions of ALDH1+ cancer cells. We isolated ALDH1+ cells from human diffuse-type gastric carcinoma cells and characterized these cells using an Aldefluor assay. ALDH1+ cells constituted 5-8% of the human diffuse-type gastric carcinoma cells, OCUM-2MLN and HSC-39; were more tumourigenic than ALDH1- cells; and were able to self-renew and generate heterogeneous cell populations. Using gene expression microarray analyses, we identified REG4 (regenerating islet-derived family, member 4) as one of the genes up-regulated in ALDH1+ cells, and thus as a novel marker for ALDH1+ tumour cells. Induced expression of REG4 enhanced the colony-forming ability of OCUM-2MLN cells, while knockdown of REG4 inhibited the tumourigenic potential of ALDH1+ cells. We further found that TGF-β signalling reduces the expression of ALDH1 and REG4, and the size of the ALDH1+ cell population. In human diffuse-type gastric carcinoma tissues, the expression of ALDH1 and REG4 correlated with each other, as assessed by immunohistochemistry, and ALDH1 expression correlated inversely with Smad3 phosphorylation as a measure of TGF-β signalling. These findings illustrate that, in diffuse-type gastric carcinoma, REG4 is up-regulated in ALDH1+ CICs, and that the increased tumourigenic ability of ALDH1+ cells depends on REG4. Moreover, TGF-β down-regulates ALDH1 and REG4 expression, which correlates with a reduction in CIC population size and tumourigenicity. Targeting REG4 in ALDH1+ CICs may provide a novel strategy in the treatment of diffuse-type gastric carcinoma.

    Topics: Adenocarcinoma; Aldehyde Dehydrogenase 1 Family; Animals; Biomarkers, Tumor; Carcinoma, Signet Ring Cell; Cell Line, Tumor; Cell Transformation, Neoplastic; Cells, Cultured; Disease Models, Animal; Down-Regulation; Gene Expression Regulation, Neoplastic; Humans; Isoenzymes; Lectins, C-Type; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplastic Stem Cells; Pancreatitis-Associated Proteins; Retinal Dehydrogenase; Signal Transduction; Stomach Neoplasms; Transforming Growth Factor beta; Transplantation, Heterologous; Up-Regulation

2012
Activation of multiple cancer pathways and tumor maintenance function of the 3q amplified oncogene FNDC3B.
    Cell cycle (Georgetown, Tex.), 2012, May-01, Volume: 11, Issue:9

    FNDC3B was recently identified in an oncogenomic screen for amplified oncogenes in hepatocellular carcinoma. It is located at 3q26 and is amplified in over 20% of cancers, usually as part of a broad amplified region encompassing the entire 3q arm. Consistent with an oncogenic role in multiple cancer types, we show here that overexpression of FNDC3B is capable of malignantly transforming mammary and kidney epithelial cells in addition to hepatocytes. To explore how FNDC3B transforms cells, we determined the cellular localization of its gene product and the cancer pathways that it activates. We found that the FNDC3B oncoprotein localizes to the Golgi network, and that its correct localization is essential for its transforming function. We found that overexpression of FNDC3B induces the epithelial-to-mesenchymal transition (EMT) and activates several cancer pathways, including PI3-kinase/Akt, Rb1 and TGFβ signaling. For TGFβ signaling, we analyzed the point in the pathway at which FNDC3B operates and obtained evidence that it induces expression of all three TGFβ ligands and also promotes TGFBR1 cell-surface localization. We found that RNAi-mediated knockdown of FNDC3B in cancer cells with 3q amplification suppressed their clonogenicity and tumorigenicity, but that the same RNAi knockdown had no effect on single-copy 3q cancer cells. These results indicate that FNDC3B is an important oncogenic driver gene of the 3q amplicon, adding to the growing list of oncogenic drivers within this commonly amplified region.

    Topics: Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Transformation, Neoplastic; Chromosomes, Human, Pair 3; Epithelial-Mesenchymal Transition; Fibronectins; Gene Dosage; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Genetic Vectors; Golgi Apparatus; Humans; Immunoprecipitation; Oncogenes; Phosphatidylinositol 3-Kinases; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Retroviridae; RNA Interference; Signal Transduction; Transcriptional Activation; Transfection; Transforming Growth Factor beta

2012
Progenitor-like traits contribute to patient survival and prognosis in oligodendroglial tumors.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2012, Aug-01, Volume: 18, Issue:15

    Patient-derived glioma-propagating cells (GPC) contain karyotypic and gene expression profiles that are found in the primary tumor. However, their clinical relevance is unclear. We ask whether GPCs contribute to disease progression and survival outcome in patients with glioma by analyzing gene expression profiles.. We tapped into public sources of GPC gene expression data and derived a gene signature distinguishing oligodendroglial from glioblastoma multiforme (GBM) GPCs. By adapting a method in glioma biology, the Connectivity Map, we interrogated its strength of association in public clinical databases. We validated the top-ranking signaling pathways Wnt, Notch, and TGFβ, in GPCs and primary tumor specimens.. We observed that patients with better prognosis correlated with oligodendroglial GPC features and lower tumor grade, and this was independent of the current clinical indicator, 1p/19q status. Patients with better prognosis had proneural tumors whereas the poorly surviving cohort had mesenchymal tumors. In addition, oligodendroglial GPCs were more sensitive to Wnt and Notch inhibition whereas GBM GPCs responded to TGFβR1 inhibition.. We provide evidence that GPCs are clinically relevant. In addition, the more favorable prognosis of oligodendroglial tumors over GBM could be recapitulated transcriptomically at the GPC level, underscoring the relevance of this cellular model. Our gene signature detects molecular heterogeneity in oligodendroglial tumors that cannot be accounted for by the 1p/19q status alone, indicating that stem-like traits contribute to clinical status. Collectively, these data highlight the limitation of morphology-based histologic analyses in tumor classification, consequently impacting on treatment decisions.

    Topics: Animals; beta Catenin; Brain Neoplasms; Cell Transformation, Neoplastic; Chromosome Deletion; Chromosomes, Human, Pair 1; Chromosomes, Human, Pair 19; Gene Expression Profiling; Gene Knockdown Techniques; Humans; Immunoblotting; Kaplan-Meier Estimate; Mice; Mice, Inbred NOD; Mice, SCID; Neoplastic Stem Cells; Oligodendroglioma; Oligonucleotide Array Sequence Analysis; Prognosis; Receptors, Notch; Signal Transduction; Transforming Growth Factor beta; Transplantation, Heterologous; Tumor Cells, Cultured; Wnt Signaling Pathway

2012
Hepatitis C viral protein NS5A induces EMT and participates in oncogenic transformation of primary hepatocyte precursors.
    Journal of hepatology, 2012, Volume: 57, Issue:5

    Apicobasal polarity, which is essential for epithelial structure and function, is targeted by several tumour-related pathogens and is generally perturbed in the course of carcinogenesis. Hepatitis C virus (HCV) infection is associated with a strong risk of hepatocellular carcinoma, typically preceded by dysplastic alterations of cell morphology. We investigated the molecular mechanisms and the functional consequences of HCV-driven perturbations of epithelial polarity.. We used biochemical, genetic, and cell biology approaches to assess the impact of hepatitis C viral protein NS5A on the polarity and function of hepatocytes and hepatic progenitors. Transgenic animals and xenograft models served for in vivo validation of the results obtained in cell culture.. We found that expression of HCV-NS5A in primary hepatic precursors and in immortalized hepatocyte cell lines gave rise to profound modifications of cell polarity, leading to epithelial to mesenchymal transition (EMT). NS5A, either alone or in the context of the full complement of viral proteins in the course of infection, acted through activating Twist2, a transcriptional regulator of EMT. The effects of NS5A were additive to those of TGF-β, a cytokine abundant in diseased liver and highly relevant to HCV-related pathology. Moreover, NS5A cooperates with oncogenic Ras, giving rise to transformed, invasive cells that are highly tumorigenic in vivo.. Our data suggest that in the context of HCV infection, NS5A favors formation of preneoplastic lesions by disrupting cell polarity and additional oncogenic events cooperate with the viral protein to give rise to motile and invasive tumour cells.

    Topics: Animals; Animals, Genetically Modified; Cell Line; Cell Polarity; Cell Transformation, Neoplastic; Cells, Cultured; Epithelial-Mesenchymal Transition; Hepatitis C; Hepatocytes; Humans; Mice; Mice, Inbred BALB C; Mice, Nude; Proto-Oncogene Proteins p21(ras); Repressor Proteins; Risk Factors; Transforming Growth Factor beta; Transplantation, Heterologous; Twist-Related Protein 1; Viral Nonstructural Proteins

2012
SOX4 induces epithelial-mesenchymal transition and contributes to breast cancer progression.
    Cancer research, 2012, Sep-01, Volume: 72, Issue:17

    Epithelial-mesenchymal transition (EMT) is a developmental program, which is associated with breast cancer progression and metastasis. Here, we report that ectopic overexpression of SOX4 in immortalized human mammary epithelial cells is sufficient for acquisition of mesenchymal traits, enhanced cell migration, and invasion, along with epithelial stem cell properties defined by the presence of a CD44(high)/CD24(low) cell subpopulation. SOX4 positively regulated expression of known EMT inducers, also activating the TGF-β pathway to contribute to EMT. SOX4 itself was induced by TGF-β in mammary epithelial cells and was required for TGF-β-induced EMT. Murine xenograft experiments showed that SOX4 cooperated with oncogenic Ras to promote tumorigenesis in vivo. Finally, in clinical specimens of human breast cancer, we found that SOX4 was abnormally overexpressed and correlated with the triple-negative breast cancer subtype (ER(-)/PR(-)/HER2(-)). Our findings define an important function for SOX4 in the progression of breast cancer by orchestrating EMT, and they implicate this gene product as a marker of poor prognosis in this disease.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Disease Progression; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Gene Expression; Gene Expression Regulation, Neoplastic; Genes, ras; Humans; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Invasiveness; Neoplastic Stem Cells; Prognosis; SOXC Transcription Factors; Transforming Growth Factor beta

2012
Heat shock factor Hsf1 cooperates with ErbB2 (Her2/Neu) protein to promote mammary tumorigenesis and metastasis.
    The Journal of biological chemistry, 2012, Oct-12, Volume: 287, Issue:42

    ErbB2/Neu oncogene is overexpressed in 25% of invasive/metastatic breast cancers. We have found that deletion of heat shock factor Hsf1 in mice overexpressing ErbB2/Neu significantly reduces mammary tumorigenesis and metastasis. Hsf1(+/-)ErbB2/Neu(+) tumors exhibit reduced cellular proliferative and invasive properties associated with reduced activated ERK1/2 and reduced epithelial-mesenchymal transition (EMT). Hsf1(+/+)Neu(+) mammary epithelial cells exposed to TGFβ show high levels of ERK1/2 activity and EMT; this is associated with reduced expression of E-cadherin and increased expression of Slug and vimentin, a mesenchymal marker. In contrast, Hsf1(-/-)Neu(+) or Hsf1(+/+)Neu(+) cells do not exhibit activated ERK1/2 and show reduced EMT in the presence of TGFβ. The ineffective activation of the RAS/RAF/MEK/ERK1/2 signaling pathway in cells with reduced levels of HSF1 is due to the low levels of HSP90 in complex with RAF1 that are required for RAF1 stability and maturation. These results indicate a powerful inhibitory effect conferred by HSF1 downstream target genes in the inhibition of ErbB2-induced breast cancers in the absence of the Hsf1 gene.

    Topics: Animals; Cadherins; Cell Transformation, Neoplastic; DNA-Binding Proteins; Female; Gene Expression Regulation, Neoplastic; Heat Shock Transcription Factors; HSP90 Heat-Shock Proteins; Mammary Neoplasms, Animal; MAP Kinase Signaling System; Mice; Mice, Knockout; Mitogen-Activated Protein Kinase 3; Neoplasm Metastasis; Proto-Oncogene Proteins c-raf; Receptor, ErbB-2; Transcription Factors; Transforming Growth Factor beta

2012
Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: connecting TGF-β signaling with "Warburg-like" cancer metabolism and L-lactate production.
    Cell cycle (Georgetown, Tex.), 2012, Aug-15, Volume: 11, Issue:16

    We have previously shown that a loss of stromal Cav-1 is a biomarker of poor prognosis in breast cancers. Mechanistically, a loss of Cav-1 induces the metabolic reprogramming of stromal cells, with increased autophagy/mitophagy, mitochondrial dysfunction and aerobic glycolysis. As a consequence, Cav-1-low CAFs generate nutrients (such as L-lactate) and chemical building blocks that fuel mitochondrial metabolism and the anabolic growth of adjacent breast cancer cells. It is also known that a loss of Cav-1 is associated with hyperactive TGF-β signaling. However, it remains unknown whether hyperactivation of the TGF-β signaling pathway contributes to the metabolic reprogramming of Cav-1-low CAFs. To address these issues, we overexpressed TGF-β ligands and the TGF-β receptor I (TGFβ-RI) in stromal fibroblasts and breast cancer cells. Here, we show that the role of TGF-β in tumorigenesis is compartment-specific, and that TGF-β promotes tumorigenesis by shifting cancer-associated fibroblasts toward catabolic metabolism. Importantly, the tumor-promoting effects of TGF-β are independent of the cell type generating TGF-β. Thus, stromal-derived TGF-β activates signaling in stromal cells in an autocrine fashion, leading to fibroblast activation, as judged by increased expression of myofibroblast markers, and metabolic reprogramming, with a shift toward catabolic metabolism and oxidative stress. We also show that TGF-β-activated fibroblasts promote the mitochondrial activity of adjacent cancer cells, and in a xenograft model, enhancing the growth of breast cancer cells, independently of angiogenesis. Conversely, activation of the TGF-β pathway in cancer cells does not influence tumor growth, but cancer cell-derived-TGF-β ligands affect stromal cells in a paracrine fashion, leading to fibroblast activation and enhanced tumor growth. In conclusion, ligand-dependent or cell-autonomous activation of the TGF-β pathway in stromal cells induces their metabolic reprogramming, with increased oxidative stress, autophagy/mitophagy and glycolysis, and downregulation of Cav-1. These metabolic alterations can spread among neighboring fibroblasts and greatly sustain the growth of breast cancer cells. Our data provide novel insights into the role of the TGF-β pathway in breast tumorigenesis, and establish a clear causative link between the tumor-promoting effects of TGF-β signaling and the metabolic reprogramming of the tumor microenvironment.

    Topics: Animals; Autocrine Communication; Autophagy; Breast Neoplasms; Caveolin 1; Cell Line, Tumor; Cell Transformation, Neoplastic; Cellular Reprogramming; Coculture Techniques; Female; Fibroblasts; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Glycolysis; Humans; Immunohistochemistry; Lactic Acid; Ligands; Mice; Mice, Nude; Mitochondria; Neovascularization, Pathologic; Oxidative Phosphorylation; Oxidative Stress; Paracrine Communication; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Stromal Cells; Transforming Growth Factor beta; Tumor Microenvironment; Xenograft Model Antitumor Assays

2012
Biological responses to TGF-β in the mammary epithelium show a complex dependency on Smad3 gene dosage with important implications for tumor progression.
    Molecular cancer research : MCR, 2012, Volume: 10, Issue:10

    TGF-β plays a dual role in epithelial carcinogenesis with the potential to either suppress or promote tumor progression. We found that levels of Smad3 mRNA, a critical mediator of TGF-β signaling, are reduced by approximately 60% in human breast cancer. We therefore used conditionally immortalized mammary epithelial cells (IMEC) of differing Smad3 genotypes to quantitatively address the Smad3 requirement for different biologic responses to TGF-β. We found that a two-fold reduction in Smad3 gene dosage led to complex effects on TGF-β responses; the growth-inhibitory response was retained, the pro-apoptotic response was lost, the migratory response was reduced, and the invasion response was enhanced. Loss of the pro-apoptotic response in the Smad3(+/-) IMECs correlated with loss of Smad3 binding to the Bcl-2 locus, whereas retention of the growth-inhibitory response in Smad3 IMECs correlated with retention of Smad3 binding to the c-Myc locus. Addressing the integrated outcome of these changes in vivo, we showed that reduced Smad3 levels enhanced metastasis in two independent models of metastatic breast cancer. Our results suggest that different biologic responses to TGF-β in the mammary epithelium are differentially affected by Smad3 dosage and that a mere two-fold reduction in Smad3 is sufficient to promote metastasis.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Disease Progression; Enhancer Elements, Genetic; Epithelium; Female; Gene Dosage; Gene Expression Regulation, Neoplastic; Humans; Mammary Glands, Animal; Mammary Glands, Human; Mice; Neoplasm Metastasis; Protein Binding; Proto-Oncogene Proteins c-bcl-2; Proto-Oncogene Proteins c-myc; Smad3 Protein; Transforming Growth Factor beta

2012
Hepatic transforming growth factor beta gives rise to tumor-initiating cells and promotes liver cancer development.
    Hepatology (Baltimore, Md.), 2012, Volume: 56, Issue:6

    Liver cirrhosis is a predominant risk factor for hepatocellular carcinoma (HCC). However, the mechanism underlying the progression from cirrhosis to HCC remains unclear. Herein we report the concurrent increase of liver progenitor cells (LPCs) and transforming growth factor-β (TGF-β) in diethylnitrosamine (DEN)-induced rat hepatocarcinogenesis and cirrhotic livers of HCC patients. Using several experimental approaches, including 2-acetylaminofluorene/partial hepatectomy (2-AAF/PHx) and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-elicited murine liver regeneration, we found that activation of LPCs in the absence of TGF-β induction was insufficient to trigger hepatocarcinogenesis. Moreover, a small fraction of LPCs was detected to coexpress tumor initiating cell (T-IC) markers during rat hepatocarcinogenesis and in human HCCs, and TGF-β levels were positively correlated with T-IC marker expression, which indicates a role of TGF-β in T-IC generation. Rat pluripotent LPC-like WB-F344 cells were exposed to low doses of TGF-β for 18 weeks imitating the enhanced TGF-β expression in cirrhotic liver. Interestingly, long-term treatment of TGF-β on WB-F344 cells impaired their LPC potential but granted them T-IC properties including expression of T-IC markers, increased self-renewal capacity, stronger chemoresistance, and tumorigenicity in NOD-SCID mice. Hyperactivation of Akt but not Notch, signal transducer and activator of transcription 3 (STAT3), or mammalian target of rapamycin (mTOR) was detected in TGF-β-treated WB-F344 cells. Introduction of the dominant-negative mutant of Akt significantly attenuated T-IC properties of those transformed WB-F344 cells, indicating Akt was required in TGF-β-mediated-generation of hepatic T-ICs. We further demonstrate that TGF-β-induced Akt activation and LPC transformation was mediated by microRNA-216a-modulated phosphatase and tensin homolog deleted on chromosome 10 (PTEN) suppression.. Hepatoma-initiating cells may derive from hepatic progenitor cells exposed to chronic and constant TGF-β stimulation in cirrhotic liver, and pharmaceutical inhibition of microRNA-216a/PTEN/Akt signaling could be a novel strategy for HCC prevention and therapy targeting hepatic T-ICs.

    Topics: AC133 Antigen; Animals; Antigens, CD; Antigens, Differentiation; Antigens, Neoplasm; Biomarkers, Tumor; Cell Adhesion Molecules; Cell Transformation, Neoplastic; Diethylnitrosamine; Epithelial Cell Adhesion Molecule; Glycoproteins; Humans; Liver; Liver Cirrhosis; Liver Neoplasms, Experimental; Male; Mice; Mice, Inbred C57BL; Mice, Inbred NOD; Mice, SCID; MicroRNAs; Neoplastic Stem Cells; Peptides; Pluripotent Stem Cells; Proto-Oncogene Proteins c-akt; PTEN Phosphohydrolase; Rats; Rats, Wistar; STAT3 Transcription Factor; Thy-1 Antigens; TOR Serine-Threonine Kinases; Transforming Growth Factor beta

2012
Runx3 protects gastric epithelial cells against epithelial-mesenchymal transition-induced cellular plasticity and tumorigenicity.
    Stem cells (Dayton, Ohio), 2012, Volume: 30, Issue:10

    The transcription factor RUNX3 functions as a tumor suppressor in the gastrointestinal epithelium, where its loss is an early event in carcinogenesis. While RUNX3 acts concurrently as a mediator of TGF-β signaling and an antagonist of Wnt, the cellular changes that follow its loss and their contribution to tumorigenicity are not fully understood. Here, we report that the loss of Runx3 in gastric epithelial cells results in spontaneous epithelial-mesenchymal transition (EMT). This produces a tumorigenic stem cell-like subpopulation, which remarkably expresses the gastric stem cell marker Lgr5. This phenomenon is due to the compounding effects of the dysregulation of the TGF-β and Wnt pathways. Specifically, Runx3(-/-) p53(-/-) gastric epithelial cells were unexpectedly sensitized for TGF-β-induced EMT, during which the resultant induction of Lgr5 was enhanced by an aberrantly activated Wnt pathway. These data demonstrate a protective role for RUNX3 in safeguarding gastric epithelial cells against aberrant growth factor signaling and the resultant cellular plasticity and stemness.

    Topics: Animals; beta Catenin; Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Core Binding Factor Alpha 3 Subunit; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Gastrointestinal Neoplasms; Gastrointestinal Tract; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Humans; Mice; Neoplasm Transplantation; Neoplastic Stem Cells; Receptors, G-Protein-Coupled; Transfection; Transforming Growth Factor beta; Wnt Proteins; Wnt Signaling Pathway

2012
Dichotomy effects of Akt signaling in breast cancer.
    Molecular cancer, 2012, Aug-24, Volume: 11

    The oncogenic roles contributed by the Akt/PKB kinase family remain controversial and presumably depend on cell context, but are perceived to be modulated by an interplay and net balance between various isoforms. This study is intended to decipher whether distinct Akt kinase isoforms exert either redundant or unique functions in regulating neoplastic features of breast cancer cells, including epithelial-mesenchymal transition (EMT), cell motility, and stem/progenitor cell expansion.. We demonstrate that overactivation of Akt signaling in nonmalignant MCF10A cells and in primary cultures of normal human mammary epithelial tissue results in previously unreported inhibitory effects on EMT, cell motility and stem/progenitor cell expansion. Importantly, this effect is largely redundant and independent of Akt isoform types. However, using a series of isogenic cell lines derived from MCF-10A cells but exhibiting varying stages of progressive tumorigenesis, we observe that this inhibition of neoplastic behavior can be reversed in epithelial cells that have advanced to a highly malignant state. In contrast to the tumor suppressive properties of Akt, activated Akt signaling in MCF10A cells can rescue cell viability upon treatment with cytotoxic agents. This feature is regarded as tumor-promoting.. We demonstrate that Akt signaling conveys novel dichotomy effects in which its oncogenic properties contributes mainly to sustaining cell viability, as opposed to the its tumor suppressing effects, which are mediated by repressing EMT, cell motility, and stem/progenitor cell expansion. While the former exerts a tumor-enhancing effect, the latter merely acts as a safeguard by restraining epithelial cells at the primary sites until metastatic spread can be moved forward, a process that is presumably dictated by the permissive tumor microenvironment or additional oncogenic insults.

    Topics: Breast Neoplasms; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Drug Resistance, Neoplasm; Enzyme Activation; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Humans; Insulin-Like Growth Factor I; Protein Isoforms; Proto-Oncogene Proteins c-akt; Signal Transduction; Stem Cells; Transforming Growth Factor beta

2012
IL-17A in the tumor microenvironment of the human colorectal adenoma-carcinoma sequence.
    Scandinavian journal of gastroenterology, 2012, Volume: 47, Issue:11

    Interleukin (IL)-17A is an important pro-inflammatory cytokine and involved in the colorectal carcinogenesis. In this study, the authors evaluated the dynamic change of IL-17A expression in the tumor microenvironment throughout the colorectal adenoma-carcinoma sequence.. Using quantitative real-time PCR (polymerase chain reaction) and semi-quantitative immunohistochemistry, the authors examined the expression level of IL-17A in 50 of human colorectal adenoma tissues, 50 of colorectal cancer (CRC) tissues and 15 controls. The relationship between IL-17A expression and clinicopathological parameters throughout the sequence was also evaluated.. The results revealed a step-up increased IL-17A mRNA level throughout the colorectal adenoma-carcinoma sequence, which began to increase in the adenomas and became even higher in the CRCs; notably, the increase of IL-17A mRNA level in the adenomatous tissues was associated with the severity of dysplasia. Immunohistochemical analysis confirmed the real-time PCR results and revealed gradually increasing IL-17A cells in both the stroma and adenomatous/cancerous epithelium. In addition, the quantitative real-time PCR result has also revealed an increased expression of TH17-stimulating factors throughout the sequence.. IL-17A and TH17 are highly activated throughout the colorectal adenoma-carcinoma sequence.

    Topics: Adenoma; Adult; Aged; Aged, 80 and over; Analysis of Variance; Carcinoma; Case-Control Studies; Cell Transformation, Neoplastic; Colorectal Neoplasms; Female; Humans; Immunohistochemistry; Interleukin-17; Interleukin-1beta; Interleukin-23; Interleukin-6; Male; Middle Aged; Real-Time Polymerase Chain Reaction; RNA, Messenger; Statistics, Nonparametric; Th17 Cells; Transforming Growth Factor beta

2012
Loss of SNAIL regulated miR-128-2 on chromosome 3p22.3 targets multiple stem cell factors to promote transformation of mammary epithelial cells.
    Cancer research, 2012, Nov-15, Volume: 72, Issue:22

    A discontinuous pattern of LOH at chromosome 3p has been reported in 87% of primary breast cancers. Despite the identification of several tumor suppressor genes in this region, there has yet to be a detailed analysis of noncoding RNAs including miRNAs in this region. In this study, we identified 16 aberrant miRNAs in this region and determined several that are frequently lost or amplified in breast cancer. miR-128-2 was the most commonly deleted miRNA. Embedded in the intron of the ARPP21 gene at chromosome 3p22.3, miR-128-2 was frequently downregulated along with ARPP21 in breast cancer, where it was negatively associated with clinicopathologic characteristics and survival outcome. Forced expression of miR-128 impeded several oncogenic traits of mammary carcinoma cells, whereas depleting miR-128-2 expression was sufficient for oncogenic transformation and stem cell-like behaviors in immortalized nontumorigenic mammary epithelial cells, both in vitro and in vivo. miR-128-2 silencing enabled transforming capacity partly by derepressing a cohort of direct targets (BMI1, CSF1, KLF4, LIN28A, NANOG, and SNAIL), which together acted to stimulate the PI3K/AKT and STAT3 signaling pathways. We also found that miR-128-2 was directly downregulated by SNAIL and repressed by TGF-β signaling, adding 2 additional negative feedback loops to this network. In summary, we have identified a novel TGF-β/SNAIL/miR-128 axis that provides a new avenue to understand the basis for oncogenic transformation of mammary epithelial cells.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Chromosome Aberrations; Chromosomes, Human, Pair 3; Down-Regulation; Female; Humans; Kruppel-Like Factor 4; Loss of Heterozygosity; Mice; Mice, Nude; MicroRNAs; Neoplasm Invasiveness; Neoplastic Stem Cells; Oncogene Protein v-akt; Snail Family Transcription Factors; STAT3 Transcription Factor; Stem Cell Factor; Transcription Factors; Transforming Growth Factor beta; Transplantation, Heterologous

2012
Oncogenic Smad3 signaling induced by chronic inflammation is an early event in ulcerative colitis-associated carcinogenesis.
    Inflammatory bowel diseases, 2011, Volume: 17, Issue:3

    Both chronic inflammation and somatic mutations likely contribute to the pathogenesis of ulcerative colitis (UC)-associated dysplasia and cancer. On the other hand, both tumor suppression and oncogenesis can result from transforming growth factor (TGF)-β signaling. TGF-β type I receptor (TβRI) and Ras-associated kinases differentially phosphorylate a mediator, Smad3, to become C-terminally phosphorylated Smad3 (pSmad3C), linker phosphorylated Smad3 (pSmad3L), and both C-terminally and linker phosphorylated Smad3 (pSmad3L/C). The pSmad3C/p21(WAF1) pathway transmits a cytostatic TGF-β signal, while pSmad3L and pSmad3L/C promote cell proliferation by upregulating c-Myc oncoprotein. The purpose of this study was to clarify the alteration of Smad3 signaling during UC-associated carcinogenesis.. By immunostaining and immunofluorescence, we compared pSmad3C-, pSmad3L-, and pSmad3L/C-mediated signaling in colorectal specimens representing colitis, dysplasia, or cancer from eight UC patients with signaling in normal colonic crypts. We also investigated p53 expression and mutations of p53 and K-ras genes. We further sought functional meaning of the phosphorylated Smad3-mediated signaling in vitro.. As enterocytes in normal crypts migrated upward toward the lumen, cytostatic pSmad3C/p21(WAF1) tended to increase, while pSmad3L/c-Myc shown by progenitor cells gradually decreased. Colitis specimens showed prominence of pSmad3L/C/c-Myc, mediated by TGF-β and tumor necrosis factor (TNF)-α, in all enterocyte nuclei throughout entire crypts. In proportion with increases in frequency of p53 and K-ras mutations during progression from dysplasia to cancer, the oncogenic pSmad3L/c-Myc pathway came to be dominant with suppression of the pSmad3C/p21(WAF1) pathway.. Oncogenic Smad3 signaling, altered by chronic inflammation and eventually somatic mutations, promotes UC-associated neoplastic progression by upregulating growth-related protein.

    Topics: Adenocarcinoma; Adult; Blotting, Western; Cell Transformation, Neoplastic; Chronic Disease; Colitis, Ulcerative; Colorectal Neoplasms; Disease Progression; Female; Fluorescent Antibody Technique; Humans; Immunoenzyme Techniques; Immunoprecipitation; Male; Middle Aged; Phosphorylation; Polymerase Chain Reaction; Polymorphism, Restriction Fragment Length; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad3 Protein; Transforming Growth Factor beta

2011
Involvement of c-Ski oncoprotein in carcinogenesis of cholangiocacinoma induced by Opisthorchis viverrini and N-nitrosodimethylamine.
    Pathology oncology research : POR, 2011, Volume: 17, Issue:2

    Opisthorchiasis is the major public health problem in the endemic areas of Thailand and Laos because Opisthorchis viverrini infection causes serious hepatobiliary diseases including CCA. The molecular mechanism of the CCA carcinogenesis induced by the infection remains obscure. To reveal the potential genes and signaling pathways to involve in the carcinogenesis, the present study investigated the expression of c-Ski, an oncogene, and two TGF-β signaling pathway relative genes, TGF-β and Smad4, during the development of CCA induced by O. viverrini infection in hamster model, and in human opisthorchiasis associated CCA. The results showed that the expression of c-Ski gene was greatly up-regulated during the carcinogenesis of CCA in hamster model. The overexpression of c-Ski was confirmed by immunohistological staining result which showed the increased expression of c-Ski protein in cytoplasm of the epithelial lining of hepatic bile ducts. Moreover, the immunohistological staining of the specimens of human opisthorchiasis associated CCA revealed the up-regulated expression of c-Ski and Smad4 proteins in the cytoplasm of the epithelial lining of hepatic bile ducts and stomal fibrosis respectively. The expression of TGF-β and Smad4 were up-regulated, which expression kinetics was time-dependent of CCA development. These results suggest that c-Ski is likely involved in the carcinogenesis of CCA induced by O. viverrini infection through regulating TGF-β signaling pathway.

    Topics: Animals; Bile Duct Neoplasms; Bile Ducts, Intrahepatic; Carcinogens; Cell Transformation, Neoplastic; Cholangiocarcinoma; Cricetinae; Dimethylnitrosamine; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Humans; Immunohistochemistry; Male; Mesocricetus; Opisthorchiasis; Opisthorchis; Proto-Oncogene Proteins; Reverse Transcriptase Polymerase Chain Reaction; Smad4 Protein; Transforming Growth Factor beta

2011
Smad4 inactivation promotes malignancy and drug resistance of colon cancer.
    Cancer research, 2011, Feb-01, Volume: 71, Issue:3

    SMAD4 is localized to chromosome 18q21, a frequent site for loss of heterozygosity in advanced stage colon cancers. Although Smad4 is regarded as a signaling mediator of the TGFβ signaling pathway, its role as a major suppressor of colorectal cancer progression and the molecular events underlying this phenomenon remain elusive. Here, we describe the establishment and use of colon cancer cell line model systems to dissect the functional roles of TGFβ and Smad4 inactivation in the manifestation of a malignant phenotype. We found that loss of function of Smad4 and retention of intact TGFβ receptors could synergistically increase the levels of VEGF, a major proangiogenic factor. Pharmacologic inhibition studies suggest that overactivation of the TGFβ-induced MEK-Erk and p38-MAPK (mitogen-activated protein kinase) auxiliary pathways are involved in the induction of VEGF expression in SMAD4 null cells. Overall, SMAD4 deficiency was responsible for the enhanced migration of colon cancer cells with a corresponding increase in matrix metalloprotease 9 enhanced hypoxia-induced GLUT1 expression, increased aerobic glycolysis, and resistance to 5'-fluoruracil-mediated apoptosis. Interestingly, Smad4 specifically interacts with hypoxia-inducible factor (HIF) 1α under hypoxic conditions providing a molecular basis for the differential regulation of target genes to suppress a malignant phenotype. In summary, our results define a molecular mechanism that explains how loss of the tumor suppressor Smad4 promotes colorectal cancer progression. These findings are also consistent with targeting TGFβ-induced auxiliary pathways, such as MEK-ERK, and p38-MAPK and the glycolytic cascade, in SMAD4-deficient tumors as attractive strategies for therapeutic intervention.

    Topics: Cell Hypoxia; Cell Movement; Cell Transformation, Neoplastic; Colonic Neoplasms; Drug Resistance, Neoplasm; Fluorouracil; Gene Expression Regulation, Neoplastic; Gene Silencing; HCT116 Cells; Humans; Matrix Metalloproteinase 9; Neoplasm Metastasis; Neovascularization, Pathologic; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Smad4 Protein; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2011
Altered TGF-β signaling in a subpopulation of human stromal cells promotes prostatic carcinogenesis.
    Cancer research, 2011, Feb-15, Volume: 71, Issue:4

    Carcinoma-associated fibroblasts (CAF) play a critical role in malignant progression. Loss of TGF-β receptor II (TGFβR2) in the prostate stroma is correlated with prostatic tumorigenesis. To determine the mechanisms by which stromal heterogeneity because of loss of TGFβR2 might contribute to cancer progression, we attenuated transforming growth factor beta (TGF-β) signaling in a subpopulation of immortalized human prostate fibroblasts in a model of tumor progression. In a tissue recombination model, loss of TGFβR2 function in 50% of the stromal cell population resulted in malignant transformation of the nontumorigenic human prostate epithelial cell line BPH1. Mixing fibroblasts expressing the empty vector and dominant negative TGFβR2 increased the expression of markers of myofibroblast differentiation [coexpression of vimentin and alpha smooth muscle actin (αSMA)] through elevation of TGF-β1 and activation of the Akt pathway. In combination, these two populations of stromal cells recapitulated the tumor inductive activity of CAFs. TGFβR2 activity in mixed stromal cell populations cultured in vitro caused secretion of factors that are known to promote tumor progression, including TGF-β1, SDF1/CXCL12, and members of the fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) families. In vivo, tissue recombination of fibroblasts overexpressing TGF-β1 and SDF1/CXCL12 not only induced transformation of BPH1 cells, but also promoted a robust growth of highly invasive cells, similar to effects produced by CAFs. While the precise nature and/or origin of the particular stromal cell populations in vivo remain unknown, these findings strongly link heterogeneity in TGF-β signaling to tumor promotion by tumor stromal cells.

    Topics: Animals; Carcinoma; Cell Transformation, Neoplastic; Cells, Cultured; Disease Progression; Embryo, Mammalian; Gene Expression Regulation, Neoplastic; Humans; Male; Mice; Mice, Inbred C57BL; Mice, SCID; Prostatic Neoplasms; Rats; Rats, Sprague-Dawley; Signal Transduction; Stromal Cells; Transforming Growth Factor beta

2011
Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth.
    Cancer cell, 2011, Feb-15, Volume: 19, Issue:2

    Carcinoma-associated fibroblasts (CAFs) that express α-smooth muscle actin (αSMA) contribute to cancer progression, but their precise origin and role are unclear. Using mouse models of inflammation-induced gastric cancer, we show that at least 20% of CAFs originate from bone marrow (BM) and derive from mesenchymal stem cells (MSCs). αSMA+ myofibroblasts (MFs) are niche cells normally present in BM and increase markedly during cancer progression. MSC-derived CAFs that are recruited to the dysplastic stomach express IL-6, Wnt5α and BMP4, show DNA hypomethylation, and promote tumor growth. Moreover, CAFs are generated from MSCs and are recruited to the tumor in a TGF-β- and SDF-1α-dependent manner. Therefore, carcinogenesis involves expansion and relocation of BM-niche cells to the tumor to create a niche to sustain cancer progression.

    Topics: Actins; Animals; Bone Marrow Cells; Cell Division; Cell Transformation, Neoplastic; Disease Models, Animal; Intercellular Signaling Peptides and Proteins; Mesenchymal Stem Cells; Mice; Neoplasms, Experimental; Receptors, CXCR4; Stomach Neoplasms; Transforming Growth Factor beta; Up-Regulation

2011
Activation of the TGF-β/Smad signaling pathway in oncogenic transformation by v-Rel.
    Virology, 2011, Apr-25, Volume: 413, Issue:1

    v-rel, encoded by the avian reticuloendotheliosis virus, is an acutely transforming member of the Rel/NF-κB family of transcription factors. Transformation by v-Rel is mediated by the aberrant expression of genes that are normally regulated by Rel/NF-κB. Here, we demonstrate activation of the TGF-β/Smad signaling pathway in Rel transformation. RNA and protein levels of key TGF-β and Smad family members (TGF-β2, -β3, TGF-β type II receptor, and Smad3) are upregulated in v-Rel transformed cells with little to no change in c-Rel-expressing cells. Treatment of v-Rel transformed lymphoid cells with kinase inhibitors of the TGF-β receptor dramatically reduces soft agar colony formation whereas addition of TGF-β2 further promotes transformation. Moreover, Smad3 but not Smad2, is selectively activated as the downstream mediator of TGF-β signaling. Blocking Smad3 expression or activity inhibits the oncogenic potential of v-Rel. Overall, TGF-β/Smad signaling is activated at multiple levels and is required for the transforming ability of v-Rel.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Chickens; Gene Expression Regulation; Oncogene Proteins v-rel; Receptors, Transforming Growth Factor beta; Reticuloendotheliosis Viruses, Avian; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2011
Smad4 binds Hoxa9 in the cytoplasm and protects primitive hematopoietic cells against nuclear activation by Hoxa9 and leukemia transformation.
    Blood, 2011, Jun-02, Volume: 117, Issue:22

    We studied leukemic stem cells (LSCs) in a Smad4(-/-) mouse model of acute myelogenous leukemia (AML) induced either by the HOXA9 gene or by the fusion oncogene NUP98-HOXA9. Although Hoxa9-Smad4 complexes accumulate in the cytoplasm of normal hematopoietic stem cells and progenitor cells (HSPCs) transduced with these oncogenes, there is no cytoplasmic stabilization of HOXA9 in Smad4(-/-) HSPCs, and as a consequence increased levels of Hoxa9 is observed in the nucleus leading to increased immortalization in vitro. Loss of Smad4 accelerates the development of leukemia in vivo because of an increase in transformation of HSPCs. Therefore, the cytoplasmic binding of Hoxa9 by Smad4 is a mechanism to protect Hoxa9-induced transformation of normal HSPCs. Because Smad4 is a potent tumor suppressor involved in growth control, we developed a strategy to modify the subcellular distribution of Smad4. We successfully disrupted the interaction between Hoxa9 and Smad4 to activate the TGF-β pathway and apoptosis, leading to a loss of LSCs. Together, these findings reveal a major role for Smad4 in the negative regulation of leukemia initiation and maintenance induced by HOXA9/NUP98-HOXA9 and provide strong evidence that antagonizing Smad4 stabilization by these oncoproteins might be a promising novel therapeutic approach in leukemia.

    Topics: Animals; Apoptosis; Blotting, Western; Bone Marrow Transplantation; Cell Nucleus; Cell Transformation, Neoplastic; Chromatin Immunoprecipitation; Cytoplasm; Flow Cytometry; Hematopoietic Stem Cells; Homeodomain Proteins; Humans; Immunoprecipitation; Leukemia, Myeloid, Acute; Mice; Mice, Inbred C57BL; Mice, Knockout; Nuclear Pore Complex Proteins; Oncogene Proteins, Fusion; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Smad4 Protein; Transforming Growth Factor beta

2011
HAb18G/CD147 promotes epithelial-mesenchymal transition through TGF-β signaling and is transcriptionally regulated by Slug.
    Oncogene, 2011, Oct-27, Volume: 30, Issue:43

    Epithelial-mesenchymal transition (EMT) induced by transforming growth factor-β (TGF-β) is implicated in hepatocarcinogenesis and hepatocellular carcinoma (HCC) metastasis. HAb18G/CD147, which belongs to the CD147 family, is an HCC-associated antigen that has a crucial role in tumor invasion and metastasis. The goal of this study was to investigate the role of HAb18G/CD147 during EMT in hepatocarcinogenesis. Human normal hepatic cell lines QZG and L02, primary mouse hepatocytes and nude mouse models were used to determine the role of HAb18G/CD147 in EMT, and the involvement of the TGF-β-driven pathway. A dual-luciferase reporter assay and ChIP were used to investigate the transcriptional regulation of the CD147 gene. Samples from patients with liver disease were assessed to determine the relationship between HAb18G/CD147 and typical markers for EMT. Our results show that upregulation of HAb18G/CD147 is induced by TGF-β coupled with downregulation of E-cadherin and upregulation of N-cadherin and vimentin. The expression of HAb18G/CD147 is controlled by the cell survival PI3K/Akt/GSK3β signaling pathway, and is directly regulated by the transcription factor Slug. Transfection of CD147 also induces an elevated expression of TGF-β. CD147-transfected hepatocytes have mesenchymal phenotypes that accelerate tumor formation and tumor metastasis in vivo. Immunohistochemistry analysis shows a negative correlation between HAb18G/CD147 and E-cadherin expression (r(s)=-0.3622, P=0.0105), and a positive correlation between HAb18G/CD147 and Slug expression (r(s)=0.3064, P=0.0323) in human HCC tissues. Our study uncovers a novel role of HAb18G/CD147 in mediating EMT in the process of HCC progression and showed that CD147 is a Slug target gene in the signaling cascade TGF-β→PI3K/Akt→GSK3β→Snail→Slug→CD147. Our results suggest that CD147 may be a potential target for the treatment and prevention of HCC.

    Topics: Animals; Basigin; Cadherins; Carcinoma, Hepatocellular; Cell Line; Cell Transformation, Neoplastic; Disease Progression; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Liver Neoplasms; Mice; Mice, Nude; Signal Transduction; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2011
Gene expression following exposure to celecoxib in humans: pathways of inflammation and carcinogenesis are activated in tumors but not normal tissues.
    Digestion, 2011, Volume: 84, Issue:3

    The Cox-2 inhibitor, celecoxib (Pfizer Inc., N.Y., USA), is a promising chemopreventive agent [Arber et al.: N Engl J Med 2006;355:885-895; Bertagnolli et al.: N Engl J Med 2006;355:873-884]. This study aims to explore its mechanism by defining changes in gene expression between neoplastic and normal tissue samples before and after treatment.. Patients with documented colorectal neoplasia in screening colonoscopy, destined to undergo surgical colectomy, were randomized for treatment with celecoxib (n = 11; 400 mg/day) or placebo (n = 3) for 30 days. Tissue samples were taken from the tumor and from normal adjacent mucosa during both colonoscopy and surgery. RNA was extracted and analyzed using Affymetrix Genechip®.. 687 genes differentiated tumor samples before and after treatment, among which 310 genes did not show the same differential expression in the placebo group or normal samples. These genes were significantly related to pathways of cell cycle regulation and inflammation, and of note was the TGF-β pathway, which held a strong association with the list of genes formerly found to be associated with the colorectal cancer expression profile in microarray analyses, as summarized in a meta-analysis by Cardoso et al. [Biochim Biophys Acta 2007;1775:103-137].. Celecoxib selectively affects genes and pathways involved in inflammation and malignant transformation in tumor but not normal tissues, this may assist in the development of safer and more effective chemopreventive agents.

    Topics: Adenocarcinoma; Celecoxib; Cell Cycle; Cell Transformation, Neoplastic; Colonic Polyps; Colorectal Neoplasms; Cyclooxygenase 2 Inhibitors; Gene Expression; Humans; Inflammation; Intestinal Mucosa; Microarray Analysis; Pyrazoles; Random Allocation; RNA, Neoplasm; Signal Transduction; Sulfonamides; Transforming Growth Factor beta

2011
TGF-beta signaling engages an ATM-CHK2-p53-independent RAS-induced senescence and prevents malignant transformation in human mammary epithelial cells.
    Proceedings of the National Academy of Sciences of the United States of America, 2011, May-24, Volume: 108, Issue:21

    Oncogene-induced senescence (OIS), the proliferative arrest engaged in response to persistent oncogene activation, serves as an important tumor-suppressive barrier. We show here that finite lifespan human mammary epithelial cells (HMEC) undergo a p16/RB- and p53-independent OIS in response to oncogenic RAS that requires TGF-β signaling. Suppression of TGF-β signaling by expression of a dominant-negative TGF-β type II receptor, use of a TGF-β type I receptor inhibitor, or ectopic expression of MYC permitted continued proliferation upon RAS expression. Surprisingly, unlike fibroblasts, shRNA-mediated knockdown of ATM or CHK2 was unable to prevent RAS-mediated OIS, arguing that the DNA damage response is not required for OIS in HMEC. Abrogation of TGF-β signaling not only allowed HMEC lacking p53 to tolerate oncogenic RAS but also conferred the capacity for anchorage-independent growth. Thus, the OIS engaged after dysregulated RAS expression provides an early barrier to malignant progression and is mediated by TGF-β receptor activation in HMEC. Understanding the mechanisms that initiate and maintain OIS in epithelial cells may provide a foundation for future therapies aimed at reengaging this proliferative barrier as a cancer therapy.

    Topics: Ataxia Telangiectasia Mutated Proteins; Cell Cycle Proteins; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Cellular Senescence; Checkpoint Kinase 2; DNA-Binding Proteins; Epithelial Cells; Humans; Mammary Glands, Human; Protein Serine-Threonine Kinases; ras Proteins; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Tumor Suppressor Proteins

2011
EW-7203, a novel small molecule inhibitor of transforming growth factor-β (TGF-β) type I receptor/activin receptor-like kinase-5, blocks TGF-β1-mediated epithelial-to-mesenchymal transition in mammary epithelial cells.
    Cancer science, 2011, Volume: 102, Issue:10

    Recently, small molecule inhibitors of transforming growth factorβ (TGF-β) type I receptor kinase ⁄ activin receptor-like kinase-5 (ALK5) have been developed to target TGF-β signalling as a therapeutic strategy for combating cancer. In the present study, the authors examined a novel small molecule inhibitor of ALK5, 3-((5- ([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)thiazol-2-ylamino)methyl)benzonitrile (EW-7203) in breast cancer cells to determine if it has potential for cancer treatment. The inhibitory effects of EW-7203 on TGF-β-induced Smad signalling and epithelial- to-mesenchymal transition (EMT) were investigated in mammary epithelial cells using luciferase reporter assays, immunoblotting, confocal microscopy and wound healing assays. In addition, the suppressive effects of EW-7203 on mammary cancer metastasis to the lung were examined using a Balb ⁄ c xenograft model system. The novel ALK5 inhibitor, EW-7203, inhibited the TGF-β1-stimulated transcriptional activation of p3TP-Lux and pCA-GA₁₂- Luc. In addition, EW-7203 decreased phosphorylated Smad2 levels and the nuclear translocation of Smad2 was increased by TGF-β1. In addition, EW-7203 inhibited TGF-β1-induced EMT and wound healing of NMuMG cells. Furthermore, in xenografted Balb ⁄ c mice, EW-7203 inhibited metastasis to the lung from breast tumors. The novel ALK5 inhibitor, EW-7203, efficiently inhibited TGF-β1-induced Smad signalling, EMT and breast tumor metastasis to the lung in vivo, demonstrating that EW-7203 has therapeutic potential for breast cancer metastasis to the lung.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Humans; Lung Neoplasms; Mammary Glands, Animal; Mammary Glands, Human; Mice; Mice, Inbred BALB C; Neoplasm Metastasis; Protein Serine-Threonine Kinases; Pyridines; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Thiazoles; Transforming Growth Factor beta; Xenograft Model Antitumor Assays

2011
Molecular signatures and prognosis of hepatocellular carcinoma.
    Minerva gastroenterologica e dietologica, 2011, Volume: 57, Issue:3

    Prognostic prediction is a vital component in clinical management of hepatocellular carcinoma (HCC) patients. Rapidly evolving genomic assays serve as flexible sources to discover molecular signatures that sensitively and specifically predict clinical outcome of the patients. Studies have identified molecular signatures of HCC tumors that depict biological aggressiveness yielded through activation of specific genes and molecular pathways such as MYC, AKT, TGF-beta, and IGF2 as well as inactivation of TP53 pathway. Despite the promise for tumor-derived signatures' role in therapeutic target discovery, their value as prognostic marker seems to be limited especially in early-stage HCC, which has been increasing as a result of successful implementation HCC surveillance for patients with liver cirrhosis. In contrast, non-tumor, diseased liver tissues turn out to be a rich source of molecular information to capture propensity to hepatocarcinogenesis and metastasis through dyregulation of growth signaling and inflammatory/oxidative stress/immune response. In addition, the liver-derived signatures hold prognostic relevance irrespective of HCC stage, suggesting their role as prognostic markers. Furthermore, they may also be utilized for development of HCC chemoprevention therapy.

    Topics: Biomarkers, Tumor; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Gene Expression Regulation; Genes, myc; Genes, p53; Humans; Insulin-Like Growth Factor II; Liver Neoplasms; Predictive Value of Tests; Prognosis; Proto-Oncogene Proteins c-akt; Sensitivity and Specificity; Signal Transduction; Transforming Growth Factor beta

2011
Genotoxic stress-mediated cell cycle activities for the decision of cellular fate.
    Cell cycle (Georgetown, Tex.), 2011, Oct-01, Volume: 10, Issue:19

    Genomic integrity maintenance is critical for prevention of a wide variety of adverse cellular effects including apoptosis, cellular senescence, and malignant cell transformation. Coupled with normal replication, the local intracellular and extracellular stresses cause damage to cellular DNA that is recognized and repaired by the DNA damage response (DDR) pathway. p53 induces the transcription of genes that negatively regulate progression of the cell cycle in response to DNA damage, and thus participates in maintaining genome stability. p53 and many other anti-proliferative factors such as TGF beta regulate the expression of different cyclin-dependent kinase inhibitors (CDKIs). Paradoxically, one of the cellular proliferative factors, c-Myc proto-oncogene also controls the expression of these CDKIs and modulates the fate of cell in response to DNA damage. Furthermore, involvement of numerous other proteins in the DDR and crosstalk between them are likely to substantiate the DDR as one of the genome's most extensive signaling networks. Versatile protein kinases in this network affect the decision about four basic cellular fates, which are quiescence, apoptosis, oncogenesis and senescence, in response to DNA damage.

    Topics: Apoptosis; Cell Cycle Checkpoints; Cell Cycle Proteins; Cell Transformation, Neoplastic; Cellular Senescence; Cyclin-Dependent Kinase Inhibitor Proteins; DNA Damage; DNA Repair; Genomic Instability; Humans; JNK Mitogen-Activated Protein Kinases; Proto-Oncogene Mas; Proto-Oncogene Proteins c-akt; Proto-Oncogene Proteins c-myc; Signal Transduction; Stress, Physiological; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2011
Snail1 induces epithelial-to-mesenchymal transition and tumor initiating stem cell characteristics.
    BMC cancer, 2011, Sep-19, Volume: 11

    Tumor initiating stem-like cells (TISCs) are a subset of neoplastic cells that possess distinct survival mechanisms and self-renewal characteristics crucial for tumor maintenance and propagation. The induction of epithelial-mesenchymal-transition (EMT) by TGFβ has been recently linked to the acquisition of TISC characteristics in breast cancer. In HCC, a TISC and EMT phenotype correlates with a worse prognosis. In this work, our aim is to elucidate the underlying mechanism by which cells acquire tumor initiating characteristics after EMT.. Gene and protein expression assays and Nanog-promoter luciferase reporter were utilized in epithelial and mesenchymal phenotype liver cancer cell lines. EMT was analyzed with migration/invasion assays. TISC characteristics were analyzed with tumor-sphere self-renewal and chemotherapy resistance assays. In vivo tumor assay was performed to investigate the role of Snail1 in tumor initiation.. TGFβ induced EMT in epithelial cells through the up-regulation of Snail1 in Smad-dependent signaling. Mesenchymal liver cancer post-EMT demonstrates TISC characteristics such as tumor-sphere formation but are not resistant to cytotoxic therapy. The inhibition of Snail1 in mesenchymal cells results in decreased Nanog promoter luciferase activity and loss of self-renewal characteristics in vitro. These changes confirm the direct role of Snail1 in some TISC traits. In vivo, the down-regulation of Snail1 reduced tumor growth but was not sufficient to eliminate tumor initiation. In summary, TGFβ induces EMT and TISC characteristics through Snail1 and Nanog up-regulation. In mesenchymal cells post-EMT, Snail1 directly regulates Nanog expression, and loss of Snail1 regulates tumor growth without affecting tumor initiation.

    Topics: Animals; Antineoplastic Agents; Carcinoma, Hepatocellular; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Homeodomain Proteins; Humans; Liver Neoplasms; Mice; Nanog Homeobox Protein; Neoplastic Stem Cells; Promoter Regions, Genetic; Signal Transduction; Smad Proteins; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta

2011
Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast cancer recurrence.
    Molecular cancer research : MCR, 2011, Volume: 9, Issue:12

    Epithelial-mesenchymal transition (EMT) is a normal developmental program that is considered to also play an important role in cancer metastasis. Ultimate inducers of EMT are transcriptional repressors that individually can induce experimental EMT, yet in many cells, particularly cancer cells, multiple inducers are expressed simultaneously. Why, and if, and how they interact to regulate EMT is unanswered. Using RNA interference technology to affect protein knockdown and avoid potential overexpression artifact coupled with transient TGFβ treatment to better mimic in vivo conditions we show, in both nontumorigenic and tumorigenic epithelial cancer cells, that Snail1 is uniquely required for EMT initiation, whereas Twist1 is required to maintain late EMT. Twist1, present in resting epithelial cells, is dispensable for EMT initiation. Mechanistically, in response to transient TGFβ treatment, transient Snail1 expression represses Twist1 transcription directly, which is subsequently upregulated, as Snail1 levels decrease, to sustain E-cadherin downregulation and growth arrest of EMT. Persistent Twist1 expression is associated with a p38 and extracellular signal-regulated kinase signal feedback loop that sustains growth-inhibitory signals characteristic of quiescent micrometastatic tumors. This Snail1-Twist1 temporal and spatial cooperation was also observed in vivo during human breast cancer progression to metastasis. Twist1 level, but not Snail1 level, and Twist1:Snail1 ratio in disseminated micrometastatic bone marrow tumor cells was found to correlate with survival and treatment resistance and is highly predictive of metastatic or recurrent disease.

    Topics: Bone Marrow Neoplasms; Breast Neoplasms; Cell Dedifferentiation; Cell Transformation, Neoplastic; Epithelial Cells; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Humans; MAP Kinase Signaling System; Nuclear Proteins; Prognosis; Promoter Regions, Genetic; Protein Binding; Recurrence; RNA Interference; Signal Transduction; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta; Twist-Related Protein 1

2011
PET with the 89Zr-labeled transforming growth factor-β antibody fresolimumab in tumor models.
    Journal of nuclear medicine : official publication, Society of Nuclear Medicine, 2011, Volume: 52, Issue:12

    Transforming growth factor-β (TGF-β) promotes cancer invasion and metastasis and is therefore a potential drug target for cancer treatment. Fresolimumab, which neutralizes all mammalian active isoforms of TGF-β, was radiolabeled with (89)Zr for PET to analyze TGF-β expression, antibody tumor uptake, and organ distribution.. (89)Zr was conjugated to fresolimumab using the chelator N-succinyldesferrioxamine-B-tetrafluorphenol. (89)Zr-fresolimumab was analyzed for conjugation ratio, aggregation, radiochemical purity, stability, and immunoreactivity. (89)Zr-fresolimumab tumor uptake and organ distribution were assessed using 3 protein doses (10, 50, and 100 μg) and compared with (111)In-IgG in a human TGF-β-transfected Chinese hamster ovary xenograft model, human breast cancer MDA-MB-231 xenograft, and metastatic model. Latent and active TGF-β1 expression was analyzed in tissue homogenates with enzyme-linked immunosorbent assay.. (89)Zr was labeled to fresolimumab with high specific activity (>1 GBq/mg), high yield, and high purity. In vitro validation of (89)Zr-fresolimumab showed a fully preserved immunoreactivity and long (>1 wk) stability in solution and in human serum. In vivo validation showed an (89)Zr-fresolimumab distribution similar to IgG in most organs, except for a higher uptake in the liver in all mice and higher kidney uptake in the 10-μg group. (89)Zr-fresolimumab induced no toxicity in mice; it accumulated in primary tumors and metastases in a manner similar to IgG. Both latent and active TGF-β was detected in tumor homogenates, whereas only latent TGF-β could be detected in liver homogenates. Remarkably high (89)Zr-fresolimumab uptake was seen in sites of tumor ulceration and in scar tissue, processes in which TGF-β is known to be highly active.. Fresolimumab tumor uptake and organ distribution can be visualized and quantified with (89)Zr-fresolimumab PET. This technique will be used to guide further clinical development of fresolimumab and could possibly identify patients most likely to benefit.

    Topics: Animals; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; CHO Cells; Cricetinae; Cricetulus; Disease Models, Animal; Female; Gene Expression Regulation, Neoplastic; Humans; Isotope Labeling; Liver; Male; Mice; Neoplasm Metastasis; Positron-Emission Tomography; Radioisotopes; Transfection; Transforming Growth Factor beta; Zirconium

2011
PGE2 receptor EP2 mediates the antagonistic effect of COX-2 on TGF-beta signaling during mammary tumorigenesis.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2010, Volume: 24, Issue:4

    The molecular mechanisms that enable cyclooxygenase-2 (COX-2) and its mediator prostaglandin E2 (PGE2) to inhibit transforming growth factor-beta (TGF-beta) signaling during mammary tumorigenesis remain unknown. We show here that TGF-beta selectively stimulated the expression of the PGE2 receptor EP2, which increased normal and malignant mammary epithelial cell (MEC) invasion, anchorage-independent growth, and resistance to TGF-beta-induced cytostasis. Mechanistically, elevated EP2 expression in normal MECs inhibited the coupling of TGF-beta to Smad2/3 activation and plasminogen activator inhibitor-1 (PAI1) expression, while EP2 deficiency in these same MECs augmented Smad2/3 activation and PAI expression stimulated by TGF-beta. Along these lines, engineering malignant MECs to lack EP2 expression prevented their growth in soft agar, restored their cytostatic response to TGF-beta, decreased their invasiveness in response to TGF-beta, and potentiated their activation of Smad2/3 and expression of PAI stimulated by TGF-beta. More important, we show that COX-2 or EP2 deficiency both significantly decreased the growth, angiogenesis, and pulmonary metastasis of mammary tumors produced in mice. Collectively, this investigation establishes EP2 as a potent mediator of the anti-TGF-beta activities elicited by COX-2/PGE2 in normal and malignant MECs. Our findings also suggest that pharmacological targeting of EP2 receptors may provide new inroads to antagonize the oncogenic activities of TGF-beta during mammary tumorigenesis.-Tian, M., Schiemann, W. P. PGE2 receptor EP2 mediates the antagonistic effect of COX-2 on TGF-beta signaling during mammary tumorigenesis.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Cyclooxygenase 2; Female; Gene Expression Regulation, Neoplastic; Mammary Neoplasms, Animal; Mice; Mice, Inbred BALB C; Neoplasm Invasiveness; Neoplasm Proteins; Plasminogen Activator Inhibitor 1; Receptors, Prostaglandin E; Receptors, Prostaglandin E, EP2 Subtype; Signal Transduction; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta

2010
Transformed epithelial cells and fibroblasts/myofibroblasts interaction in breast tumor: a mathematical model and experiments.
    Journal of mathematical biology, 2010, Volume: 61, Issue:3

    It is well known that tumor and its microenvironment, or stroma, interact with each other and that this interaction plays a critical role in tumor initiation, growth, and metastasis. This interaction consists of complex relations between tumor cells, stromal cells such as fibroblasts, epithelial cells and immunocytes, the vascular system, the extracellular matrix, and cytokines secreted by the cells. Understanding these relationships may lead to new therapeutic approaches to cancer. In the present paper, we consider tumor-stroma crosstalk in a simple in vitro situation which involves interaction between tumor epithelial cells from breast cancer and a microenvironment consisting of just fibroblasts. The two populations of cells are separated by a semi-permeable membrane that allows only cytokines to cross over. We develop a mathematical model that includes two critical growth factors: TGF-beta, produced by the tumor cells, and EGF, secreted by the fibroblasts. The TGF-beta modifies the microenvironment by transforming fibroblasts into myofibroblasts. Myofibroblasts secrete higher concentrations of EGF than fibroblasts, thereby, increasing the proliferation of tumor cells. Thus already in this simple setup one sees a mutual interaction between tumor cells and their microenvironment. We conducted experiments which show good agreement with the model's simulations, hence confirming the model's ability to predict aspects of tumor cell behavior in response to signaling from fibroblasts.

    Topics: Breast Neoplasms; Cell Communication; Cell Transformation, Neoplastic; Computer Simulation; Epidermal Growth Factor; Epithelial Cells; Female; Fibroblasts; Humans; Models, Immunological; Transforming Growth Factor beta

2010
Smad3 protein levels are modulated by Ras activity and during the cell cycle to dictate transforming growth factor-beta responses.
    The Journal of biological chemistry, 2010, Feb-26, Volume: 285, Issue:9

    Transforming growth factor beta (TGF-beta) regulates many biological processes, and aberrant TGF-beta signaling is implicated in tumor development. Smad3 is a central component of the TGF-beta signaling pathway, and once activated, Smad3 forms complexes with Smad4 or other receptor-regulated Smads, which accumulate in the nucleus to transcriptionally regulate TGF-beta target genes. Because Smad3 plays a significant role in mediating the activities of TGF-beta, we examined its regulation during tumor development using a well characterized tumor model. We demonstrate that Smad3 levels are dramatically reduced in the tumorigenic cell line transformed with activated H-Ras compared with the normal parental epithelial cells. Interestingly, we also observe a cell cycle-dependent regulation of Smad3 in both cell types, with high Smad3 levels in quiescent cells and a significant drop in Smad3 protein levels in proliferating cells. Smad3 is regulated at the mRNA level and at the level of protein stability. In addition, functional analysis indicates that down-regulation of Smad3 levels is required for the tumor cells to proliferate in the presence of TGF-beta, because ectopic expression of Smad3 in the tumorigenic cell line restores the growth inhibitory response to TGF-beta. In contrast, expression of high levels of Smad3 did not interfere with the ability of these cells to undergo epithelial to mesenchymal transition upon TGF-beta stimulation. Altogether, our results suggest that the level of Smad3 protein is an important determinant of the progression of tumorigenesis. High levels of Smad3 are required for the tumor suppressor activities of TGF-beta, whereas lower levels are sufficient for the tumor promoting functions.

    Topics: Cell Cycle; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; Protein Stability; ras Proteins; RNA, Messenger; Smad3 Protein; Transforming Growth Factor beta

2010
A multi-strategy approach to informative gene identification from gene expression data.
    Journal of bioinformatics and computational biology, 2010, Volume: 8, Issue:1

    An unsupervised multi-strategy approach has been developed to identify informative genes from high throughput genomic data. Several statistical methods have been used in the field to identify differentially expressed genes. Since different methods generate different lists of genes, it is very challenging to determine the most reliable gene list and the appropriate method. This paper presents a multi-strategy method, in which a combination of several data analysis techniques are applied to a given dataset and a confidence measure is established to select genes from the gene lists generated by these techniques to form the core of our final selection. The remainder of the genes that form the peripheral region are subject to exclusion or inclusion into the final selection. This paper demonstrates this methodology through its application to an in-house cancer genomics dataset and a public dataset. The results indicate that our method provides more reliable list of genes, which are validated using biological knowledge, biological experiments, and literature search. We further evaluated our multi-strategy method by consolidating two pairs of independent datasets, each pair is for the same disease, but generated by different labs using different platforms. The results showed that our method has produced far better results.

    Topics: Animals; Artificial Intelligence; Cell Transformation, Neoplastic; Computational Biology; Databases, Genetic; Decision Trees; Gene Expression Profiling; Genomics; Humans; Leukemia, Myeloid, Acute; Mice; Oligonucleotide Array Sequence Analysis; Transforming Growth Factor beta

2010
Increased 18F-FDG uptake and expression of Glut1 in the EMT transformed breast cancer cells induced by TGF-beta.
    Neoplasma, 2010, Volume: 57, Issue:3

    As carcinomas progress, the tumors may lose epithelial morphology and acquire mesenchymal characteristics typically called epithelial-mesenchymal transition (EMT), which is commonly associated with increased cell migration, enables cells to dissociate from their original tissue and form metastasis in distant organs. In addition to molecular and morphologic changes, the EMT transformed cells also showed the change of sensitivity to chemotherapeutics. In order to detect the EMT transition in vivo clinically, we detected the change of metabolism of MCF-7 cells after being induced by TGF-beta to form EMT condition by MTT and 18F-FDG uptake.

    Topics: Breast Neoplasms; Cadherins; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Epithelial Cells; Female; Fluorodeoxyglucose F18; Glucose Transporter Type 1; Humans; Mesoderm; Radiopharmaceuticals; Transforming Growth Factor beta; Vimentin

2010
Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors.
    Cancer research, 2010, May-15, Volume: 70, Issue:10

    Transforming growth factor-beta (TGF-beta) is a potent inducer of epithelial to mesenchymal transition (EMT). However, it remains elusive about which molecular mechanisms determine the cellular capacity to undergo EMT in response to TGF-beta. We have found that both epidermal growth factor receptor (EGFR) overexpression and mutant p53 tumor suppressor genes contribute to the enrichment of an EMT-competent cellular subpopulation among telomerase-immortalized human esophageal epithelial cells during malignant transformation. EGFR overexpression triggers oncogene-induced senescence, accompanied by the induction of cyclin-dependent kinase inhibitors p15(INK4B), p16(INK4A), and p21. Interestingly, a subpopulation of cells emerges by negating senescence without loss of EGFR overexpression. Such cell populations express increased levels of zinc finger E-box binding (ZEB) transcription factors ZEB1 and ZEB2, and undergo EMT on TGF-beta stimulation. Enrichment of EMT-competent cells was more evident in the presence of p53 mutation, which diminished EGFR-induced senescence. RNA interference directed against ZEB resulted in the induction of p15(INK4B) and p16(INK4A), reactivating the EGFR-dependent senescence program. Importantly, TGF-beta-mediated EMT did not take place when cellular senescence programs were activated by either ZEB knockdown or the activation of wild-type p53 function. Thus, senescence checkpoint functions activated by EGFR and p53 may be evaded through the induction of ZEB, thereby allowing the expansion of an EMT-competent unique cellular subpopulation, providing novel mechanistic insights into the role of ZEB in esophageal carcinogenesis.

    Topics: Blotting, Western; Carcinoma, Squamous Cell; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Cellular Senescence; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p16; Epithelial Cells; ErbB Receptors; Esophageal Neoplasms; Esophagus; Fluorescent Antibody Technique; Homeodomain Proteins; Humans; Luciferases; Mesoderm; Repressor Proteins; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Telomerase; Transcription Factors; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Zinc Finger E-box Binding Homeobox 2; Zinc Finger E-box-Binding Homeobox 1

2010
Co-regulated expression of matrix metalloproteinase-2 and transforming growth factor-beta in melanoma development and progression.
    Oncology reports, 2010, Volume: 24, Issue:1

    Previous studies have found that matrix metalloproteinase-2 (MMP-2) and transforming growth factor-beta (TGF-beta) can be considered as biomarkers and indices of disease progression in several human cancers. In this study, we investigated the plasma levels of MMP-2 and TGF-beta and their correlation in 49 primary cutaneous melanoma and 10 metastatic melanoma. Plasma MMP-2 and TGF-beta levels in patients with primary melanoma were significantly higher than those of healthy controls. These protein levels were significantly higher in patients with metastatic melanoma. A positive correlation between plasma levels of MMP-2 and TGF-beta in melanoma patients supports the hypothesis that TGF-beta triggers the release of MMP-2. The immunohistochemistry analysis shows that MMP-2 and TGF-beta were highly expressed in tumor tissues as well as in matched plasma samples. This finding suggests that these proteins are released from tumor cells. Overall, our data indicate that MMP-2 and TGF-beta may represent novel diagnostic markers and therapeutic targets in melanoma and the determination of their concentration could be a useful diagnostic and prognostic indicator. TGF-beta, leading the tissue invasion mediated by MMP-2, is a strong promoter of tumor progression. Therefore, reducing or blocking the activity of TGF-beta may represent a promising target in therapeutic strategies for limiting the growth of melanoma.

    Topics: Adult; Aged; Aged, 80 and over; Biomarkers, Tumor; Case-Control Studies; Cell Transformation, Neoplastic; Disease Progression; Female; Humans; Immunohistochemistry; Male; Matrix Metalloproteinase 2; Melanoma; Middle Aged; Neoplasm Metastasis; Skin Neoplasms; Transforming Growth Factor beta

2010
Increased expression of Smad proteins, and in particular Smad3, in oral lichen planus compared to normal oral mucosa.
    Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology, 2010, Volume: 39, Issue:8

    Oral lichen planus (OLP) is a chronic inflammatory disease of the oral mucosa which the World Health Organisation (WHO) considers a premalignant condition. One step in malignant development is so called epithelial mesenchymal transition (EMT), a process whereby epithelial cells acquire mesenchymal characteristics. EMT occurs during embryogenesis and wound healing but also in some human diseases such as cancer and fibrosis. A factor known to induce EMT is transforming growth factor-β (TGF-β), which uses the Smad proteins as mediators for its signalling. TGF-β is also often over-expressed in squamous cell carcinoma of the head and neck (SCCHN).. In the present study we mapped expression of Smad proteins in OLP lesions by immunohistochemistry, and compared to expression in normal and sensitive oral mucosa. The latter group of patients had developed SCCHN after shorter or longer periods of diffuse oral symptoms. The aim was to see if there were any signs of EMT related changes in the OLP lesions, as judged by changes in the TGF-β pathway.. Changes in the TGF-β pathway related to EMT are seen in the very earliest stages of oral malignancy and become more severe as lesions progress.

    Topics: Adult; Aged; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Disease Progression; Epithelial-Mesenchymal Transition; Female; Head and Neck Neoplasms; Humans; Immunohistochemistry; Lichen Planus, Oral; Male; Middle Aged; Mouth Mucosa; Precancerous Conditions; Smad Proteins; Smad2 Protein; Smad3 Protein; Smad4 Protein; Smad7 Protein; Transforming Growth Factor beta; Young Adult

2010
Cytosolic phospholipase A2alpha and peroxisome proliferator-activated receptor gamma signaling pathway counteracts transforming growth factor beta-mediated inhibition of primary and transformed hepatocyte growth.
    Hepatology (Baltimore, Md.), 2010, Volume: 52, Issue:2

    Hepatocellular carcinoma often develops in the setting of abnormal hepatocyte growth associated with chronic hepatitis and liver cirrhosis. Transforming growth factor beta (TGF-beta) is a multifunctional cytokine pivotal in the regulation of hepatic cell growth, differentiation, migration, extracellular matrix production, stem cell homeostasis, and hepatocarcinogenesis. However, the mechanisms by which TGF-beta influences hepatic cell functions remain incompletely defined. We report herein that TGF-beta regulates the growth of primary and transformed hepatocytes through concurrent activation of Smad and phosphorylation of cytosolic phospholipase A(2)alpha (cPLA(2)alpha), a rate-limiting key enzyme that releases arachidonic acid for the production of bioactive eicosanoids. The interplays between TGF-beta and cPLA(2)alpha signaling pathways were examined in rat primary hepatocytes, human hepatocellular carcinoma cells, and hepatocytes isolated from newly developed cPLA(2)alpha transgenic mice.. Our data show that cPLA(2)alpha activates peroxisome proliferator-activated receptor gamma (PPAR-gamma) and thus counteracts Smad2/3-mediated inhibition of cell growth. Therefore, regulation of TGF-beta signaling by cPLA(2)alpha and PPAR-gamma may represent an important mechanism for control of hepatic cell growth and hepatocarcinogenesis.

    Topics: Animals; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Group IV Phospholipases A2; Hepatocytes; Humans; Mice; Mice, Transgenic; PPAR gamma; Signal Transduction; Transforming Growth Factor beta

2010
ERBB receptor activation is required for profibrotic responses to transforming growth factor beta.
    Cancer research, 2010, Oct-01, Volume: 70, Issue:19

    Engagement of the transforming growth factor-β (TGF-β) receptor complex activates multiple signaling pathways that play crucial roles in both health and disease. TGF-β is a key regulator of fibrogenesis and cancer-associated desmoplasia; however, its exact mode of action in these pathologic processes has remained poorly defined. Here, we report a novel mechanism whereby signaling via members of the ERBB or epidermal growth factor family of receptors serves as a central requirement for the biological responses of fibroblasts to TGF-β. We show that TGF-β triggers upregulation of ERBB ligands and activation of cognate receptors via the canonical SMAD pathway in fibroblasts. Interestingly, activation of ERBB is commonly observed in a subset of fibroblast but not epithelial cells from different species, indicating cell type specificity. Moreover, using genetic and pharmacologic approaches, we show that ERBB activation by TGF-β is essential for the induction of fibroblast cell morphologic transformation and anchorage-independent growth. Together, these results uncover important aspects of TGF-β signaling that highlight the role of ERBB ligands/receptors as critical mediators in fibroblast responses to this pleiotropic cytokine.

    Topics: Animals; Cell Adhesion; Cell Growth Processes; Cell Line; Cell Transformation, Neoplastic; Dogs; Fibroblasts; Gene Knockout Techniques; HeLa Cells; Humans; Mice; Oncogene Proteins v-erbB; Signal Transduction; Smad Proteins; Swiss 3T3 Cells; Transforming Growth Factor beta

2010
Requirement of TGFbeta signaling for SMO-mediated carcinogenesis.
    The Journal of biological chemistry, 2010, Nov-19, Volume: 285, Issue:47

    Hedgehog (Hh) signaling, via the key signal transducer Smoothened (SMO) and Gli transcription factors, is essential for embryonic development and carcinogenesis. At present, the molecular mechanism of Hh signaling-mediated carcinogenesis is not completely understood. Using a mouse model (K14cre/R26SmoM2) of SMO-mediated basal cell carcinoma development, we identified TGFβ2 as a major Hh-regulated gene. TGFβ2 expression was high in the keratinocytes, with activated TGFβ signaling (indicated by elevated expression of phosphorylated SMAD2/3) detected in both tumor and stroma. The significance of TGFβ signaling for SMO function was demonstrated in two assays. Down-regulation of TGFβ2 expression prevented Hh signaling-dependent osteoblast differentiation and motor neuron differentiation. Furthermore, inhibition of TGFβ signaling by TGFβ receptor I inhibitor SD208 significantly reduced tumor area in K14cre/R26SmoM2 mice. Tumor shrinkage in mice was associated with an increased number of lymphocytes, suggesting an immune suppression role of TGFβ signaling. The relevance of our results to human cancer is reflected by the fact that human basal cell carcinomas, which almost always harbor activated Hh signaling, have activated TGFβ signaling, as indicated by high levels of phosphorylated SMAD2 and SMAD3 in tumor and stroma. Together, our data indicate that TGFβ signaling is critical for Hh signaling-mediated carcinogenesis.

    Topics: Animals; Biomarkers; Blotting, Western; Carcinoma, Basal Cell; Cell Differentiation; Cell Transformation, Neoplastic; Cells, Cultured; Embryonic Stem Cells; Female; Gene Expression Profiling; Hedgehog Proteins; Humans; Integrases; Keratinocytes; Male; Mice; Mice, Inbred C3H; Motor Neurons; Oligonucleotide Array Sequence Analysis; Osteoblasts; Phosphorylation; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, G-Protein-Coupled; Receptors, Transforming Growth Factor beta; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Skin Neoplasms; Smad2 Protein; Smad3 Protein; Smoothened Receptor; Transforming Growth Factor beta

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
Maintaining mesenchymal properties of ovarian surface epithelial cells: a potential early protective role for TGF-beta in ovarian carcinogenesis.
    Endocrinology, 2010, Volume: 151, Issue:11

    Topics: Carcinoma; Cell Transformation, Neoplastic; Epithelial Cells; Female; Humans; Ovarian Neoplasms; Ovary; Transforming Growth Factor beta

2010
ILEI requires oncogenic Ras for the epithelial to mesenchymal transition of hepatocytes and liver carcinoma progression.
    Oncogene, 2009, Feb-05, Volume: 28, Issue:5

    In human hepatocellular carcinoma (HCC), epithelial to mesenchymal transition (EMT) correlates with aggressiveness of tumors and poor survival. We employed a model of EMT based on immortalized p19(ARF) null hepatocytes (MIM), which display tumor growth upon expression of oncogenic Ras and undergo EMT through the synergism of Ras and transforming growth factor (TGF)-beta. Here, we show that the interleukin-related protein interleukin-like EMT inducer (ILEI), a novel EMT-, tumor- and metastasis-inducing protein, cooperates with oncogenic Ras to cause TGF-beta-independent EMT. Ras-transformed MIM hepatocytes overexpressing ILEI showed cytoplasmic E-cadherin, loss of ZO-1 and induction of alpha-smooth muscle actin as well as platelet-derived growth factor (PDGF)/PDGF-R isoforms. As shown by dominant-negative PDGF-R expression in these cells, ILEI-induced PDGF signaling was required for enhanced cell migration, nuclear accumulation of beta-catenin, nuclear pY-Stat3 and accelerated growth of lung metastases. In MIM hepatocytes expressing the Ras mutant V12-C40, ILEI collaborated with PI3K signaling resulting in tumor formation without EMT. Clinically, human HCC samples showed granular or cytoplasmic localization of ILEI correlating with well and poorly differentiated tumors, respectively. In conclusion, these data indicate that ILEI requires cooperation with oncogenic Ras to govern hepatocellular EMT through mechanisms involving PDGF-R/beta-catenin and PDGF-R/Stat3 signaling.

    Topics: Animals; beta Catenin; Carcinoma; Cell Transformation, Neoplastic; Cells, Cultured; Cytokines; Disease Progression; Epithelial Cells; Female; Genes, ras; Hepatocytes; Humans; Liver Neoplasms; Male; Mesoderm; Mice; Mice, SCID; Neoplasm Proteins; Receptors, Platelet-Derived Growth Factor; STAT3 Transcription Factor; Tissue Distribution; Transforming Growth Factor beta

2009
TGF-beta-mediated activation of RhoA signalling is required for efficient (V12)HaRas and (V600E)BRAF transformation.
    Oncogene, 2009, Feb-19, Volume: 28, Issue:7

    Transforming growth factor beta-1 (TGF-beta) acts as both a tumour suppressor and a tumour promoter in a context-dependent manner. The tumour-promoting activities of TGF-beta are likely to result from a combination of Smad and non-Smad signalling pathways but remain poorly understood. Here we show that TGF-beta-mediated activation of RhoA is dependent on the kinase activity of ALK5 and that continuous ALK5 activity maintains basal RhoA-ROCK signalling, cell morphology and actin dynamics in serum-starved rodent fibroblasts independently of Smad2, Smad3 and Smad4. In immortalized human diploid fibroblasts, we show that oncogenic rewiring by transduction of (V12)HaRas instigates regulation of RhoA-ROCK signalling through an autocrine TGF-beta1-ALK5 pathway. Furthermore, we show that ALK5-mediated activation of RhoA is required for efficient (V12)HaRas, V-Raf and (V600E)BRAF transformation and (V12)HaRas-mediated anchorage-independent growth. These findings identify a new pro-oncogenic activity of TGF-beta and indicate that tumours harbouring (V12)HaRas and (V600E)BRAF mutations may be susceptible to TGF-beta signalling inhibitors.

    Topics: Actins; Animals; Benzamides; Blotting, Western; Cell Transformation, Neoplastic; Cells, Cultured; Cytoskeleton; Dioxoles; Enzyme-Linked Immunosorbent Assay; Fibroblasts; Fluorescent Antibody Technique; Genes, ras; Guanosine Triphosphate; Humans; Mice; NIH 3T3 Cells; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins B-raf; Rats; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; rho-Associated Kinases; rhoA GTP-Binding Protein; Signal Transduction; Smad3 Protein; Smad4 Protein; Transfection; Transforming Growth Factor beta

2009
TGFbeta-stimulated Smad1/5 phosphorylation requires the ALK5 L45 loop and mediates the pro-migratory TGFbeta switch.
    The EMBO journal, 2009, Jan-21, Volume: 28, Issue:2

    During the course of breast cancer progression, normally dormant tumour-promoting effects of transforming growth factor beta (TGFbeta), including migration, invasion, and metastasis are unmasked. In an effort to identify mechanisms that regulate the pro-migratory TGFbeta 'switch' in mammary epithelial cells in vitro, we found that TGFbeta stimulates the phosphorylation of Smad1 and Smad5, which are typically associated with bone morphogenetic protein signalling. Mechanistically, this phosphorylation event requires the kinase activity and, unexpectedly, the L45 loop motif of the type I TGFbeta receptor, ALK5, as evidenced by studies using short hairpin RNA-resistant ALK5 mutants in ALK5-depleted cells and in vitro kinase assays. Functionally, Smad1/5 co-depletion studies demonstrate that this phosphorylation event is essential to the initiation and promotion of TGFbeta-stimulated migration. Moreover, this phosphorylation event is preferentially detected in permissive environments such as those created by tumorigenic cells or oncogene activation. Taken together, our data provide evidence that TGFbeta-stimulated Smad1/5 phosphorylation, which occurs through a non-canonical mechanism that challenges the notion of selective Smad phosphorylation by ALK5, mediates the pro-migratory TGFbeta switch in mammary epithelial cells.

    Topics: Activins; Animals; Benzamides; Bone Morphogenetic Proteins; Breast Neoplasms; Cell Line; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Dioxoles; Humans; Mice; Phosphorylation; Protein Binding; Protein Isoforms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad1 Protein; Smad5 Protein; Transforming Growth Factor beta

2009
Presto change-o: the epithelial-mesenchymal transition.
    Journal of proteome research, 2009, Volume: 8, Issue:1

    Topics: Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Chromatography, Liquid; Epithelium; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Mass Spectrometry; Mesoderm; Neoplasm Metastasis; Proteomics; Transforming Growth Factor beta

2009
GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation.
    Genes & development, 2009, Jan-01, Volume: 23, Issue:1

    Pancreatic ductal adenocarcinoma (PDAC) is characterized by the deregulation of the hedgehog signaling pathway. The Sonic Hedgehog ligand (Shh), absent in the normal pancreas, is highly expressed in pancreatic tumors and is sufficient to induce neoplastic precursor lesions in mouse models. We investigated the mechanism of Shh signaling in PDAC carcinogenesis by genetically ablating the canonical bottleneck of hedgehog signaling, the transmembrane protein Smoothened (Smo), in the pancreatic epithelium of PDAC-susceptible mice. We report that multistage development of PDAC tumors is not affected by the deletion of Smo in the pancreas, demonstrating that autocrine Shh-Ptch-Smo signaling is not required in pancreatic ductal cells for PDAC progression. However, the expression of Gli target genes is maintained in Smo-negative ducts, implicating alternative means of regulating Gli transcription in the neoplastic ductal epithelium. In PDAC tumor cells, we find that Gli transcription is decoupled from upstream Shh-Ptch-Smo signaling and is regulated by TGF-beta and KRAS, and we show that Gli1 is required both for survival and for the KRAS-mediated transformed phenotype of cultured PDAC cancer cells.

    Topics: Animals; Carcinoma, Pancreatic Ductal; Cell Line; Cell Survival; Cell Transformation, Neoplastic; Cells, Cultured; Gene Expression Regulation, Neoplastic; Hedgehog Proteins; Humans; Kruppel-Like Transcription Factors; Mice; Pancreatic Ducts; Pancreatic Neoplasms; Proto-Oncogene Proteins p21(ras); Receptors, G-Protein-Coupled; Signal Transduction; Smoothened Receptor; Transforming Growth Factor beta; Zinc Finger Protein GLI1

2009
New TGF-beta and Ras crosstalk in EMT.
    Cell cycle (Georgetown, Tex.), 2009, Jan-15, Volume: 8, Issue:2

    Topics: Cell Transformation, Neoplastic; Epithelial Cells; Mesoderm; ras Proteins; Signal Transduction; Transforming Growth Factor beta

2009
Hepatitis B virus X protein shifts human hepatic transforming growth factor (TGF)-beta signaling from tumor suppression to oncogenesis in early chronic hepatitis B.
    Hepatology (Baltimore, Md.), 2009, Volume: 49, Issue:4

    Hepatitis B virus X (HBx) protein is suspected to participate in oncogenesis during chronic hepatitis B progression. Transforming growth factor beta (TGF-beta) signaling involves both tumor suppression and oncogenesis. TGF-beta activates TGF-beta type I receptor (TbetaRI) and c-Jun N-terminal kinase (JNK), which differentially phosphorylate the mediator Smad3 to become C-terminally phosphorylated Smad3 (pSmad3C) and linker-phosphorylated Smad3 (pSmad3L). Reversible shifting of Smad3-mediated signaling between tumor suppression and oncogenesis in HBx-expressing hepatocytes indicated that TbetaRI-dependent pSmad3C transmitted a tumor-suppressive TGF-beta signal, while JNK-dependent pSmad3L promoted cell growth. We used immunostaining, immunoblotting, and in vitro kinase assay to compare pSmad3L- and pSmad3C-mediated signaling in biopsy specimens representing chronic hepatitis, cirrhosis, or hepatocellular carcinoma (HCC) from 90 patients chronically infected with hepatitis B virus (HBV) with signaling in liver specimens from HBx transgenic mice. In proportion to plasma HBV DNA levels, early chronic hepatitis B specimens showed prominence of pSmad3L in hepatocytic nuclei. HBx-activated JNK/pSmad3L/c-Myc oncogenic pathway was enhanced, while the TbetaRI/pSmad3C/p21(WAF1) tumor-suppressive pathway was impaired as human and mouse HBx-associated hepatocarcinogenesis progressed. Of 28 patients with chronic hepatitis B who showed strong oncogenic pSmad3L signaling, six developed HCC within 12 years; only one of 32 patients showing little pSmad3L developed HCC. In contrast, seven of 30 patients with little Smad3C phosphorylation developed HCC, while no patient who retained hepatocytic tumor-suppressive pSmad3C developed HCC within 12 years.. HBx shifts hepatocytic TGF-beta signaling from the tumor-suppressive pSmad3C pathway to the oncogenic pSmad3L pathway in early carcinogenic process. Hepatocytic pSmad3L and pSmad3C assessment in HBV-infected liver specimens should prove clinically useful for predicting risk of HCC.

    Topics: Adult; Animals; Carcinoma, Hepatocellular; Cell Nucleus; Cell Proliferation; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; DNA-Binding Proteins; DNA, Viral; Female; Hepatitis B, Chronic; Hepatocytes; Humans; JNK Mitogen-Activated Protein Kinases; Liver Cirrhosis; Liver Neoplasms; Male; Mice; Mice, Transgenic; Middle Aged; Phosphorylation; Signal Transduction; Smad3 Protein; Trans-Activators; Transcription Factors; Transforming Growth Factor beta; Viral Regulatory and Accessory Proteins; Young Adult

2009
Intercellular communication of cellular stress monitored by gamma-H2AX induction.
    Carcinogenesis, 2009, Volume: 30, Issue:10

    When cells are exposed to ionizing radiation (IR), unexposed cells that share media with damaged cells exhibit similar effects to irradiated cells including increased levels of DNA double-strand breaks (DSBs). Hypothesizing that this effect, known as the radiation-induced bystander effect, may be a specific instance of communication between damaged and undamaged cells regardless of damage source, we demonstrated that exposure of target cells to non-IR induces bystander damage in non-targeted cells as measured by gamma-H2AX and 53BP1 focal formation. Initially, bystander damage was found primarily in S-phase cells, but at later times, non-S-phase cells were also affected. In addition, media from undamaged malignant and senescent cells also was found to induce DSBs in primary cultures. Media conditioned on cells targeted with either ionizing or non-IR as well as on undamaged malignant and senescent cells contained elevated levels of several cytokines. One of these, transforming growth factor beta (TGF-beta), and nitric oxide (NO) were found to elevate numbers of gamma-H2AX/53BP1 foci in normal cell cultures similar to levels found in bystander cells, and this elevation was abrogated by NO synthase inhibitors, TGF-beta blocking antibody and antioxidants. These findings support the hypothesis that damage in bystander cells results from their exposure to cytokines or reactive compounds released from stressed cells, regardless of damage source. These results have implications for oncogenesis in that they indicate that damaged normal cells or undamaged tumor cells may induce genomic instability, leading to an increased risk of oncogenic transformation in other cells with which they share media or contact directly.

    Topics: Breast; Cell Communication; Cell Division; Cell Transformation, Neoplastic; DNA, Neoplasm; Enzyme Inhibitors; Female; Fibroblasts; HeLa Cells; Histones; Humans; Nitric Oxide; Nitric Oxide Synthase; S Phase; Stress, Physiological; Transforming Growth Factor beta; Ultraviolet Rays; Uterine Cervical Neoplasms

2009
Hepatic tumor-stroma crosstalk guides epithelial to mesenchymal transition at the tumor edge.
    Oncogene, 2009, Nov-12, Volume: 28, Issue:45

    The tumor-stroma crosstalk is a dynamic process fundamental in tumor development. In hepatocellular carcinoma (HCC), the progression of malignant hepatocytes frequently depends on transforming growth factor (TGF)-beta provided by stromal cells. TGF-beta induces an epithelial to mesenchymal transition (EMT) of oncogenic Ras-transformed hepatocytes and an upregulation of platelet-derived growth factor (PDGF) signaling. To analyse the influence of the hepatic tumor-stroma crosstalk onto tumor growth and progression, we co-injected malignant hepatocytes and myofibroblasts (MFBs). For this, we either used in vitro-activated p19(ARF) MFBs or in vivo-activated MFBs derived from physiologically inflamed livers of Mdr2/p19(ARF) double-null mice. We show that co-transplantation of MFBs with Ras-transformed hepatocytes strongly enhances tumor growth. Genetic interference with the PDGF signaling decreases tumor cell growth and maintains plasma membrane-located E-cadherin and beta-catenin at the tumor-host border, indicating a blockade of hepatocellular EMT. We further generated a collagen gel-based three dimensional HCC model in vitro to monitor the MFB-induced invasion of micro-organoid HCC spheroids. This invasion was diminished after inhibition of TGF-beta or PDGF signaling. These data suggest that the TGF-beta/PDGF axis is crucial during hepatic tumor-stroma crosstalk, regulating both tumor growth and cancer progression.

    Topics: Animals; Cell Communication; Cell Transformation, Neoplastic; Epithelial Cells; Hepatocytes; Immunohistochemistry; Liver Neoplasms, Experimental; Mesoderm; Mice; Mice, Inbred BALB C; Mice, SCID; Platelet-Derived Growth Factor; Transforming Growth Factor beta

2009
The transcriptional regulatory function of p53 is essential for suppression of mouse skin carcinogenesis and can be dissociated from effects on TGF-beta-mediated growth regulation.
    The Journal of pathology, 2009, Volume: 219, Issue:2

    Transcriptional regulation by p53 is critical for p53-mediated tumour suppression; however, p53-mediated transactivation has been dissociated from p53-mediated biological processes including apoptosis, DNA repair, and differentiation. We compared the effects of a mutant allele, p53(QS - val135), containing a double mutation in the amino-terminus abrogating transactivation activity and a modification at amino acid 135 partially affecting DNA binding, to complete loss of p53. We applied in vitro endpoints correlated with epithelial tumourigenesis and an in vivo assay of tumour phenotype to assess whether loss of p53-mediated transcriptional regulation underlies the malignant phenotype of p53(-/-)/v-ras(Ha)-overexpressing keratinocytes. Transactivation deficiency of p53QS-val135 was confirmed by reporter gene assays in fibroblasts and differentiating keratinocytes. Ras oncogene-induced senescence was lost in both p53(QS - val135/QS - val135) and p53(-/-) keratinocytes. Similarly, p53(QS - val135/QS - val135), like p53(-/-), cooperated with v-ras(Ha) to enhance malignant conversion. The tumours arising in p53(QS - val135/QS - val135) keratinocytes displayed strong nuclear p53 expression; thus, the p53(QS - val135) allele was maintained and the deficient transactivation function of the expressed p53QS mutant protein was supported by absence of p21(waf1) in these tumours. The p53(QS - val135) allele did not confer a dominant-negative phenotype, as p53(+/QS - val135) keratinocytes senesced normally in response to v-ras(Ha) expression and formed benign tumours. While p53(-/-) keratinocytes displayed diminished response to TGF-beta, p53(QS - val135/QS - val135) and p53(+/+) keratinocytes responded equivalently, indicating that the requirement for p53 in maximizing TGF-beta-mediated growth regulation is independent of its transactivation domain and that the ability of keratinocytes to respond to TGF-beta is insufficient to suppress the malignant phenotype in this model. Furthermore, TGF-beta enhances p53QS-induced activation of a dual p53-TGF-beta responsive reporter in a keratinocyte cell line. These findings support an essential role for p53-mediated transcriptional regulation in suppressing malignancies arising from ras-induced skin tumours, consistent with previous findings in spontaneous carcinogenesis in other organs, and highlight the potential importance of senescence for tumour suppression in vivo.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Cells, Cultured; Cellular Senescence; Genes, p53; Genotype; Keratinocytes; Mice; Mice, Nude; Mutation; Neoplasm Transplantation; Skin Neoplasms; Transcriptional Activation; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2009
Dysfunctional transforming growth factor-beta receptor II accelerates prostate tumorigenesis in the TRAMP mouse model.
    Cancer research, 2009, Sep-15, Volume: 69, Issue:18

    The contribution of a dysfunctional transforming growth factor-beta type II receptor (TGF beta RII) to prostate cancer initiation and progression was investigated in an in vivo mouse model. Transgenic mice harboring the dominant-negative mutant TGF-beta type II receptor (DNTGF beta RII) in mouse epithelial cell were crossed with the TRAMP prostate cancer transgenic mouse to characterize the in vivo consequences of inactivated TGF-beta signaling on prostate tumor initiation and progression. Histopathologic diagnosis of prostate specimens from the TRAMP+/DNTGF beta RII double transgenic mice revealed the appearance of early malignant changes and subsequently highly aggressive prostate tumors at a younger age, compared with littermates TRAMP+/Wt TGF beta RII mice. Immunohistochemical and Western blotting analysis revealed significantly increased proliferative and apoptotic activities, as well as vascularity and macrophage infiltration that correlated with an elevated vascular endothelial growth factor and MCP-1 protein levels in prostates from TRAMP+/DNTGF beta RII+ mice. An epithelial-mesenchymal transition (EMT) effect was also detected in prostates of TRAMP+/DNTGF beta RII mice, as documented by the loss of epithelial markers (E-cadherin and beta-catenin) and up-regulation of mesenchymal markers (N-cadherin) and EMT-transcription factor Snail. A significant increase in the androgen receptor mRNA and protein levels was associated with the early onset of prostate tumorigenesis in TRAMP+/DNTGF beta RII mice. Our results indicate that in vivo disruption of TGF-beta signaling accelerates the pathologic malignant changes in the prostate by altering the kinetics of prostate growth and inducing EMT. The study also suggests that a dysfunctional TGF beta RII augments androgen receptor expression and promotes inflammation in early stage tumor growth, thus conferring a significant contribution by TGF-beta to prostate cancer progression.

    Topics: Adenocarcinoma; Animals; Apoptosis; Cell Growth Processes; Cell Transformation, Neoplastic; Disease Models, Animal; Inflammation; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Androgen; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

2009
Oncogene and tumour suppressor: the two faces of SnoN.
    The EMBO journal, 2009, Nov-18, Volume: 28, Issue:22

    SnoN was first identified based on its homology with the proto-oncogene c-Ski, and has since been implicated as a promoter of oncogenic transformation and cancer progression. Consistent with a role as proto-oncogene, SnoN negatively regulates TGF-beta signalling, through its interactions with Smad complexes. Thus, SnoN inhibits the growth inhibitory effect of TGF-beta, which is considered as the basis for the tumour suppressor activity of TGF-beta signalling. In this issue of The EMBO Journal, Pan et al (2009) now demonstrate that SnoN also functions as a tumour suppressor, independently of its role in Smad signalling. The tumour suppressor role of SnoN results from its interaction with the promyelocytic leukaemia (PML) protein and the accumulation of SnoN in PML nuclear bodies, thus allowing SnoN to stabilize p53 and induce premature senescence.

    Topics: Animals; Cell Transformation, Neoplastic; Genes, Tumor Suppressor; Humans; Intracellular Signaling Peptides and Proteins; Mice; Models, Biological; Oncogenes; Proto-Oncogene Mas; Proto-Oncogene Proteins; Signal Transduction; Smad Proteins; Transcription Factors; Transforming Growth Factor beta

2009
Disruption of transforming growth factor-beta signaling by five frequently methylated genes leads to head and neck squamous cell carcinoma pathogenesis.
    Cancer research, 2009, Dec-15, Volume: 69, Issue:24

    Head and neck squamous cell carcinoma (HNSCC) is an aggressive cancer with low survival rates in advanced stages. To facilitate timely diagnosis and improve outcome, early detection markers (e.g., DNA methylation) are crucial for timely cancer diagnosis. In a recent publication, an epigenome-wide screen revealed a set of genes that are commonly methylated and downregulated in head and neck cancers (SEPT9, SLC5A8, FUSSEL18, EBF3, and IRX1). Interestingly, these candidates are potentially involved in the transforming growth factor-beta (TGF-beta) signaling pathway, which is often disrupted in HNSCC. Therefore, we sought to determine coordinated epigenetic silencing of these candidate genes in HNSCC as potential key disruptors of TGF-beta signaling, which could ultimately result in HNSCC progression. Through immunoprecipitation studies, all five of the investigated candidate genes were found to interact with components of the TGF-beta pathway. Overexpression of SLC5A8, EBF3, and IRX1 resulted in decreased mitotic activity and increased apoptosis. In addition, EBF3 was found to increase p21 promoter activity, and SMAD2 significantly increased IRX1 promoter activity. These findings are significant because they reveal a set of genes that interact with components of the TGF-beta pathway, and their silencing via methylation in HNSCC results in coordinated decrease in apoptosis, increased proliferation, and decreased differentiation.

    Topics: Apoptosis; Carcinoma, Squamous Cell; Cation Transport Proteins; Cell Differentiation; Cell Growth Processes; Cell Line, Tumor; Cell Transformation, Neoplastic; Cytoskeletal Proteins; Disease Progression; DNA Methylation; Gene Expression Regulation, Neoplastic; Gene Silencing; GTP-Binding Proteins; Head and Neck Neoplasms; Homeodomain Proteins; Humans; Microtubule-Associated Proteins; Monocarboxylic Acid Transporters; Nerve Tissue Proteins; Proto-Oncogene Proteins; Septins; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2009
MMPs, TIMP-2, and TGF-beta1 in the cancerization of oral lichen planus.
    Head & neck, 2008, Volume: 30, Issue:9

    Matrix metalloproteinases (MMPs) and transforming growth factor-beta1 (TGF-beta1) are important in many physiological and pathological processes.. Immunohistochemistry for MMP-2, MMP-9, membrane-type 1 MMP (MT1-MMP, MMP-14), tissue inhibitor of matrix metalloproteinase (TIMP)-2, and TGF-beta were performed on normal mucosa, nonatrophic oral lichen planus, atrophic oral lichen planus, and oral squamous cell carcinomas (OSCC) resulting from lichen planus.. Expression of MMPs progressively increased from normal mucosa to nonatrophic oral lichen planus, atrophic oral lichen planus, and OSCCs. Immunoscores of MMPs in atrophic oral lichen planus was significantly greater than nonatrophic oral lichen planus. Moreover, immunoscore of MMP-9 of OSCCs was significantly greater than both atrophic and nonatrophic lichen planus. Furthermore, expression of TIMP-2 and TGF-beta1 paralleled increases seen with MMPs.. Imbalance between MMPs and TIMPs may be involved in cancerization of oral lichen planus. MMP-2, MT1-MMP, and especially MMP-9 may be useful markers for judging potency of malignant transformation from oral lichen planus.

    Topics: Adult; Age Factors; Biomarkers, Tumor; Biopsy, Needle; Case-Control Studies; Cell Transformation, Neoplastic; Cytokines; Disease Progression; Female; Humans; Immunohistochemistry; Lichen Planus, Oral; Male; Metalloproteases; Middle Aged; Mouth Neoplasms; Precancerous Conditions; Probability; Prognosis; Reference Values; Retrospective Studies; Risk Factors; Sensitivity and Specificity; Severity of Illness Index; Sex Factors; Tissue Inhibitor of Metalloproteinase-1; Transforming Growth Factor beta

2008
Transforming growth factor-beta (TGF-beta) and TGF-beta-associated kinase 1 are required for R-Ras-mediated transformation of mammary epithelial cells.
    Cancer research, 2008, Aug-01, Volume: 68, Issue:15

    Transforming growth factor-beta (TGF-beta) cooperates with oncogenic members of the Ras superfamily to promote cellular transformation and tumor progression. Apart from the classic (H-, K-, and N-) Ras GTPases, only the R-Ras subfamily (R-Ras, R-Ras2/TC21, and R-Ras3/M-Ras) has significant oncogenic potential. In this study, we show that oncogenic R-Ras transformation of EpH4 cells requires TGF-beta signaling. When murine EpH4 cells were stably transfected with a constitutively active R-Ras(G38V) mutant, they were no longer sensitive to TGF-beta-mediated growth inhibition and showed increased proliferation and transformation in response to exogenous TGF-beta. R-Ras/EpH4 cells require TGF-beta signaling for transformation to occur and they produce significantly elevated levels of endogenous TGF-beta, which signals in an autocrine fashion. The effects of TGF-beta are independent of Smad2/3 activity and require activation of TGF-beta-associated kinase 1 (TAK1) and its downstream effectors c-Jun NH(2)-terminal kinase and p38 mitogen-activated protein kinase as well as the phosphoinositide 3-kinase/Akt and mammalian target of rapamycin pathways. Thus, TAK1 is a novel link between TGF-beta signaling and oncogenic R-Ras in the promotion of tumorigenesis.

    Topics: Blotting, Western; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Humans; Mammary Glands, Human; MAP Kinase Kinase Kinases; Oncogene Protein p21(ras); Signal Transduction; Transforming Growth Factor beta

2008
Role of the prostanoid FP receptor in action potential generation and phenotypic transformation of NRK fibroblasts.
    Cellular signalling, 2008, Volume: 20, Issue:11

    By using an shRNA approach to knockdown the expression of the prostaglandin (PG)-F(2alpha) receptor (FP-R), the role of PGF(2alpha) in the process of phenotypic transformation of normal rat kidney (NRK) fibroblasts has been studied. Our data show that PGF(2alpha) up-regulates Cox-2 expression both at the mRNA and protein level, indicating that activation of FP-R in NRK fibroblasts induces a positive feedback loop in the production PGF(2alpha). Knockdown of FP-R expression fully impaired the ability of PGF(2alpha) to induce a calcium response and subsequent depolarization in NRK cells. However, these cells could still undergo phenotypic transformation when treated with a combination of EGF and retinoic acid, but in contrast to the wild-type cells, this process was not accompanied by a membrane depolarization to -20 mV. Knockdown of FP-R expression also impaired the spontaneous firing of calcium action potentials by density-arrested NRK cells. These data show that a membrane depolarization is not a prerequisite for the acquisition of a transformed phenotype. Furthermore, our data provide the first direct evidence that activity of PGF(2alpha) by putative pacemaker cells underlies the generation of calcium action potentials in NRK monolayers.

    Topics: Action Potentials; Animals; Cell Count; Cell Line, Transformed; Cell Proliferation; Cell Transformation, Neoplastic; Cyclooxygenase 1; Cyclooxygenase 2; Dinoprost; Down-Regulation; Enzyme Induction; Fibroblasts; Humans; Membrane Proteins; Phenotype; Rats; Receptors, Prostaglandin; RNA, Small Interfering; Transforming Growth Factor beta; Tretinoin

2008
Fibulin-5 initiates epithelial-mesenchymal transition (EMT) and enhances EMT induced by TGF-beta in mammary epithelial cells via a MMP-dependent mechanism.
    Carcinogenesis, 2008, Volume: 29, Issue:12

    Epithelial-mesenchymal transition (EMT) is a normal physiological process that regulates tissue development, remodeling and repair; however, aberrant EMT also elicits disease development in humans, including lung fibrosis, rheumatoid arthritis and cancer cell metastasis. Transforming growth factor-beta (TGF-beta) is a master regulator of EMT in normal mammary epithelial cells (MECs), wherein this pleiotropic cytokine also functions as a potent suppressor of mammary tumorigenesis. In contrast, malignant MECs typically evolve resistance to TGF-beta-mediated cytostasis and develop the ability to proliferate, invade and metastasize when stimulated by TGF-beta. It therefore stands to reason that establishing how TGF-beta promotes EMT may offer new insights into targeting the oncogenic activities of TGF-beta in human breast cancers. By monitoring alterations in the actin cytoskeleton and various markers of EMT, we show here that the TGF-beta gene target, fibulin-5 (FBLN5), initiates EMT and enhances that induced by TGF-beta. Whereas normal MECs contain few FBLN5 transcripts, those induced to undergo EMT by TGF-beta show significant upregulation of FBLN5 messenger RNA, suggesting that EMT and the dedifferentiation of MECs override the repression of FBLN5 expression in polarized MECs. We also show that FBLN5 stimulated matrix metalloproteinase expression and activity, leading to MEC invasion and EMT, to elevated Twist expression and to reduced E-cadherin expression. Finally, FBLN5 promoted anchorage-independent growth in normal and malignant MECs, as well as enhanced the growth of 4T1 tumors in mice. Taken together, these findings identify a novel EMT and tumor-promoting function for FBLN5 in developing and progressing breast cancers.

    Topics: Animals; Breast Neoplasms; Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Epithelial Cells; Extracellular Matrix Proteins; Female; Immunohistochemistry; Mammary Glands, Animal; Metalloproteases; Mice; Recombinant Proteins; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Transforming Growth Factor beta

2008
Sustained induction of epithelial to mesenchymal transition activates DNA methylation of genes silenced in basal-like breast cancers.
    Proceedings of the National Academy of Sciences of the United States of America, 2008, Sep-30, Volume: 105, Issue:39

    The active acquisition of epigenetic changes is a poorly understood but important process in development, differentiation, and disease. Our work has shown that repression of the p16/pRb pathway in human epithelial cells, a condition common to stem cells and many tumor cells, induces dynamic epigenetic remodeling resulting in the targeted methylation of a selected group of CpG islands. We hypothesized that cells in this epigenetically plastic state could be programmed by the microenvironment to acquire epigenetic changes associated with tumorigenesis. Here, we describe an in vitro model system where epigenetically plastic cells were placed in an environment that induced epithelial to mesenchymal transition (EMT) and led to a program of acquired de novo DNA methylation at targeted sites. In this model, we found that repression of E-cadherin transcription preceded the subsequent acquisition of methylated CpG sites. Furthermore, the induction of EMT was accompanied by de novo methylation of several other gene promoters, including those of the estrogen receptor and Twist. These data demonstrate that signals from the microenvironment can induce phenotypic and gene expression changes associated with targeted de novo epigenetic alterations important in tumor progression, and that these alterations occur through a deterministic, rather than stochastic, mechanism. Given the dynamic epigenetic reprogramming that occurs in these cells, DNA methylation profiles observed in human tumors may reflect the history of environmental exposures during the genesis of a tumor.

    Topics: Breast Neoplasms; Cadherins; Cell Line, Tumor; Cell Transformation, Neoplastic; DNA Methylation; Epithelial Cells; Gene Expression Regulation, Neoplastic; Gene Silencing; Humans; Mesoderm; Promoter Regions, Genetic; Serum; Smad2 Protein; Transforming Growth Factor beta

2008
Transformation by oncogenic Ras expands the early genomic response to transforming growth factor beta in intestinal epithelial cells.
    Neoplasia (New York, N.Y.), 2008, Volume: 10, Issue:10

    A substantial body of evidence implicates TGFbeta as a tumor promoter in epithelial cells that have become resistant to its tumor suppressor activity. To better understand early, genome-wide TGFbeta responses in cells resistant to growth inhibition by TGFbeta, we used microarray analysis in a well-defined cell culture system of sensitive and resistant intestinal epithelial cells. TGFbeta-regulated gene expression in TGFbeta-growth-sensitive, nontransformed rat intestinal epithelial cells (RIE-1) was compared to expression in TGFbeta-growth-resistant RIE cells stably transformed by oncogenic Ras(12V). Treatment of RIE-1 cells with 2 ng/ml TGFbeta1 for 1 hour increased the expression of eight gene sequences by 2.6-fold or more, whereas eight were down regulated 2.6-fold. In RIE-Ras(12V) cells, 42 gene sequences were upregulated and only 3 were down-regulated. Comparison of RIE and RIE-Ras(12V) identified 37 gene sequences as unique, Ras-dependent genomic targets of TGFbeta1. TGFbeta-regulation of connective tissue growth factor and vascular endothelial growth factor, two genes up-regulated in RIE-Ras cells and previously implicated in tumor promotion, was independently confirmed and further characterized by Northern analysis. Our data indicate that overexpression of oncogenic Ras in intestinal epithelial cells confers a significantly expanded repertoire of robust, early transcriptional responses to TGFbeta via signaling pathways yet to be fully elucidated but including the canonical Raf-1/MAPK/Erk pathway. Loss of sensitivity to growth inhibition by TGFbeta does not abrogate TGFbeta signaling and actually expands the early transcriptional response to TGFbeta1. Expression of some of these genes may confer to Ras-transformed cells characteristics favorable for tumor promotion.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Connective Tissue Growth Factor; Disease Progression; Epithelial Cells; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Genes, ras; Genome; Intestinal Mucosa; Oligonucleotide Array Sequence Analysis; Rats; Time Factors; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2008
Extensive alteration in the expression profiles of TGFB pathway signaling components and TP53 is observed along the gastric dysplasia-carcinoma sequence.
    Histology and histopathology, 2008, Volume: 23, Issue:12

    The expression patterns of TGFB signaling proteins, such as TGFB1/2, TGFBR1(ALK5), TGFBR2, SMAD1/2/3, SMAD2/3, SMAD4, SMAD7, and of downstream targets of TGFB signaling, CDKN1A (p21CIP1), CDKN1B (p27KIP1), MYC, CDC25A, TP53, and RELA (p65NF-kB) were investigated in gastric carcinomas and other gastric lesions.. A total of 112 gastric carcinomas, 37 dysplasias, 54 intestinal metaplasias, 29 chronic atrophic gastritis and 54 normal gastric epithelium were analyzed by tissue microarray-based immunohistochemical analysis. Extensive changes in expression profiles of these proteins were observed. Three types of expression patterns were observed along the normal epithelium-atrophic gastritis-dysplasia-carcinoma sequence. (1) Expression of TGFB1/2, TGFBR1, MYC, and TP53 continually increased along this sequence. (2) Expression of SMAD4, CDKN1A, SMAD1/2/3, SMAD2/3, and CDKN1B was enhanced in dysplasia but decreased in carcinoma. (3) Expression of TGFBR2, SMAD7, RELA, and CDC25A was enhanced in dysplasia and the enhanced level was maintained in carcinoma. In addition, we also evaluated the clinical significance of the expression of TGFB signaling proteins in gastric carcinoma. TGFB and MYC were positively correlated with advanced stages, whereas SMAD1/2/3 and SMAD4 were strongly associated with earlier stages.. The extensive change in expression of TGFB signaling components is implicated during tumorigenesis of gastric neoplasias.

    Topics: Adenocarcinoma; Aged; Aged, 80 and over; Cell Transformation, Neoplastic; Female; Gene Expression; Gene Expression Profiling; Humans; Immunohistochemistry; Male; Middle Aged; Precancerous Conditions; Signal Transduction; Stomach Neoplasms; Tissue Array Analysis; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2008
Sustained TGF beta exposure suppresses Smad and non-Smad signalling in mammary epithelial cells, leading to EMT and inhibition of growth arrest and apoptosis.
    Oncogene, 2008, Feb-21, Volume: 27, Issue:9

    To better understand the dual, tumour-suppressive and tumour-promoting function of transforming growth factor-beta (TGFbeta), we analysed mammary epithelial NMuMG cells in response to short and long-term TGFbeta exposure. NMuMG cells became proliferation-arrested and apoptotic after exposure to TGFbeta for 2-5 days, whereas surviving cells underwent epithelial-mesenchymal transition (EMT). After chronic TGFbeta exposure (2-3 weeks), however, NMuMG cells became resistant to proliferation arrest and apoptosis, showing sustained EMT instead (TD cells). EMT was fully reversed by a pharmacologic TGFbeta-receptor-I kinase inhibitor or withdrawal of TGFbeta for 6-12 days. Interestingly, both cell cycle arresting/proapoptotic (Smads, p38 kinase) and antiapoptotic, proliferation and EMT-promoting signalling pathways (PI3K-PKB/Akt, ERK) were co-suppressed to low, but significant levels. Except for PI3K-Akt, TGFbeta-dependent downregulation of these signalling pathways in transdifferentiated (TD) cells was fully reversed upon TGFbeta withdrawal, together with partial re-induction of proliferation arrest and apoptosis. Co-injection of non-tumorigenic NMuMG cells with tumour-forming CHO cells oversecreting exogenous TGFbeta1 (CHO-TGFbeta1) allowed outgrowth of epithelioid cells in CHO-TGFbeta1 cell-induced tumours. These epithelial islands enhanced CHO-TGFbeta1 tumour cell proliferation, possibly due to chemokines (for example, JE/MCP-1) secreted by NMuMG/TD cells. We conclude that suppression of antiproliferative, proapoptotic TGFbeta signalling in TD cells may permit TGFbeta-dependent proliferation, survival and EMT-enhancing signalling pathways to act at low levels. Thus, TGFbeta may modulate its own signalling to facilitate switching from tumour suppression to tumour progression.

    Topics: Animals; Apoptosis; Cell Culture Techniques; Cell Line; Cell Transformation, Neoplastic; CHO Cells; Cricetinae; Cricetulus; Epithelial Cells; Female; Genes, Tumor Suppressor; Mammary Glands, Animal; Mesoderm; Mice; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2008
Mutual paracrine effects of oral squamous cell carcinoma cells and normal oral fibroblasts: induction of fibroblast to myofibroblast transdifferentiation and modulation of tumor cell proliferation.
    Oral oncology, 2008, Volume: 44, Issue:5

    Several lines of evidence demonstrated that the stroma surrounding the tumors plays an important role in the growth and progression of several neoplasms, including oral squamous cell carcinomas (OSCC). We evaluated the presence of myofibroblasts in OSCC and determined whether their presence is associated with clinicopathological features of the tumors. We also investigated the mutual paracrine effects of tumor cells and myofibroblasts on fibroblast-myofibroblast transdifferentiation and tumor cell proliferation. Immunohistochemical analysis showed the approximately 60% of the OSCCs contained myofibroblasts in the stroma of the tumor. Abundant presence of myofibroblasts significantly correlated with N stage, disease stage, regional recurrence, and proliferative potential of the tumor cells. Using OSCC cell lines and primary oral normal fibroblasts (ONF), we demonstrated that tumor cells induced transdifferentiation of ONFs to myofibroblasts via secretion of transforming growth factor-beta 1 (TGF-beta 1). In turn, myofibroblasts secreted factors that stimulated OSCC cell proliferation, as revealed by measuring BrdU incorporation and Ki67 expression. The results of the study suggest that during tumor invasion OSCC-derived TGF-beta 1 promote fibroblast-myofibroblast transdifferentiation, and that tumor cellular proliferation can be induced by factors released from myofibroblasts, which may favor tumor growth.

    Topics: Adult; Aged; Aged, 80 and over; Carcinoma, Squamous Cell; Cell Proliferation; Cell Transdifferentiation; Cell Transformation, Neoplastic; Female; Fibroblasts; Gene Expression Profiling; Humans; Immunohistochemistry; Male; Middle Aged; Mouth Neoplasms; Paracrine Communication; Phenotype; Stromal Cells; Transforming Growth Factor beta; Tumor Cells, Cultured

2008
TGFbeta-induced EMT requires focal adhesion kinase (FAK) signaling.
    Experimental cell research, 2008, Jan-01, Volume: 314, Issue:1

    The epithelial-to-mesenchymal transition (EMT) is a crucial process, occurring both during development and tumor progression, by which an epithelial cell undergoes a conversion to a mesenchymal phenotype, dissociates from initial contacts and migrates to secondary sites. We recently reported that in hepatocytes the multifunctional cytokine TGFbeta induces a full EMT characterized by (i) Snail induction, (ii) E-cadherin delocalization and down-regulation, (iii) down-regulation of the hepatocyte transcriptional factor HNF4alpha and (iv) up-regulation of mesenchymal and invasiveness markers. In particular, we showed that Snail directly causes the transcriptional down-regulation of E-cadherin and HNF4, while it is not sufficient for the up-regulation of mesenchymal and invasiveness EMT markers. In this paper, we show that in hepatocytes TGFbeta induces a Src-dependent activation of the focal adhesion protein FAK. More relevantly, we gathered results indicating that FAK signaling is required for (i) transcriptional up-regulation of mesenchymal and invasiveness markers and (ii) delocalization of membrane-bound E-cadherin. Our results provide the first evidence of FAK functional role in TGFbeta-mediated EMT in hepatocytes.

    Topics: Animals; Biomarkers, Tumor; Cadherins; Cell Line; Cell Transformation, Neoplastic; Enzyme Activation; Epithelial Cells; Focal Adhesion Protein-Tyrosine Kinases; Hepatocytes; Liver Neoplasms; Mesoderm; Mice; Neoplasm Invasiveness; Signal Transduction; src-Family Kinases; Transcriptional Activation; Transforming Growth Factor beta; Up-Regulation

2008
Smad3 deficiency inhibits v-ras-induced transformation by suppression of JNK MAPK signaling and increased farnesyl transferase inhibition.
    Oncogene, 2008, Apr-10, Volume: 27, Issue:17

    The ability of transforming growth factor-beta (TGF-beta) to modulate various effects on distinct cell lineages has been a central feature of its multi-faceted nature. The purpose of this study was to access the effects of deletion of a key TGF-beta signal transducer, Smad3, on MAPK activation and v-Ras(Ha)-transformation of primary mouse embryonic fibroblasts (MEFs). We observe reduced TGF-beta1 and v-ras(Ha) mediated activation of the JNK and ERK MAPK pathway upon ablation of Smad3. Further, Smad3-deficient MEFs demonstrate resistance to v-ras(Ha)-induced transformation while the absence of Smad3 results in increased inhibition of farnesyl transferase activity. Taken together, these observations demonstrate that the absence of Smad3 protects fibroblasts from oncogenic transformation by (i) augmenting farnesyl transferase inhibition and (ii) suppressing the Ras-JNK MAPK pathway. These results provide new insights into the molecular mechanisms involved in v-Ras(Ha) oncogene-induced mesenchymal phenotypic transformation.

    Topics: Alkyl and Aryl Transferases; Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Chlorocebus aethiops; Enzyme Activation; Gene Expression Regulation; MAP Kinase Signaling System; Mice; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; Oncogene Protein p21(ras); Phenotype; Smad3 Protein; Transforming Growth Factor beta; Transgenes

2008
Sonic hedgehog signaling promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway.
    Carcinogenesis, 2008, Volume: 29, Issue:3

    It is known that the activation of hedgehog (Hh) signaling is involved in the progression and invasion of various tumors, including gastric carcinoma. In this study, we investigated the impact of transforming growth factor (TGF)-beta signaling on the sonic hedgehog (Shh)-mediated invasion of gastric cancer cells. We found that higher concentrations of N-Shh enhanced cell motility and invasiveness in gastric cancer cells, whereas no increase was observed in cells that were treated with KAAD-cyclopamine (a Shh signaling inhibitor) or anti-Shh blocking antibodies. In addition, the N-Shh-induced migration and invasiveness of gastric cancer cells were reduced by treatment with anti-TGF-beta blocking antibody or TGF-beta1 small interfering RNA (siRNA) in presence of N-Shh when compared with control groups. Furthermore, TGF-beta1 secretion, TGF-beta-mediated transcriptional response, expression of activin receptor-like kinase (ALK) 5 protein and phosphorylation of Smad 3 were also enhanced by treatment with N-Shh, but not KAAD-cyclopamine, anti-Shh or TGF-beta1 blocking antibodies. Blockade of the ALK5 kinase in the presence of N-Shh significantly inhibited phosphorylation of Smad 3, activity of matrix metalloproteinases and Shh-induced cell motility/invasiveness. Importantly, transient expression of ALK5 siRNA or Smad 3 siRNA reduced the ability of N-Shh to stimulate migration and invasion of those cells compared with the cells treated with non-specific control siRNA. In summary, these results indicate that Shh promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway. Additionally, our findings are the first to suggest a role and mechanism for Shh signaling as it relates to the metastatic potential of gastric cancer, thereby indicating potential therapeutic molecular targets to decrease metastasis.

    Topics: Base Sequence; Cell Line, Tumor; Cell Transformation, Neoplastic; DNA Primers; Hedgehog Proteins; Humans; Immunohistochemistry; Lymphatic Metastasis; Matrix Metalloproteinase 2; Matrix Metalloproteinase 9; Neoplasm Invasiveness; Neoplasm Metastasis; Phosphorylation; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; RNA, Small Interfering; Signal Transduction; Smad3 Protein; Stomach Neoplasms; Transforming Growth Factor beta

2008
Ski/SnoN expression in the sequence metaplasia-dysplasia-adenocarcinoma of Barrett's esophagus.
    Human pathology, 2008, Volume: 39, Issue:3

    Barrett's esophagus (BE) is a precancerous condition. However, the mechanisms underlying the transformation from metaplastic to dysplastic to adenocarcinomatous epithelium are still poorly understood. As loss of transforming growth factor-beta growth inhibition is considered a hallmark of several human neoplasms, we evaluated the expression of Ski and SnoN (proteins that antagonize transforming growth factor-beta signaling through physical interaction with Smad complex and by recruiting histone deacetylases), as markers of the transforming growth factor-beta signaling pathway, in BE with and without dysplasia. Biopsy samples from 37 patients (26 men, aged 60 +/- 8 years) with histologically proven BE were evaluated; 10 patients had concomitant low-grade dysplasia, 7 high-grade dysplasia (HGD), and 6 HGD associated with adenocarcinoma. Ski and SnoN expression was assessed immunohistochemically. Neither Ski nor SnoN was expressed in normal esophageal epithelium, but both were strongly expressed in BE tissue, with intense cytoplasmic positivity. Expression of these proteins decreased markedly in dysplastic areas in patients with low-grade dysplasia and was absent in those with HGD or HGD/adenocarcinoma. Ski and SnoN proteins are overexpressed in BE and may be involved in abnormal signaling elicited by transforming growth factor-beta in this epithelium, enhancing the tumorigenesis process. These observations might help to elucidate the molecular mechanisms involved in the BE tumorigenesis process.

    Topics: Adenocarcinoma; Aged; Barrett Esophagus; Cell Transformation, Neoplastic; DNA-Binding Proteins; Esophageal Neoplasms; Humans; Immunohistochemistry; Intracellular Signaling Peptides and Proteins; Male; Metaplasia; Middle Aged; Precancerous Conditions; Proto-Oncogene Proteins; Transforming Growth Factor beta

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
TGF-beta and hepatocellular carcinoma.
    Hepatology (Baltimore, Md.), 2008, Volume: 47, Issue:3

    Topics: Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Hepatitis C, Chronic; Humans; Liver Neoplasms; Transforming Growth Factor beta

2008
Cancer-associated transforming growth factor beta type II receptor gene mutant causes activation of bone morphogenic protein-Smads and invasive phenotype.
    Cancer research, 2008, Mar-15, Volume: 68, Issue:6

    Transforming growth factor beta (TGFbeta) plays a key role in maintaining tissue homeostasis by inducing cell cycle arrest, differentiation and apoptosis, and ensuring genomic integrity. Furthermore, TGFbeta orchestrates the response to tissue injury and mediates repair by inducing epithelial to mesenchymal transition and by stimulating cell motility and invasiveness. Although loss of the homeostatic activity of TGFbeta occurs early on in tumor development, many advanced cancers have coopted the tissue repair function to enhance their metastatic phenotype. How these two functions of TGFbeta become uncoupled during cancer development remains poorly understood. Here, we show that, in human keratinocytes, TGFbeta induces phosphorylation of Smad2 and Smad3 as well as Smad1 and Smad5 and that both pathways are dependent on the kinase activities of the type I and II TGFbeta receptors (T beta R). Moreover, cancer-associated missense mutations of the T beta RII gene (TGFBR2) are associated with at least two different phenotypes. One type of mutant (TGFBR2(E526Q)) is associated with loss of kinase activity and all signaling functions. In contrast, a second mutant (TGFBR2(R537P)) is associated with high intrinsic kinase activity, loss of Smad2/3 activation, and constitutive activation of Smad1/5. Furthermore, this TGFBR2 mutant endows the carcinoma cells with a highly motile and invasive fibroblastoid phenotype. This activated phenotype is T beta RI (Alk-5) independent and can be reversed by the action of a dual T beta RI and T beta RII kinase inhibitor. Thus, identification of such activated T beta RII receptor mutations in tumors may have direct implications for appropriately targeting these cancers with selective therapeutic agents.

    Topics: Bone Morphogenetic Proteins; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Head and Neck Neoplasms; Humans; Keratinocytes; Mutation, Missense; Phosphorylation; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Recombinant Proteins; Smad Proteins; Transfection; Transforming Growth Factor beta

2008
B-Raf is required for ERK activation and tumor progression in a mouse model of pancreatic beta-cell carcinogenesis.
    Oncogene, 2008, Aug-14, Volume: 27, Issue:35

    Activation of the Raf/MEK/ERK pathway, often by gain-of-function mutations of RAS or RAF, is observed in many human cancers. The extracellular signal-regulated kinase (ERK) pathway is required for the proliferation of cancer cell lines harboring activating BRAF or, to a lesser extent, activating RAS mutations. It is still unclear, however, whether the pathway is required in vivo for tumor development, particularly in tumors in which B-Raf is not mutationally activated. During embryonic development, B-Raf is essential for angiogenesis in the placenta. To address the question of whether B-Raf contributed to tumor angiogenesis in vivo we conditionally ablated B-Raf in a model of pancreatic islet carcinoma driven by the functional inactivation of tumor suppressors (RIP1Tag2), which critically depends on angiogenesis for growth. We find that B-Raf is dispensable for the proliferation of tumor cells in culture, but necessary for ERK activation and for the expression of angiogenic factors by tumor cells in vivo and in vitro. In vivo, these defects result in the formation of hollow tumors with decreased vessel density and strongly reduced proliferation. The progression from adenoma to carcinoma is also significantly impaired. Thus, endogenous B-Raf contributes to the development of RIP1Tag2 tumors by supporting the stromal response and tumor progression.

    Topics: Animals; Base Sequence; Cell Transformation, Neoplastic; Disease Models, Animal; Disease Progression; DNA Primers; Enzyme Activation; Extracellular Signal-Regulated MAP Kinases; Immunohistochemistry; Islets of Langerhans; Mice; Mice, Knockout; Pancreatic Neoplasms; Proto-Oncogene Proteins B-raf; Reverse Transcriptase Polymerase Chain Reaction; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A

2008
Dual role of SnoN in mammalian tumorigenesis.
    Molecular and cellular biology, 2007, Volume: 27, Issue:1

    SnoN is an important negative regulator of transforming growth factor beta signaling through its ability to interact with and repress the activity of Smad proteins. It was originally identified as an oncoprotein based on its ability to induce anchorage-independent growth in chicken embryo fibroblasts. However, the roles of SnoN in mammalian epithelial carcinogenesis have not been well defined. Here we show for the first time that SnoN plays an important but complex role in human cancer. SnoN expression is highly elevated in many human cancer cell lines, and this high level of SnoN promotes mitogenic transformation of breast and lung cancer cell lines in vitro and tumor growth in vivo, consistent with its proposed pro-oncogenic role. However, this high level of SnoN expression also inhibits epithelial-to-mesenchymal transdifferentiation. Breast and lung cancer cells expressing the shRNA for SnoN exhibited an increase in cell motility, actin stress fiber formation, metalloprotease activity, and extracellular matrix production as well as a reduction in adherens junction proteins. Supporting this observation, in an in vivo breast cancer metastasis model, reducing SnoN expression was found to moderately enhance metastasis of human breast cancer cells to bone and lung. Thus, SnoN plays both pro-tumorigenic and antitumorigenic roles at different stages of mammalian malignant progression. The growth-promoting activity of SnoN appears to require its ability to bind to and repress the Smad proteins, while the antitumorigenic activity can be mediated by both Smad-dependent and Smad-independent pathways and requires the activity of small GTPase RhoA. Our study has established the importance of SnoN in mammalian epithelial carcinogenesis and revealed a novel aspect of SnoN function in malignant progression.

    Topics: Animals; Breast Neoplasms; cdc42 GTP-Binding Protein; Cell Line, Tumor; Cell Transformation, Neoplastic; Female; Gene Expression Regulation, Neoplastic; Humans; Intracellular Signaling Peptides and Proteins; Mice; Mice, Inbred BALB C; Neoplasm Metastasis; Neoplasm Transplantation; Neoplasms, Glandular and Epithelial; Proto-Oncogene Proteins; rhoA GTP-Binding Protein; Transforming Growth Factor beta

2007
Transcriptional cooperation between the transforming growth factor-beta and Wnt pathways in mammary and intestinal tumorigenesis.
    Cancer research, 2007, Jan-01, Volume: 67, Issue:1

    Transforming growth factor-beta (TGF-beta) and Wnt ligands function in numerous developmental processes, and alterations of both signaling pathways are associated with common pathologic conditions, including cancer. To obtain insight into the extent of interdependence of the two signaling cascades in regulating biological responses, we used an oligonucleotide microarray approach to identify Wnt and TGF-beta target genes using normal murine mammary gland epithelial cells as a model. Combination treatment of TGF-beta and Wnt revealed a novel transcriptional program that could not have been predicted from single ligand treatments and included a cohort of genes that were cooperatively induced by both pathways. These included both novel and known components or modulators of TGF-beta and Wnt pathways, suggesting that mutual feedback is a feature of the coordinated activities of the ligands. The majority of the cooperative targets display increased expression in tumors derived from either Min (many intestinal neoplasia) or mouse mammary tumor virus (MMTV)-Wnt1 mice, two models of Wnt-induced tumors, with nine of these genes (Ankrd1, Ccnd1, Ctgf, Gpc1, Hs6st2, IL11, Inhba, Mmp14, and Robo1) showing increases in both. Reduction of TGF-beta signaling by expression of a dominant-negative TGF-beta type II receptor in bigenic MMTV-Wnt1/DNIIR mice increased mammary tumor latency and was correlated with a decrease in expression of Gpc1, Inhba, and Robo1, three of the TGF-beta/Wnt cooperative targets. Our results indicate that the TGF-beta and Wnt/beta-catenin pathways are firmly intertwined and generate a unique gene expression pattern that can contribute to tumor progression.

    Topics: Adenoma; Animals; Cell Transformation, Neoplastic; Female; Gene Expression Regulation, Neoplastic; Humans; Intestinal Neoplasms; L Cells; Mammary Neoplasms, Experimental; Mice; Mice, Inbred C57BL; Mice, Transgenic; Signal Transduction; Transcription, Genetic; Transforming Growth Factor beta; Wnt Proteins; Wnt3 Protein

2007
Low-dose irradiation of nontransformed cells stimulates the selective removal of precancerous cells via intercellular induction of apoptosis.
    Cancer research, 2007, Feb-01, Volume: 67, Issue:3

    An important stage in tumorigenesis is the ability of a precancerous cell to escape natural anticancer signals imposed on it by neighboring cells and its microenvironment. We have previously characterized a system of intercellular induction of apoptosis whereby nontransformed cells selectively remove transformed cells from coculture via cytokine and reactive oxygen/nitrogen species (ROS/RNS) signaling. We report that irradiation of nontransformed cells with low doses of either high linear energy transfer (LET) alpha-particles or low-LET gamma-rays leads to stimulation of intercellular induction of apoptosis. The use of scavengers and inhibitors confirms the involvement of ROS/RNS signaling and of the importance of transformed cell NADPH oxidase in the selectivity of the system. Doses as low as 2-mGy gamma-rays and 0.29-mGy alpha-particles were sufficient to produce an observable increase in transformed cell apoptosis. This radiation-stimulated effect saturates at very low doses (50 mGy for gamma-rays and 25 mGy for alpha-particles). The use of transforming growth factor-beta (TGF-beta) neutralizing antibody confirms a role for the cytokine in the radiation-induced signaling. The system may represent a natural anticancer mechanism stimulated by extremely low doses of ionizing radiation.

    Topics: Alpha Particles; Animals; Antibodies; Apoptosis; Cell Line, Transformed; Cell Transformation, Neoplastic; Dose-Response Relationship, Radiation; Fibroblasts; Gamma Rays; NADPH Oxidases; Precancerous Conditions; Rats; Reactive Nitrogen Species; Reactive Oxygen Species; Signal Transduction; 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
"Smad"eningly erratic: target gene methylation determines whether TGFbeta promotes or suppresses malignant glioma.
    Developmental cell, 2007, Volume: 12, Issue:3

    TGFbeta functions as a tumor suppressor in some contexts and a tumor promoter in others. In a recent issue of Cancer Cell, Bruna et al. (2007) shed light on an epigenetic mechanism that underlies this schizophrenic behavior in malignant glioma. Their findings highlight a stem cell/cancer link...and a potential blind spot in large-scale cancer genome sequencing projects.

    Topics: Animals; Brain Neoplasms; Cell Transformation, Neoplastic; DNA Methylation; Epigenesis, Genetic; Gene Expression Regulation, Neoplastic; Glioma; Humans; Smad Proteins; Stem Cells; Transforming Growth Factor beta

2007
Cross-talk between paracrine-acting cytokine and chemokine pathways promotes malignancy in benign human prostatic epithelium.
    Cancer research, 2007, May-01, Volume: 67, Issue:9

    The present study explores the mechanisms by which human prostatic carcinoma-associated fibroblasts (CAF) induce tumorigenesis in initiated but nonmalignant human prostatic epithelial cells (BPH-1). CAF express elevated levels of both transforming growth factor-beta1 (TGF-beta1) and stromal cell-derived factor-1 (SDF-1/CXCL12). TGF-beta inhibits the growth of BPH-1 cells in vitro, but was found to be necessary for the tumorigenic response to CAF. This counterintuitive result suggested that the TGF-beta signaling system was involved in other processes relating to tumorigenesis. The SDF-1 receptor, CXCR4, is expressed at low levels in benign prostate tissue and in BPH-1 cells in culture. However, CXCR4 levels increase during prostate cancer progression. CXCR4 was found to be induced and localized to the cell membrane in BPH1 cells by CAF-conditioned medium and by CAF cells in tissue recombinants. TGF-beta was both necessary and sufficient to allow the detection of membrane-localized CXCR4 in BPH1 cells. Suppression of epithelial cell CXCR4 expression abrogated the tumorigenic response to CAF. SDF-1, secreted by CAF, acts via the TGF-beta-regulated CXCR4 to activate Akt in the epithelial cells. This mechanism elicits tumorigenesis and obviates the growth-inhibitory effects of TGF-beta. Thus, tumor stroma can contribute to carcinogenesis through synergism between TGF-beta, SDF-1, and CXCR4. These experiments suggest mechanisms by which TGF-beta can shift its role from an inhibitor to a promoter of proliferation during tumor progression. Both the TGF-beta and SDF-1 pathways are targets of drug discovery efforts; these data suggest potential benefits in the cotargeting of these pathways.

    Topics: Animals; Cell Communication; Cell Transformation, Neoplastic; Chemokine CXCL12; Chemokines, CXC; Culture Media, Conditioned; Epithelial Cells; Female; Fibroblasts; Humans; Male; Pregnancy; Prostate; Prostatic Neoplasms; Rats; Rats, Sprague-Dawley; Receptors, CXCR4; Signal Transduction; Stromal Cells; Transforming Growth Factor beta; Transplantation, Heterologous

2007
Reversible Smad-dependent signaling between tumor suppression and oncogenesis.
    Cancer research, 2007, Jun-01, Volume: 67, Issue:11

    Cancer cells often gain advantage by reducing the tumor-suppressive activity of transforming growth factor-beta (TGF-beta) together with stimulation of its oncogenic activity as in Ras-transformed cells; however, molecular mechanisms remain largely unknown. TGF-beta activates both its type I receptor (TbetaRI) and c-Jun NH2-terminal kinase (JNK), which phosphorylate Smad2 and Smad3 at the COOH-terminal (pSmad2/3C) and linker regions (pSmad2/3L). Here, we report that Ras transformation suppresses TbetaRI-mediated pSmad3C signaling, which involves growth inhibition by down-regulating c-Myc. Instead, hyperactive Ras constitutively stimulates JNK-mediated pSmad2/3L signaling, which fosters tumor invasion by up-regulating plasminogen activator inhibitor-1 and matrix metalloproteinase-1 (MMP-1), MMP-2, and MMP-9. Conversely, selective blockade of linker phosphorylation by a mutant Smad3 lacking JNK-dependent phosphorylation sites results in preserved tumor-suppressive function via pSmad3C in Ras-transformed cells while eliminating pSmad2/3L-mediated invasive capacity. Thus, specific inhibition of the JNK/pSmad2/3L pathway should suppress cancer progression by shifting Smad-dependent signaling from oncogenesis to tumor suppression.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Gastric Mucosa; Genes, ras; Genes, Tumor Suppressor; MAP Kinase Kinase 4; Matrix Metalloproteinases; Phosphorylation; Plasminogen Activator Inhibitor 1; ras Proteins; Rats; Signal Transduction; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta

2007
Mammary epithelial cells treated concurrently with TGF-alpha and TGF-beta exhibit enhanced proliferation and death.
    Experimental biology and medicine (Maywood, N.J.), 2007, Volume: 232, Issue:8

    Transforming growth factor-alpha (TGF-alpha) stimulates while TGF-beta inhibits mammary epithelial cell growth, suggesting that when cells are treated concurrently with the growth factors their combined effects would result in no net growth. However, combined treatments stimulate proliferation and cellular transformation in several cell lines. The objective of this paper was to describe the effect of long-term (6 days) concurrent TGF-alpha and TGF-beta treatment on normal mammary epithelial cell growth pattern, morphology, and gene expression. Growth curve analysis showed that TGF-alpha enhanced while TGF-beta suppressed growth rate until Day 4, when cells entered lag phase. However, cells treated concurrently with both growth factors exhibited a dichotomous pattern of growth marked by growth and death phases (with no intermittent lag phase). These changes in growth patterns were due to a marked induction of cell death from Day 2 (16.5%) to Day 4 (89.5%), resulting in the transition from growth to death phases, even though the combined treated cultures had significantly more (P < 0.05) cells in S phase on Day 4. TGF-beta stimulated epithelial to mesenchyme transdifferentiation (EMT) in the presence of TGF-alpha, as characterized by increased expression of fibronectin and changes in TGF-beta receptor binding. Expression patterns of genes that regulate the cell cycle showed significant interaction between treatment and days, with TGF-beta overriding TGF-alpha-stimulated effects on gene expression. Overall, the combined treatments were marked by enhanced rates of cellular proliferation, death, and trans-differentiation, behaviors reminiscent of breast tumors, and thus this system may serve as a good model to study breast tumorigenesis.

    Topics: Animals; Cell Cycle; Cell Cycle Proteins; Cell Death; Cell Line; Cell Transformation, Neoplastic; Epithelial Cells; Fibronectins; Mammary Glands, Animal; Mammary Neoplasms, Animal; Mesoderm; Mice; Transforming Growth Factor alpha; Transforming Growth Factor beta

2007
Ionizing radiation predisposes nonmalignant human mammary epithelial cells to undergo transforming growth factor beta induced epithelial to mesenchymal transition.
    Cancer research, 2007, Sep-15, Volume: 67, Issue:18

    Transforming growth factor beta1 (TGFbeta) is a tumor suppressor during the initial stage of tumorigenesis, but it can switch to a tumor promoter during neoplastic progression. Ionizing radiation (IR), both a carcinogen and a therapeutic agent, induces TGFbeta activation in vivo. We now show that IR sensitizes human mammary epithelial cells (HMEC) to undergo TGFbeta-mediated epithelial to mesenchymal transition (EMT). Nonmalignant HMEC (MCF10A, HMT3522 S1, and 184v) were irradiated with 2 Gy shortly after attachment in monolayer culture or treated with a low concentration of TGFbeta (0.4 ng/mL) or double treated. All double-treated (IR + TGFbeta) HMEC underwent a morphologic shift from cuboidal to spindle shaped. This phenotype was accompanied by a decreased expression of epithelial markers E-cadherin, beta-catenin, and ZO-1, remodeling of the actin cytoskeleton, and increased expression of mesenchymal markers N-cadherin, fibronectin, and vimentin. Furthermore, double treatment increased cell motility, promoted invasion, and disrupted acinar morphogenesis of cells subsequently plated in Matrigel. Neither radiation nor TGFbeta alone elicited EMT, although IR increased chronic TGFbeta signaling and activity. Gene expression profiling revealed that double-treated cells exhibit a specific 10-gene signature associated with Erk/mitogen-activated protein kinase (MAPK) signaling. We hypothesized that IR-induced MAPK activation primes nonmalignant HMEC to undergo TGFbeta-mediated EMT. Consistent with this, Erk phosphorylation was transiently induced by irradiation and persisted in irradiated cells treated with TGFbeta, and treatment with U0126, a MAP/Erk kinase (MEK) inhibitor, blocked the EMT phenotype. Together, these data show that the interactions between radiation-induced signaling pathways elicit heritable phenotypes that could contribute to neoplastic progression.

    Topics: Breast; Cell Transformation, Neoplastic; Enzyme Activation; Epithelial Cells; Humans; MAP Kinase Signaling System; Mesoderm; Mitogen-Activated Protein Kinase Kinases; Transforming Growth Factor beta

2007
Keeping order in the neighborhood: new roles for TGFbeta in maintaining epithelial homeostasis.
    Cancer cell, 2007, Volume: 12, Issue:4

    TGFbetas are thought to have tumor suppressor activity in many organ systems, but receptor inactivation in mouse models has not previously resulted in increased spontaneous tumorigenesis. A study in this issue of Cancer Cell shows that mice with a targeted knockout of the type II TGFbeta receptor in stratified epithelia specifically develop spontaneous squamous cell carcinomas in the anogenital region, but not in the skin. Loss of TGFbeta signaling appears to destabilize the epithelium such that homeostasis fails in the face of persistent proliferative challenge, a normal feature of the anogenital site, and latent invasive and migratory phenotypes are unmasked.

    Topics: Animals; Anus Neoplasms; Apoptosis; Carcinoma, Squamous Cell; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Epithelial Cells; Extracellular Matrix; Focal Adhesion Protein-Tyrosine Kinases; Homeostasis; Humans; Integrins; Keratin-14; Keratinocytes; Mice; Mice, Knockout; Mutation; Neoplasm Invasiveness; Papilloma; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; ras Proteins; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Skin; Skin Neoplasms; src-Family Kinases; Time Factors; Transforming Growth Factor beta; Urogenital Neoplasms; Wound Healing

2007
Loss of TGFbeta signaling destabilizes homeostasis and promotes squamous cell carcinomas in stratified epithelia.
    Cancer cell, 2007, Volume: 12, Issue:4

    Although TGFbeta is a potent inhibitor of proliferation, epithelia lacking the essential receptor (TbetaRII) for TGFbeta signaling display normal tissue homeostasis. By studying asymptomatic TbetaRII-deficient stratified epithelia, we show that tissue homeostasis is maintained by balancing hyperproliferation with elevated apoptosis. Moreover, rectal and genital epithelia, which are naturally proliferative, develop spontaneous squamous cell carcinomas with age when TbetaRII is absent. This progression is associated with a reduction in apoptosis and can be accelerated in phenotypically normal epidermis by oncogenic mutations in Ras. We show that TbetaRII deficiency leads to enhanced keratinocyte motility and integrin-FAK-Src signaling. Together, these mechanisms provide a molecular framework to account for many of the characteristics of TbetaRII-deficient invasive SQCCs.

    Topics: Animals; Anus Neoplasms; Apoptosis; Carcinoma, Squamous Cell; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Epithelial Cells; Extracellular Matrix; Focal Adhesion Protein-Tyrosine Kinases; Homeostasis; Humans; Integrins; Keratin-14; Keratinocytes; Male; Mice; Mice, Knockout; Mutation; Neoplasm Invasiveness; Papilloma; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; ras Proteins; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Skin; Skin Neoplasms; src-Family Kinases; Time Factors; Transforming Growth Factor beta; Urogenital Neoplasms; Wound Healing

2007
Bone morphogenetic protein 2 opposes Shh-mediated proliferation in cerebellar granule cells through a TIEG-1-based regulation of Nmyc.
    The Journal of biological chemistry, 2007, Dec-21, Volume: 282, Issue:51

    Nmyc is a potent regulator of cell cycle in cerebellar granular neuron precursors (CGNPs) and has been proposed to be the main effector of Shh (Sonic hedgehog) proliferative activity. Nmyc ectopic expression is sufficient to promote cell autonomous proliferation and can lead to tumorigenesis. Bone morphogenetic protein 2 (BMP2) antagonizes Shh proliferative effect by promoting cell cycle exit and differentiation in CGNPs. Here we report that BMP2 opposes Shh mitogenic activity by blocking Nmyc expression. We have identified TIEG-1 (KLF10) as the intermediary factor that blocks Nmyc expression through the occupancy of the Sp1 sites present in its promoter. We also demonstrate that TIEG-1 ectopic expression in CGNPs induces cell cycle arrest that can lead to apoptosis but fails to promote differentiation. Moreover, TIEG-1 synergizes with BMP2 activity to terminally differentiate CGNPs and independent differentiator signals such as dibutyryl cAMP and prevents apoptosis in TIEG-1 arrested cells. All together, these data strongly suggest that the BMP2 pathway triggers cell cycle exit and differentiation as two separated but coordinated processes, where TIEG-1 acts as a mediator of the cell cycle arrest.

    Topics: Animals; Apoptosis; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Bucladesine; Cell Cycle; Cell Differentiation; Cell Transformation, Neoplastic; Cells, Cultured; Cerebellum; DNA-Binding Proteins; Gene Expression; Hedgehog Proteins; Mice; Neurons; Proto-Oncogene Proteins c-myc; Response Elements; Signal Transduction; Stem Cells; Transcription Factors; Transforming Growth Factor beta

2007
Overexpression of N-Myc rapidly causes acute myeloid leukemia in mice.
    Cancer research, 2007, Nov-15, Volume: 67, Issue:22

    N-MYC encodes a basic helix-loop-helix/leucine zipper (bHLH/LZ) transcription factor that is frequently overexpressed in human neuroblastoma. N-MYC overexpression has also been reported in human acute myeloid leukemias (AML), which we show here is a frequent event. Myeloid cells in N-Myc-overexpressing mouse bone marrow hyperproliferate but those in c-MYC-overexpressing bone marrow do not. The NH(2)-terminal transactivation domain, nuclear localization signal, and bHLH/LZ domain of N-Myc are essential for this effect. Microarray analysis revealed 969 differentially expressed genes between N-Myc- and c-MYC-overexpressing myeloid cells. N-Myc-overexpressing cells showed decreased transforming growth factor beta signaling and increased c-Jun-NH(2)-kinase signaling, both of which are associated with proliferation and leukemic transformation of myeloid cells. Mice transplanted with bone marrow expressing wild-type N-Myc developed clonal and transplantable AML after approximately 1 month; those transplanted with bone marrow expressing mutant N-Myc did not. Twist, a known suppressor of the p19Arf/p53 pathway, was up-regulated in all tumors. These results show that N-Myc overexpression is highly oncogenic in mouse myeloid cells and suggest that N-MYC up-regulation contributes to human myeloid leukemogenesis.

    Topics: Animals; Bone Marrow Cells; Cell Proliferation; Cell Separation; Cell Transformation, Neoplastic; Gene Expression Regulation, Leukemic; JNK Mitogen-Activated Protein Kinases; Leukemia, Myeloid, Acute; Mice; Myeloid Cells; Oligonucleotide Array Sequence Analysis; Protein Structure, Tertiary; Proto-Oncogene Proteins c-myc; Signal Transduction; Transforming Growth Factor beta

2007
Differential expression of TGFbeta-stimulated clone 22 in normal prostate and prostate cancer.
    International journal of cancer, 2006, Feb-15, Volume: 118, Issue:4

    The transforming growth factor-beta (TGFbeta) superfamily and its downstream effector genes are key regulators of epithelial homeostasis. Altered expression of these genes may be associated with malignant transformation of the prostate gland. The cDNA array analysis of differential expression of the TGFbeta superfamily and functionally related genes between patient-matched noncancerous prostate (NP) and prostate cancer (PC) bulk tissue specimens highlighted two genes, namely TGFbeta-stimulated clone-22 (TSC-22) and Id4. Verification of their mRNA expression by real-time PCR in patient-matched NP and PC bulk tissue, in laser-captured pure epithelial and cancer cells and in NP and PC cell lines confirmed TSC-22 underexpression, but not Id4 overexpression, in PC and in human PC cell lines. Immunohistochemical analysis showed that TSC-22 protein expression in NP is restricted to the basal cells and colocalizes with the basal cell marker cytokeratin 5. In contrast, all matched PC samples lack TSC-22 immunoreactivity. Likewise, PC cell lines do not show detectable TSC-22 protein expression as shown by immunoblotting. TSC-22 should be considered as a novel basal cell marker, potentially useful for studying lineage determination within the epithelial compartment of the prostate. Conversely, lack of TSC-22 seems to be a hallmark of malignant transformation of the prostate epithelium. Accordingly, TSC-22 immunohistochemistry may prove to be a diagnostic tool for discriminating benign lesions from malignant ones of the prostate. The suggested tumour suppressor function of TSC-22 warrants further investigation on its role in prostate carcinogenesis and on the TSC-22 pathway as a candidate therapeutic target in PC.

    Topics: Aged; Cell Transformation, Neoplastic; Gene Expression Profiling; Humans; Immunohistochemistry; Inhibitor of Differentiation Proteins; Male; Oligonucleotide Array Sequence Analysis; Polymerase Chain Reaction; Prostate; Prostatic Neoplasms; Repressor Proteins; RNA, Messenger; Transforming Growth Factor beta; Tumor Cells, Cultured

2006
Effects of human papillomavirus type 16 E7 protein on the growth of cervical carcinoma cells and immuno-escape through the TGF-beta1 signaling pathway.
    Gynecologic oncology, 2006, Volume: 101, Issue:1

    E7 is regarded as one of the main oncoproteins of high-risk human papillomaviruses (HPVs). It may affect the transforming growth factor beta 1 (TGF-beta1) signaling pathway. In this study, the relationship between HPV-16 infection and the functions of three critical factors of the TGF-beta1/Smads pathway was explored to assess the possible role of E7 in the development of cervical cancer.. The expression of E7, TGF-beta1, TbetaR-II and Smad4 was detected by immunohistochemistry in paraffin-embedded cervical samples, and by RT-PCR and Western blotting in cervical cancer cell lines. The effect of TGF-beta1 on the growth of cervical cancer cells were tested by methyl thiazolyl tetrazolium (MTT), and the effects of HPV-16 E7 protein on normal and malignant cervical cells were investigated by flow cytometry.. During the progression from benign to malignant lesions, the expression levels of TGF-beta1 and Smad4 increased significantly in cervical carcinoma tissues. The expression of TGF-beta1 was positively correlated with E7 expression. In vitro experiments showed that TGF-beta1 could not inhibit the proliferation of several cervical carcinoma cell lines in long-term regulation, but could inhibit immunologic reactions of peripheral blood mononuclear cells (PBMCs). Blocking E7 expression could lower the expression level of TGF-beta1 and induce cells to enter apoptosis.. Our data indicate that HPV-16 E7 protein plays an important role during the development of cervical cancer by immuno-inhibition and stimulation of tumor cell proliferation through the TGF-beta1/Smads signaling pathway.

    Topics: Cell Growth Processes; Cell Line, Tumor; Cell Transformation, Neoplastic; Enzyme-Linked Immunosorbent Assay; Female; HeLa Cells; Human papillomavirus 16; Humans; Interferon-gamma; Interleukin-2; Leukocytes, Mononuclear; Oligonucleotides, Antisense; Oncogene Proteins, Viral; Papillomavirus E7 Proteins; Papillomavirus Infections; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Messenger; Signal Transduction; Smad4 Protein; Transfection; Transforming Growth Factor beta; Transforming Growth Factor beta1; Uterine Cervical Dysplasia; Uterine Cervical Neoplasms

2006
TGF-beta1 drives partial myofibroblastic differentiation in chondromyxoid fibroma of bone.
    The Journal of pathology, 2006, Volume: 208, Issue:1

    Chondromyxoid fibroma (CMF) is a rare benign cartilaginous bone tumour with a lobular architecture containing stellate and myofibroblast-like spindle cells. The aim of this study was to investigate the presence, spatial distribution, and extent of myoid differentiation in CMF and to evaluate a possible causative role for TGF-beta1 signalling, which is known to promote smooth muscle actin (SMA) expression. Twenty cases were studied for immunoreactivity for muscle-specific actin (MSA), SMA, desmin, h-caldesmon, calponin, TGF-beta1, and plasminogen activator inhibitor type 1 (PAI-1). The extent of myofibroblastic differentiation was further investigated ultrastructurally, including immuno-electron microscopy using antibodies against MSA and SMA, focusing upon the different cell types in CMF. The expression of potential genes driving this process was quantified by Q-RT-PCR (TGF-beta1, fibronectin, its EDA splice variant, and PAI-1). Tumour cells, especially those with a spindled morphology, showed diffuse immunoreactivity for MSA, SMA, TGF-beta1, and PAI-1, while desmin, h-caldesmon, and calponin were absent. Ultrastructurally, neoplastic cells showed the presence of myofilaments and rare dense bodies, which were more prominent in spindle cells and less so in chondroblast-like cells. Immuno-electron microscopy confirmed the actin nature of these myofilaments. No fibronexus was identified. The functional activity of TGF-beta1 was demonstrated by the identification of PAI-1, a related downstream molecule both immunohistochemically as well as by Q-RT-PCR. There was a linear correlation between TGF-beta1 and PAI-1 expression. Fibronectin-EDA levels were low. We have therefore substantiated the presence of morphological, immunohistochemical, and immuno-electron microscopic partial myofibroblastic differentiation in CMF, driven by TGF-beta1 signalling.

    Topics: Actins; Adolescent; Adult; Bone Neoplasms; Calcium-Binding Proteins; Calmodulin-Binding Proteins; Calponins; Cell Transformation, Neoplastic; Child; Chondroblastoma; Chondrocytes; Desmin; Female; Fibroblasts; Fibronectins; Genes, Neoplasm; Humans; Immunohistochemistry; Male; Microfilament Proteins; Microscopy, Electron; Microscopy, Immunoelectron; Middle Aged; Muscle Proteins; Muscle, Smooth; Neoplasm Proteins; Plasminogen Activator Inhibitor 1; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factor beta1

2006
Impaired transforming growth factor beta signalling in Barrett's carcinogenesis due to frequent SMAD4 inactivation.
    Gut, 2006, Volume: 55, Issue:6

    Transforming growth factor beta (TGF-beta) is frequently involved in gastrointestinal carcinogenesis although its contribution to oesophageal adenocarcinoma (AC) and its precursor Barrett's oesophageal epithelium (BE) metaplasia are unclear.. Expression of TGF-beta signalling components was assessed by reverse transcription-polymerase chain reaction (PCR), western blot, and immunohistochemistry in oesophageal endoscopic biopsies and cell lines. Genomic alterations in SMAD4 were characterised by fluorescence in situ hybridisation, methylation specific PCR, and sequencing. Functional integrity of TGF-beta signalling was assessed by characterisation of p21 and proliferation status. Smad4 negative BIC-1 cells were transiently transfected with smad4 and TGF-beta responsiveness evaluated.. smad4 mRNA expression was progressively reduced in the metaplasia-dysplasia-adenocarcinoma sequence (p<0.01). A quarter of AC samples displayed an abnormal Smad4 protein isoform, with no corresponding changes in gene sequence or organisation. Methylation of smad4 has not been described previously but we found promoter methylation in 70% of primary AC samples. In 6/8 oesophageal cell lines, chromosomal rearrangements affected the smad4 locus. Lack of smad4 expression in BIC-1 cells occurred secondary to loss of one copy and extensive deletion of the second allele's promoter region. TGF-beta dependent induction of p21 and downregulation of minichromosome maintenance protein 2 was lost in >80% of BE and AC. TGF-beta failed to inhibit proliferation in 5/8 oesophageal cell lines. In BIC-1, the antiproliferative response was restored following transient transfection of smad4 cDNA.. In BE carcinogenesis, downregulation of Smad4 occurs due to several different mechanisms, including methylation, deletion, and protein modification. Frequent alterations in TGF-beta signalling lead to a functionally significant impairment of TGF-beta mediated growth suppression.

    Topics: Adenocarcinoma; Barrett Esophagus; Base Sequence; Cell Proliferation; Cell Transformation, Neoplastic; Disease Progression; DNA Methylation; Esophageal Neoplasms; Gene Expression Regulation, Neoplastic; Genome; Humans; Molecular Sequence Data; Neoplasm Proteins; Precancerous Conditions; Prospective Studies; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; RNA, Neoplasm; Signal Transduction; Smad4 Protein; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2006
Caveolin-1 deficiency (-/-) conveys premalignant alterations in mammary epithelia, with abnormal lumen formation, growth factor independence, and cell invasiveness.
    The American journal of pathology, 2006, Volume: 168, Issue:1

    During breast cancer development, the luminal space of the mammary acinar unit fills with proliferating epithelial cells that exhibit growth factor-independence, cell attachment defects, and a more invasive fibroblastic phenotype. Here, we used primary cultures of mammary epithelial cells derived from genetically engineered mice to identify caveolin-1 (Cav-1) as a critical factor for maintaining the normal architecture of the mammary acinar unit. Isolated cultures of normal mammary epithelial cells retained the capacity to generate mammary acini within extracellular matrix. However, those from Cav-1 (-/-) mice exhibited defects in three-dimensional acinar architecture, including disrupted lumen formation and epidermal growth factor-independent growth due to hyperactivation of the p42/44 mitogen-activated protein kinase cascade. In addition, Cav-1-null mammary epithelial cells deprived of exogenous extracellular matrix underwent a spontaneous epithelial-mesenchymal transition, with reorganization of the actin cytoskeleton, and E-cadherin redistribution. Mechanistically, these phenotypic changes appear to be caused by increases in matrix metalloproteinase-2/9 secretion and transforming growth factor-beta/Smad-2 hyperactivation. Finally, loss of Cav-1 potentiated the ability of growth factors (hepatocyte growth factor and basic fibroblast growth factor) to induce mammary acini branching, indicative of a more invasive fibroblastic phenotype. Thus, a Cav-1 deficiency profoundly affects mammary epithelia by modulating the activation state of important signaling cascades. Primary cultures of Cav-1-deficient mammary epithelia will provide a valuable new model to study the spatial/temporal progression of mammary cell transformation.

    Topics: Animals; Blotting, Western; Caveolin 1; Cell Transformation, Neoplastic; Cells, Cultured; Female; Fluorescent Antibody Technique; Growth Substances; Mammary Glands, Animal; Matrix Metalloproteinase 2; Matrix Metalloproteinase 9; Mice; Microscopy, Electron, Transmission; Neoplasm Invasiveness; Precancerous Conditions; Signal Transduction; Smad2 Protein; Transforming Growth Factor beta

2006
In vivo disruption of TGF-beta signaling by Smad7 leads to premalignant ductal lesions in the pancreas.
    Proceedings of the National Academy of Sciences of the United States of America, 2006, Feb-07, Volume: 103, Issue:6

    TGF-beta has been postulated to play an important role in the development of pancreatic cancers. More than 50% of human pancreatic cancers bear mutations of Sma- and Mad-related protein (Smad) 4, a critical protein required for TGF-beta signaling. To evaluate the in vivo function of TGF-beta in the development of pancreatic cancers, we generated a transgenic mouse model with pancreas-specific expression of Smad7, a specific inhibitor of TGF-beta signaling. Through the use of elastase I promoter, we directed the tissue specific expression of exogenous Smad7. Consistently, the exogenous Smad7 was detected only in the pancreas in the transgenic mice, and, furthermore, phosphorylation of Smad2 was blocked in the pancreatic tissues. At 6 months of age, most transgenic animals developed premalignant ductal lesions in the pancreas, with characteristics of pancreatic intraepithelial neoplasia (PanIN), a precursor to invasive pancreatic cancers. The premalignant lesions of the pancreas were accompanied by accelerated proliferation of the ductal epithelium and acinar cells, as well as increased fibrosis around the ductal lesions. This study not only demonstrated that in vivo inactivation of TGF-beta signaling is implicated in the development of early stage of pancreatic cancers, but also provided a promising animal model useful for the investigation and intervention of pancreatic cancers in humans.

    Topics: Animals; Cell Proliferation; Cell Transformation, Neoplastic; Gene Expression Regulation; Mice; Mice, Transgenic; Organ Size; Organ Specificity; Pancreas; Precancerous Conditions; Signal Transduction; Smad7 Protein; Transforming Growth Factor beta; Transgenes

2006
Tenascin C and annexin II expression in the process of pancreatic carcinogenesis.
    The Journal of pathology, 2006, Volume: 208, Issue:5

    Tenascin C (TNC) is a component of the provisional extracellular matrix (ECM) that characterizes solid tumours. Cell surface annexin II is a high-affinity receptor for large TNC splice variants. The aim of this study was to analyse whether TNC and annexin II play a role in the development of pancreatic ductal adenocarcinoma (PDAC). PDAC is characterized by a rich ECM populated by pancreatic stellate cells, which play a crucial role in pancreatic desmoplasia. The mRNA and protein levels of TNC and of annexin II were analysed in pancreatic tissues by DNA array, quantitative reverse transcriptase-polymerase chain reaction (QRT-PCR) and immunohistochemistry. TNC large splice variants were detected by RT-PCR. Enzyme linked immunosorbent assay (ELISA) was used to measure TNC levels in serum and culture supernatants. TNC and annexin II mRNA levels were significantly higher in pancreatic cancer tissues than in the normal pancreas. TNC expression was detected with increased frequency in the progression from PanIN-1 lesions to PDAC, and a parallel switch from cytoplasmic to cell surface expression of annexin II was observed. Large TNC transcripts were found in pancreatic cancer and in chronic pancreatitis, but not in the normal pancreas. TNC expression was demonstrated in pancreatic stellate cells, where it could be induced by tumour necrosis factor alpha (TNFalpha), transforming growth factor beta1 (TGF-beta1) and by cancer cell supernatants supplemented with TGF-beta1. In conclusion, the expression of TNC and cell surface annexin II increases in the progression from low-grade PanIN lesions to pancreatic cancer. Pancreatic stellate cells are identified as a source of TNC in pancreatic tissues, possibly under the influence of soluble factors released by the tumour cells.

    Topics: Adenocarcinoma; Annexin A2; Blotting, Western; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Humans; Immunoenzyme Techniques; Neoplasm Proteins; Pancreatic Neoplasms; Pancreatitis, Chronic; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; RNA, Neoplasm; Tenascin; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

2006
Expression of bone morphogenetic proteins and their associated molecules in ameloblastomas and adenomatoid odontogenic tumors.
    Oral diseases, 2006, Volume: 12, Issue:2

    To further clarify the roles of regulators of embryonic development, bone morphogenetic protein (BMPs) and their associated molecules, in oncogenesis and cytodifferentiation of odontogenic tumors, the expression of these regulator molecules were analyzed in epithelial odontogenic tumors as well as in tooth germs.. Tooth germs, ameloblastomas, adenomatoid odontogenic tumors, and malignant ameloblastomas were examined by RT-PCR and immunohistochemistry for detection of BMP-2, -4, -7, BMP receptors I and II (BMPR-I, BMPR-II), core-binding factor alpha1 (CBFA1), and osterix.. mRNA expression of BMPs, BMPRs, CBFA1, and osterix was detected in all odontogenic tissues. Immunohistochemical reactivity for BMPs, BMPRs, and CBFA1 was detected in both epithelial and mesenchymal cells of tooth germs and epithelial odontogenic tumors. BMPs and BMPRs were evidently expressed in odontogenic epithelial cells in tooth germs and epithelial odontogenic tumors. Acanthomatous ameloblastomas showed increased BMP-7 reactivity in keratinizing cells. Nuclear CBFA1 expression was detected scatteredly in odontogenic epithelial cells in normal and neoplastic odontogenic tissues, as well as in some mesenchymal cells in tooth germs and in some stromal cells in epithelial odontogenic tumors. Ameloblastic carcinomas showed low reactivity for BMPs, BMPRs, and CBFA1.. BMPs and their associated molecules might play a role in cytodifferentiation of normal and neoplastic odontogenic epithelium via epithelial-mesenchymal interactions.

    Topics: Ameloblastoma; Bone Morphogenetic Protein 2; Bone Morphogenetic Protein 4; Bone Morphogenetic Protein 7; Bone Morphogenetic Protein Receptors, Type I; Bone Morphogenetic Protein Receptors, Type II; Bone Morphogenetic Proteins; Cell Differentiation; Cell Nucleus; Cell Transformation, Neoplastic; Core Binding Factor Alpha 1 Subunit; Epithelial Cells; Epithelium; Humans; Immunohistochemistry; Mesoderm; Odontogenic Tumors; Reverse Transcriptase Polymerase Chain Reaction; Sp7 Transcription Factor; Stromal Cells; Tooth Germ; Transcription Factors; Transforming Growth Factor beta

2006
E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines.
    British journal of cancer, 2006, Mar-13, Volume: 94, Issue:5

    Using genome-wide expression profiling of a panel of 27 human mammary cell lines with different mechanisms of E-cadherin inactivation, we evaluated the relationship between E-cadherin status and gene expression levels. Expression profiles of cell lines with E-cadherin (CDH1) promoter methylation were significantly different from those with CDH1 expression or, surprisingly, those with CDH1 truncating mutations. Furthermore, we found no significant differentially expressed genes between cell lines with wild-type and mutated CDH1. The expression profile complied with the fibroblastic morphology of the cell lines with promoter methylation, suggestive of epithelial-mesenchymal transition (EMT). All other lines, also the cases with CDH1 mutations, had epithelial features. Three non-tumorigenic mammary cell lines derived from normal breast epithelium also showed CDH1 promoter methylation, a fibroblastic phenotype and expression profile. We suggest that CDH1 promoter methylation, but not mutational inactivation, is part of an entire programme, resulting in EMT and increased invasiveness in breast cancer. The molecular events that are part of this programme can be inferred from the differentially expressed genes and include genes from the TGFbeta pathway, transcription factors involved in CDH1 regulation (i.e. ZFHX1B, SNAI2, but not SNAI1, TWIST), annexins, AP1/2 transcription factors and members of the actin and intermediate filament cytoskeleton organisation.

    Topics: Breast Neoplasms; Cadherins; Cell Line, Tumor; Cell Transformation, Neoplastic; DNA Methylation; DNA Mutational Analysis; Down-Regulation; Epithelial Cells; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Mesoderm; Neoplasm Invasiveness; Polymerase Chain Reaction; Promoter Regions, Genetic; Transcription, Genetic; Transforming Growth Factor beta

2006
TGFbeta/BMP inhibits the bone marrow transformation capability of Hoxa9 by repressing its DNA-binding ability.
    The EMBO journal, 2006, Apr-05, Volume: 25, Issue:7

    Homeobox (Hox) gene mutations and their altered expressions are frequently linked to human leukemia. Here, we report that transforming growth factor beta (TGFbeta)/bone morphogenetic protein (BMP) inhibits the bone marrow transformation capability of Hoxa9 and Nup98-Hoxa9, the chimeric fusion form of Hoxa9 identified in human acute myeloid leukemia (AML), through Smad4, the common Smad (Co-Smad) in the TGFbeta/BMP signaling pathway. Smad4 interacts directly with the homeodomain of Hoxa9 and blocks the ability of Nup98-Hoxa9 to bind DNA, thereby suppressing its ability to regulate downstream gene transcription. Mapping data revealed that the amino-terminus of Smad4 mediates this interaction and overexpression of the Hoxa9 interaction domain of Smad4 was sufficient to inhibit the enhanced serial replating ability of primary bone marrow cells induced by Nup98-Hoxa9. These studies establish a novel mechanism by which TGFbeta/BMP regulates hematopoiesis and suggest that modification of Hox DNA-binding activity may serve as a novel therapeutic intervention for those leukemias that involve deregulation of Hox.

    Topics: Animals; Bone Marrow Cells; Bone Morphogenetic Proteins; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Hematopoiesis; Homeodomain Proteins; Mice; Mutation; Nuclear Pore Complex Proteins; Oncogene Proteins, Fusion; Protein Binding; Protein Structure, Tertiary; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta

2006
Cell cycle control related proteins (p53, p21, and Rb) and transforming growth factor beta (TGFbeta) in benign and carcinomatous (in situ and infiltrating) human breast: implications in malignant transformations.
    Cancer investigation, 2006, Volume: 24, Issue:2

    A comparative study of the products of the cell cycle control genes p53 (mutated form), p21, Rb (nonphosphorylated and phosphorylated form) and TGFbeta was performed by immunohistochemistry and Western blot, in benign breast disorders and breast cancer (in situ and infiltrating tumors). For the five proteins studied, the relative numbers of positively stained cells were higher in in situ carcinoma than in benign breast diseases. In infiltrating breast tumors, the relative numbers of positively stained cells were even higher than in in situ tumors except for the percentage of pRb immunostained cells, which decreased slightly in infiltrative tumors. For the other four proteins, the percentages of positively stained cases were similar to those found in in situ tumors. In the three groups of patients, TGFbeta immunoreaction appeared in the cytoplasm while immunoreactions to p53, p21, Rb, and pRb were found always in the nucleus except for p21 in in situ tumors, which showed cytoplasmic immunoreaction. Present results suggest that accumulation of mutated p53, cytoplasmic p21, and pRb in breast gland epithelium might be a crucial point in the development of in situ adenocarcinoma. In the infiltrating tumors, the expression of p21 in the nuclei and the decrease in pRb expression suggest an insufficient attempt to hinder cell proliferation.

    Topics: Adult; Aged; Blotting, Western; Breast Neoplasms; Carcinoma, Ductal, Breast; Carcinoma, Intraductal, Noninfiltrating; Cell Cycle Proteins; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; Female; Humans; Immunohistochemistry; Middle Aged; Retinoblastoma Protein; Transforming Growth Factor beta; Tumor Suppressor Protein p53

2006
Altered levels of Smad2 and Smad4 are associated with human prostate carcinogenesis.
    Prostate cancer and prostatic diseases, 2006, Volume: 9, Issue:2

    Alterations have been demonstrated in ligand and cognate receptor system of the transforming growth factor beta (TGF-beta) pathway in prostate cancer (PC). Still, little is known about changes in the activity of the intracellular Smad cascade of TGF-beta signaling during prostate carcinogenesis. We used immunohistochemistry to analyze phosphorylated Smad2 (p-Smad2), nuclear Smad4 and inhibitory-Smad7 in epithelial cells of normal, hyperplastic and malignant prostate. Specimens comprised 49 tissue cores of PC, 10 benign prostate hypertrophies and three normal prostates. Nuclear p-Smad2 (P<0.001) and nuclear Smad4 (P=0.023) were significantly decreased in PC with remarkable variations in cytoplasmic Smad7 levels. Substantial decreases in p-Smad2 and Smad4 levels were found in specimens with primary Gleason grades 3 and 4, whereas in grade 5, levels were markedly higher. Our results provide the first evidence for changes and reversible attenuation in the Smad system of the TGF-beta pathway during prostate carcinogenesis.

    Topics: Biomarkers, Tumor; Biopsy, Needle; Case-Control Studies; Cell Transformation, Neoplastic; Disease Progression; Gene Expression Regulation, Neoplastic; Humans; Immunohistochemistry; Male; Probability; Prostatic Hyperplasia; Prostatic Neoplasms; Reference Values; Sampling Studies; Sensitivity and Specificity; Smad2 Protein; Smad4 Protein; Tissue Culture Techniques; Transforming Growth Factor beta

2006
TGF-beta signaling pathway inactivation and cell cycle deregulation in the development of gastric cancer: role of the beta-spectrin, ELF.
    Biochemical and biophysical research communications, 2006, Jun-16, Volume: 344, Issue:4

    We have shown that loss of ELF, a stem cell adaptor protein, disrupts TGF-beta signaling through Smad3 and Smad4 localization. Notably elf(+/-)/smad4(+/-) mice develop gastric cancer presenting this as an important model for analyzing molecular event in gastric carcinogenesis. To gain further insight into the functional role of ELF in gastric cancer suppression, we carried out a detailed characterization of cell cycle events leading to gastric tumorigenesis. elf(-/-) cells and elf(+/-)/smad4(+/-) mice demonstrate a marked alteration of cell cycle regulators, such as Cdk4, K-Ras, and p21. Levels of Cdk4 increased compared to normal controls, suggesting loss of ELF results in functional abnormalities in cell cycle regulation. We further demonstrate that the elf(-/-) MEFs show a disruption of G1/S cell cycle transition and a significant reduction in senescence. Thus, in response to ELF deficiency, the abnormalities of G1/S checkpoint and senescence contribute their increment of susceptibility to malignant transformation.

    Topics: Aging; Animals; Cell Cycle; Cell Cycle Proteins; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase 4; Mice; Mice, Mutant Strains; Signal Transduction; Spectrin; Stomach Neoplasms; Transforming Growth Factor beta; Up-Regulation

2006
Raf plus TGFbeta-dependent EMT is initiated by endocytosis and lysosomal degradation of E-cadherin.
    Oncogene, 2006, Nov-16, Volume: 25, Issue:54

    Oncogenic Ras interferes with adhesive functions of epithelial cells, but requires tumor growth factor beta (TGFbeta) signaling to cause epithelial-mesenchymal transition (EMT) and tumor progression in model systems. To investigate the mechanisms by which Ras and TGFbeta pathways cooperate in EMT induction, we introduced a tamoxifen-inducible version of Raf-1 (RafER) into fully polarized, mammary epithelial cells (EpH4). EMT characterized by loss of E-cadherin expression and upregulation of invasiveness-promoting genes was induced by TGFbeta plus 4-hydroxytamoxifen (4HT) activation of RafER. Downregulation of E-cadherin by RafER plus TGFbeta was detectable in total cell lysates after 48 h and much earlier in detergent-insoluble fractions of E-cadherin. Both pathways cooperated to strongly enhance endocytosis of E-cadherin, mainly via the clathrin-dependent route. Pulse-chase experiments showed decreased E-cadherin protein stability in cells stimulated with TGFbeta and 4HT and increased E-cadherin half-life in the presence of monensin. Monensin and chloroquine prevented E-cadherin degradation to different extent, but only monensin effectively blocked the loss of E-cadherin from the junctional complexes. Both lysosome inhibitors caused accumulation of E-cadherin vesicles, some of which were positive for Cathepsin D and lysosome-associated membrane protein 1 (LAMP-1). In addition, TGFbeta and mitogen-activated protein kinase hyperactivation synergistically induced E-cadherin ubiquitination, suggesting that the cooperation of Raf and TGFbeta favors lysosomal degradation of E-cadherin instead of its recycling. Our data indicate that early stages of EMT involve cooperative, post-translational downregulation of E-cadherin, whereas loss of E-cadherin via transcriptional repression is a late event in EMT.

    Topics: Animals; Blotting, Northern; Blotting, Western; Cadherins; Cell Line, Tumor; Cell Transformation, Neoplastic; Down-Regulation; Endocytosis; Epithelial Cells; Fluorescent Antibody Technique; Immunoprecipitation; Lysosomes; Mice; Microscopy, Confocal; Protein Processing, Post-Translational; raf Kinases; Recombinant Fusion Proteins; Transforming Growth Factor beta

2006
Nucleophosmin/anaplastic lymphoma kinase (NPM/ALK) oncoprotein induces the T regulatory cell phenotype by activating STAT3.
    Proceedings of the National Academy of Sciences of the United States of America, 2006, Jun-27, Volume: 103, Issue:26

    The mechanisms of malignant cell transformation mediated by the oncogenic, chimeric nucleophosmin/anaplastic lymphoma kinase (NPM/ALK) tyrosine kinase remain only partially understood. Here we report that the NPM/ALK-carrying T cell lymphoma (ALK+TCL) cells secrete IL-10 and TGF-beta and express FoxP3, indicating their T regulatory (Treg) cell phenotype. The secreted IL-10 suppresses proliferation of normal immune, CD3/CD28-stimulated peripheral blood mononuclear cells and enhances viability of the ALK+TCL cells. The Treg phenotype of the affected cells is strictly dependent on NPM/ALK expression and function as demonstrated by transfection of the kinase into BaF3 cells and inhibition of its enzymatic activity and expression in ALK+TCL cells. NPM/ALK, in turn, induces the phenotype through activation of its key signal transmitter, signal transducer and activator of transcription 3 (STAT3). These findings identify a mechanism of NPM/ALK-mediated oncogenesis based on induction of the Treg phenotype of the transformed CD4(+) T cells. These results also provide an additional rationale to therapeutically target the chimeric kinase and/or STAT3 in ALK+TCL.

    Topics: CD28 Antigens; Cell Line, Tumor; Cell Transformation, Neoplastic; Forkhead Transcription Factors; Humans; Immunosuppression Therapy; Interleukin-10; Lymphoma, T-Cell; Oncogene Proteins, Fusion; Protein-Tyrosine Kinases; Receptors, IgE; STAT3 Transcription Factor; T-Lymphocytes, Regulatory; Transforming Growth Factor beta

2006
Oncogenic function of a novel WD-domain protein, STRAP, in human carcinogenesis.
    Cancer research, 2006, Jun-15, Volume: 66, Issue:12

    The development and progression of malignancies is a complex multistage process that involves the contribution of a number of genes giving growth advantage to cells when transformed. The role of transforming growth factor-beta (TGF-beta) in carcinogenesis is complex with tumor-suppressor or prooncogenic activities depending on the cell type and the stage of the disease. We have previously reported the identification of a novel WD-domain protein, STRAP, that associates with both TGF-beta receptors and that synergizes with the inhibitory Smad, Smad7, in the negative regulation of TGF-beta-induced transcription. Here, we show that STRAP is ubiquitously expressed and is localized in both cytoplasm and nucleus. STRAP is up-regulated in 60% colon and in 78% lung carcinomas. Stable expression of STRAP results in activation of mitogen-activated protein kinase/extracellular signal-regulated kinase pathway and in down-regulation of the cyclin-dependent kinase inhibitor p21(Cip1), which results in retinoblastoma protein hyperphosphorylation. In addition, we have observed that Smad2/3 phosphorylation, TGF-beta-mediated transcription, and growth inhibition are induced in STRAP-knockout mouse embryonic fibroblasts compared with wild-type cells. Ectopic expression of STRAP in A549 lung adenocarcinoma cell line inhibits TGF-beta-induced growth inhibition and enhances anchorage-independent growth of these cells. Moreover, overexpression of STRAP increases tumorigenicity in athymic nude mice. Knockdown of endogenous STRAP by small interfering RNA increases TGF-beta signaling, reduces ERK activity, increases p21(Cip1) expression, and decreases tumorigenicity. Taken together, these results suggest that up-regulation of STRAP in human cancers may provide growth advantage to tumor cells via TGF-beta-dependent and TGF-beta-independent mechanisms, thus demonstrating the oncogenic function of STRAP.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Cell Transformation, Neoplastic; Chlorocebus aethiops; Colorectal Neoplasms; COS Cells; Cyclin-Dependent Kinase Inhibitor p21; Enzyme Activation; Gene Expression Regulation, Neoplastic; Humans; Lung Neoplasms; MAP Kinase Signaling System; Mice; Mink; Mitogen-Activated Protein Kinase Kinases; Neoplasm Proteins; NIH 3T3 Cells; Oncogenes; Phosphorylation; Proteins; Retinoblastoma Protein; RNA-Binding Proteins; Transforming Growth Factor beta

2006
TGF-beta inhibits Akt-induced transformation in intestinal epithelial cells.
    Surgery, 2006, Volume: 140, Issue:2

    During the early stages of colorectal carcinogenesis, the phosphatidylinositol 3-kinase (PI3K)/Akt pathway is activated, enabling the transformed cells to survive and grow in the absence of anchorage to extracellular matrix. Transforming growth factor beta (TGF-beta) is an important tumor suppressor in the colon, and it is inactivated during later stages of colorectal carcinogenesis. The purpose of this study was to determine whether TGF-beta inhibits Akt-induced anchorage-independent growth and resistance to anoikis in gut epithelial cells.. Rat intestinal epithelial cells (RIE-1) were infected with a retrovirus containing pLXSN-mAkt, and three independent clones were selected. Anchorage-independent growth was examined by colony formation in soft agar and cell counting in ultralow attachment plates. Anoikis was analyzed with the use of Annexin V staining.. All three clones of RIE-1/mAkt formed colonies in soft agar, which were decreased by TGF-beta. TGF-beta induced anoikis and treatment with a general caspase inhibitor, zVAD-fluoromethyl ketone, blocked TGF-beta-mediated decrease in colony formation.. TGF-beta attenuated Akt-induced anchorage-independent growth in RIE-1 cells in part by enhancing anoikis. Our data demonstrate a novel tumor-suppressor activity of TGF-beta and provide the molecular justification for the required activation of the PI3K/Akt pathway and the subsequent inactivation of TGF-beta signaling during colorectal carcinogenesis.

    Topics: Animals; Apoptosis; Cell Culture Techniques; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Epithelial Cells; Intestinal Mucosa; Proto-Oncogene Proteins c-akt; Rats; Transforming Growth Factor beta

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
JNK mediates TGF-beta1-induced epithelial mesenchymal transdifferentiation of mouse transformed keratinocytes.
    FEBS letters, 2006, Oct-02, Volume: 580, Issue:22

    In this study we analyzed the role of the c-Jun N-terminal kinases (JNK) pathway in the TGF-beta1 stimulation of urokinase-type plasminogen activator (uPA), initial stages of epithelial-mesenchymal transdifferentiation (EMT) and cell migration. TGF-beta1 induces JNK phosphorylation, c-Jun transactivation and AP1 activation. The involvement of JNK was evaluated using dominant negative mutants SEK-1 AL, JNK and cJun, depletion of JNK1,2 proteins by treatment of cells with antisense oligonucleotides, as well as the chemical inhibitor SP600125. Our results demonstrated that the JNK pathway is required in the TGF-beta1 enhancement of uPA, fibronectin, E-cadherin delocalization, actin re-organization and vimentin expression, concomitant with the induction of cell migration. These results allow us to suggest a role of JNK in the TGF-beta1 induction of EMT in relation with the stimulation of malignant properties of mouse transformed keratinocytes.

    Topics: Animals; Cadherins; Cell Differentiation; Cell Line, Transformed; Cell Movement; Cell Transformation, Neoplastic; Fibronectins; Gene Expression Regulation; Keratinocytes; MAP Kinase Kinase 4; Mice; Mitogen-Activated Protein Kinase 8; Mitogen-Activated Protein Kinase 9; Protein Transport; Proto-Oncogene Proteins c-jun; Signal Transduction; Transcriptional Activation; Transforming Growth Factor beta; Transforming Growth Factor beta1; Urokinase-Type Plasminogen Activator; Vimentin

2006
NSAID activated gene (NAG-1), a modulator of tumorigenesis.
    Journal of biochemistry and molecular biology, 2006, Nov-30, Volume: 39, Issue:6

    The NSAID activated gene (NAG-1), a member of the TGF-beta superfamily, is involved in tumor progression and development. The over-expression of NAG-1 in cancer cells results in growth arrest and increase in apoptosis, suggesting that NAG-1 has anti-tumorigenic activity. This conclusion is further supported by results of experiments with transgenic mice that ubiquitously express human NAG-1. These transgenic mice are resistant to the development of intestinal tumors following treatment with azoxymethane or by introduction of a mutant APC gene. In contrast, other data suggest a pro-tumorigenic role for NAG-1, for example, high expression of NAG-1 is frequently observed in tumors. NAG-1 may be like other members of the TGF-beta superfamily, acting as a tumor suppressor in the early stages, but acting pro-tumorigenic at the later stages of tumor progression. The expression of NAG-1 can be increased by treatment with drugs and chemicals documented to prevent tumor formation and development. Most notable is the increase in NAG-1 expression by the inhibitors of cyclooxygenases that prevent human colorectal cancer development. The regulation of NAG-1 is complex, but these agents act through either p53 or EGR-1 related pathways. In addition, an increase in NAG-1 is observed in inhibition of the AKT/GSK-3beta pathway, suggesting NAG-1 alters cell survival. Thus, NAG-1 expression is regulated by tumor suppressor pathways and appears to modulate tumor progression.

    Topics: Animals; Anti-Inflammatory Agents, Non-Steroidal; Carcinogenicity Tests; Cell Transformation, Neoplastic; Cytokines; Gene Expression Regulation; Genes, Tumor Suppressor; Growth Differentiation Factor 15; Humans; Male; Mice; Mice, Transgenic; Prostatic Neoplasms; Transforming Growth Factor beta

2006
Cutaneous T-cell lymphoma: malignant proliferation of T-regulatory cells.
    Blood, 2005, Feb-15, Volume: 105, Issue:4

    Studies in an in vitro model of cutaneous T-cell lymphoma (CTCL) demonstrated that CTCL cell proliferation is stimulated by direct contact with autologous, immature dendritic cells (DCs), suggesting that CD4(+) CTCL cell division is driven by antigens presented by DC major histocompatibility complex (MHC) class 2. We now report that the T-cell receptor (TCR) of the CD4(+) CTCL cells is triggered after interaction with DCs loaded with apoptotic CTCL cells, as shown by reduced membrane expression of CD3 and the TCR, up-regulation of cytotoxic T lymphocyte antigen-4 (CTLA-4), and calcium mobilization. CTCL cells adopt a T-regulatory (Treg) phenotype expressing CD25/CTLA-4 and FoxP3 and secreting interleukin-10 (IL-10) and transforming growth factor-beta (TGF-beta). Treg CTCL cells suppress normal T-cell antigen-driven secretion of IL-2 and interferon-gamma (IFN-gamma). Blocking DC MHC class 2 expression or transport inhibited CTCL cell adoption of a Treg phenotype. Allogeneic CTCL cells or normal CD4 T cells served as sources of apoptotic material for CTCL cell conversion to a Treg phenotype. Conversion of CTCL cells to Treg cells may explain the anergic, immunosuppressive nature of the malignancy.

    Topics: Antibodies, Blocking; Antigens; Antigens, CD; Antigens, Differentiation; Apoptosis; Calcium; Cell Proliferation; Cell Transformation, Neoplastic; CTLA-4 Antigen; Cytokines; Dendritic Cells; Dose-Response Relationship, Immunologic; Histocompatibility Antigens Class II; Humans; Immunophenotyping; Interleukin-10; Lymphocyte Activation; Lymphoma, T-Cell, Cutaneous; Skin Neoplasms; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Cells, Cultured

2005
Transforming growth factor (TGF)-beta in conjunction with H-ras activation promotes malignant progression of MCF10A breast epithelial cells.
    Cytokine, 2005, Jan-21, Volume: 29, Issue:2

    To address how transforming growth factor (TGF)-beta and oncogenic H-ras signal transduction pathways interact with each other in the malignant progression of breast epithelial cells, we investigated the role of TGF-beta signaling pathway in invasive and migrative properties of H-ras-transformed MCF10A human breast epithelial cells in this study. Here we show that TGF-beta treatment significantly enhanced invasion and migration of H-ras MCF10A cells. H-ras-mediated activation of p38 MAPK and ERK-1/2 was stimulated by TGF-beta. TGF-beta increased expression of matrix metalloproteinase (MMP)-2 through transcriptional activation while TGF-beta-stimulated MMP-9 up-regulation did not occur at transcription level. Activation of p38 MAPK pathway was required for TGF-beta-induced cell migration, invasion and MMP-2/-9 up-regulation, indicating a critical role of p38 MAPK signaling in TGF-beta-promoted tumor progression of H-ras-activated cells. ERKs signaling was also crucial for TGF-beta-enhanced invasive and migrative phenotypes but the up-regulation of MMP-2/-9 was not dependent on ERKs activity. Taken together, we show that TGF-beta promotes H-ras-mediated cell migration and invasive phenotypes in which p38 MAPK and ERKs signaling pathways are involved. Our findings revealing how H-ras and TGF-beta signal pathways interact with each other in MCF10A human breast cells may provide an insight into molecular mechanisms for contribution of TGF-beta to a malignant progression of breast cancer in collaboration with activated H-ras.

    Topics: Cell Line; Cell Transformation, Neoplastic; Enzyme Activation; Gene Expression Regulation; Genes, ras; Humans; Mammary Glands, Human; Mitogen-Activated Protein Kinases; Transforming Growth Factor beta

2005
Acceleration of Smad2 and Smad3 phosphorylation via c-Jun NH(2)-terminal kinase during human colorectal carcinogenesis.
    Cancer research, 2005, Jan-01, Volume: 65, Issue:1

    Conversion of normal epithelial cells to tumors is associated with a shift in transforming growth factor-beta (TGF-beta) function: reduction of tumor suppressor activity and increase of oncogenic activity. However, specific mechanisms of this functional alteration during human colorectal carcinogenesis remain to be elucidated. TGF-beta signaling involves Smad2/3 phosphorylated at linker regions (pSmad2/3L) and COOH-terminal regions (pSmad2/3C). Using antibodies specific to each phosphorylation site, we herein showed that Smad2 and Smad3 were phosphorylated at COOH-terminal regions but not at linker regions in normal colorectal epithelial cells and that pSmad2/3C were located predominantly in their nuclei. However, the linker regions of Smad2 and Smad3 were phosphorylated in 31 sporadic colorectal adenocarcinomas. In particular, late-stage invasive and metastatic cancers typically showed a high degree of phosphorylation of Smad2/3L. Their extent of phosphorylation in 11 adenomas was intermediate between those in normal epithelial cells and adenocarcinomas. Whereas pSmad2L remained in the cytoplasm, pSmad3L was located exclusively in the nuclei of Ki-67-immunoreactive adenocarcinomas. In contrast, pSmad3C gradually decreased as the tumor stage progressed. Activated c-Jun NH(2)-terminal kinase in cancers could directly phosphorylate Smad2/3L. Although Mad homology 2 region sequencing in the Smad4 gene revealed a G/A substitution at codon 361 in one adenocarcinoma, the mutation did not correlate with phosphorylation. No mutations in the type II TGF-beta receptor and Smad2 genes were observed in the tumors. In conclusion, pSmad3C, which favors tumor suppressor activity of TGF-beta, was found to decrease, whereas c-Jun NH(2)-terminal kinase tended to induce the phosphorylation of Smad2/3L in human colorectal adenoma-carcinoma sequence.

    Topics: Adenocarcinoma; Binding Sites; Cell Transformation, Neoplastic; Colorectal Neoplasms; DNA-Binding Proteins; Humans; JNK Mitogen-Activated Protein Kinases; Neoplasm Staging; Phosphorylation; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Smad2 Protein; Smad3 Protein; Trans-Activators; Transforming Growth Factor beta

2005
Integration of Ras subeffector signaling in TGF-beta mediated late stage hepatocarcinogenesis.
    Carcinogenesis, 2005, Volume: 26, Issue:5

    Immortalized p19(ARF) null hepatocytes (MIM) feature a high degree of functional differentiation and are susceptible to transforming growth factor (TGF)-beta driven growth arrest and apoptosis. In contrast, polarized MIM hepatocytes expressing hyperactive Ha-Ras continue proliferation in cooperation with TGF-beta, and adopt an invasive phenotype by executing an epithelial to mesenchymal transition (EMT). In this study, we analyzed the involvement of Ras subeffectors in TGF-beta mediated hepatocellular EMT by employing MIM hepatocytes, which express Ras mutants allowing selective activation of either mitogen-activated protein kinase (MAPK) signaling (V12-S35) or phosphoinositide 3-OH (PI3)3 kinase (PI3K) signaling (V12-C40). We found that MAPK signaling in MIM-S35 hepatocytes was necessary and sufficient to promote resistance to TGF-beta mediated inhibition of proliferation in vitro and in vivo. MIM-S35 hepatocytes showed also PI3K activation during EMT, however, MAPK signaling on its own protected hepatocytes from apoptosis. Yet, MIM-C40 hepatocytes failed to form tumors and required additional MAPK stimulation to overcome TGF-beta mediated growth arrest. In vivo, the collaboration of MAPK signaling and TGF-beta activity drastically accelerated the cell-cycle progression of the hepatocytes, leading to vast tumor formation. From these data we conclude that MAPK is crucial for the cooperation with TGF-beta to regulate the proliferation as well as the survival of hepatocytes during EMT, and causes the fatal increase in hepatocellular tumor progression.

    Topics: Animals; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Humans; Mice; Mitogen-Activated Protein Kinases; Neoplasm Invasiveness; ras Proteins; Signal Transduction; Transforming Growth Factor beta

2005
Cancer. An anchor for tumor cell invasion.
    Science (New York, N.Y.), 2005, Mar-18, Volume: 307, Issue:5716

    Topics: Animals; Carcinoma, Squamous Cell; Cell Adhesion Molecules; Cell Transformation, Neoplastic; Collagen Type VII; Disease Susceptibility; Epidermolysis Bullosa Dystrophica; Genes, ras; Humans; I-kappa B Proteins; Kalinin; Keratinocytes; Mice; Mutation; Neoplasm Invasiveness; NF-KappaB Inhibitor alpha; Protein Structure, Tertiary; Skin Neoplasms; Transduction, Genetic; Transforming Growth Factor beta

2005
Mouse p10, an alternative spliced form of p15INK4b, inhibits cell cycle progression and malignant transformation.
    Cancer research, 2005, Apr-15, Volume: 65, Issue:8

    The INK4 family of proteins negatively regulates cell cycle progression at the G(1)-S transition by inhibiting cyclin-dependent kinases. Two of these cell cycle inhibitors, p16(INK4A) and p15(INK4B), have tumor suppressor activities and are inactivated in human cancer. Interestingly, both INK4 genes express alternative splicing variants. In addition to p16(INK4A), the INK4A locus encodes a splice variant, termed p12--specifically expressed in human pancreas--and ARF, a protein encoded by an alternative reading frame that acts as a tumor suppressor through the p53 pathway. Similarly, the human INK4B locus encodes the p15(INK4B) tumor suppressor and one alternatively spliced form, termed as p10. We show here that p10, which arises from the use of an alternative splice donor site within intron 1, is conserved in the mouse genome and is widely expressed in mouse tissues. Similarly to mouse p15(INK4B), p10 expression is also induced by oncogenic insults and transforming growth factor-beta treatment and acts as a cell cycle inhibitor. Importantly, we show that mouse p10 is able to induce cell cycle arrest in a p53-dependent manner. We also show that mouse p10 is able to inhibit foci formation and anchorage-independent growth in wild-type mouse embryonic fibroblasts, and that these antitransforming properties of mouse p10 are also p53-dependent. These results indicate that the INK4B locus, similarly to INK4A-ARF, harbors two different splicing variants that can be involved in the regulation of both the p53 and retinoblastoma pathways, the two major molecular pathways in tumor suppression.

    Topics: Alternative Splicing; Amino Acid Sequence; Animals; Base Sequence; Cell Cycle; Cell Cycle Proteins; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p15; Genes, ras; Mice; Molecular Sequence Data; NIH 3T3 Cells; Protein Isoforms; Retinoblastoma Protein; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Tumor Suppressor Proteins

2005
Induction of an epithelial to mesenchymal transition in human immortal and malignant keratinocytes by TGF-beta1 involves MAPK, Smad and AP-1 signalling pathways.
    Journal of cellular biochemistry, 2005, Aug-01, Volume: 95, Issue:5

    Recent data indicate that transforming growth factor-beta1 (TGF-beta1) can act to promote tumour progression in the late stages of carcinogenesis. The mechanism by which this occurs is unknown although a ligand-induced epithelial-mesenchymal transition (EMT) is thought to be important. In this study, we demonstrate that active Ras is required for TGF-beta1-induced EMT in human keratinocytes and that epidermal growth factor (EGF) can substitute for mutant Ras. EMT was reversed by the removal of TGF-beta1. Under conditions of TGF-beta1-induced EMT, cells were growth inhibited by the ligand resulting in G1 arrest. In cells containing normal Ras, TGF-beta1-activated ERK and p38 mitogen-activated protein kinases (MAPKs), and levels of activation were further increased by co-treatment with EGF. Inhibition of MAPK pathways and Smad2/3 signalling blocked the induction of EMT by TGF-beta1. Further, inhibition of the AP-1 transcriptional complex by [6]-Gingerol, or by the ectopic expression of JDP2, blocked TGF-beta1-induced EMT and conversely, stimulation of AP-1 by 12-O-tetradecanoylphorbol 13-acetate (TPA) substituted for EGF in the induction of EMT by TGF-beta1 in cells containing normal Ras. The presence of oncogenic Ras, the treatment of cells with EGF, or the treatment of cells with TPA to activate AP-1, potentiated TGF-beta1-induced Smad-dependent transcription, an effect that was attenuated by the inhibition of MAPKs and AP-1. The results demonstrate that active Ras and TGF-beta1 co-operate to reversibly induce EMT in human keratinocytes by mechanisms that involve MAPKs, Smad2/3 and AP-1. Further we demonstrate that MAPK/AP-1 signalling enhances Smad transcriptional activity under conditions associated with TGF-beta1-induced EMT.

    Topics: Blotting, Western; Catechols; Cell Cycle; Cell Proliferation; Cell Transformation, Neoplastic; DNA-Binding Proteins; Epithelial Cells; Fatty Alcohols; Genes, ras; Humans; Keratinocytes; Luciferases; Mesoderm; Mutagens; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Plasmids; Signal Transduction; Skin Neoplasms; Smad3 Protein; Trans-Activators; Transcription Factor AP-1; Transforming Growth Factor beta; Transforming Growth Factor beta1; Zingiber officinale

2005
Oligomerization of Evi-1 regulated by the PR domain contributes to recruitment of corepressor CtBP.
    Oncogene, 2005, Sep-08, Volume: 24, Issue:40

    Evi-1 is a transcription factor that is implicated in leukemic transformation of hematopoietic cells. Two distinct alternative forms, Evi-1a and Evi-1c, are generated from the EVI-1 gene. Whereas Evi-1a is widely recognized as an oncoprotein, a role for Evi-1c, which has an additional PR domain in the amino-terminus of Evi-1a, in leukemogenesis, has not been elucidated thus far. Aberrant oligomerization of transcription factors has recently emerged as a prevalent mechanism for activating their oncogenic potential in hematopoietic malignancies. Here, to study the mechanisms that underlie Evi-1-mediated oncogenesis, we investigated formation of oligomeric complexes by the Evi-1 proteins. We show that Evi-1a forms homo-oligomers, whereas Evi-1c exclusively exists as a monomer in mammalian cells. Remarkably, Evi-1c has lost the ability to interact with CtBP, a transcriptional corepressor that associates with Evi-1a. As a consequence, the ability of Evi-1c to repress transforming growth factor-beta (TGF-beta) signaling is significantly abrogated. These results identify a novel function of a PR domain to regulate oligomerization of transcription factors and suggest that homo-oligomerization may play a critical role in corepressor recruitment by the Evi-1 proteins. In addition, we found that the chimeric oncoprotein acute myelocytic leukemia (AML)1-Evi-1, generated in t(3;21) leukemia, also forms homo-oligomers and hetero-oligomers with Evi-1a, while it did not interact with Evi-1c. Consistent with the results, repression of TGF-beta by AML1-Evi-1 was significantly enhanced by Evi-1a, whereas it was hardly affected by the presence of Evi-1c. These results suggest that oligomerization may contribute to the oncogenic potential of Evi-1-containing proteins.

    Topics: Alcohol Oxidoreductases; Amino Acid Motifs; Animals; Cell Line; Cell Transformation, Neoplastic; DNA-Binding Proteins; Humans; MDS1 and EVI1 Complex Locus Protein; Phosphoproteins; Protein Binding; Protein Structure, Tertiary; Proto-Oncogenes; Recombinant Fusion Proteins; Signal Transduction; Transcription Factors; Transforming Growth Factor beta; Tumor Cells, Cultured

2005
Gene expression profiling of cancer progression reveals intrinsic regulation of transforming growth factor-beta signaling in ErbB2/Neu-induced tumors from transgenic mice.
    Oncogene, 2005, Aug-04, Volume: 24, Issue:33

    Upregulation of HER2/ErbB2/Neu occurs in 15-30% of human breast cancers and correlates with poor prognosis. Identification of ErbB2/Neu transcriptional targets should facilitate development of novel therapeutic approaches. Development of breast cancer is a multistep process; thus, to identify the transcriptomes associated with different stages of progression of tumorigenesis, we compared expression profiles of mammary tumors and preneoplastic mammary tissue from MMTV-Neu transgenic mice to expression profiles of wild-type mammary glands using Affymetrix microarrays. We identified 324 candidate genes that were unique to ErbB2/Neu-induced tumors relative to normal mammary gland tissue from wild-type controls. Expression of a subset of these genes (82) was also changed in the preneoplastic mammary glands compared to wild-type controls, indicating that they may play a pivotal role during early events of ErbB2/Neu-initiated mammary tumorigenesis. Further analysis of the microarray data revealed that expression of several known transforming growth factor (TGF)-beta target genes was altered, suggesting that the TGF-beta signaling cascade is downregulated in ErbB2/Neu-induced tumors. Western blot analysis for TGF-beta-Receptor-I/ALK5 and immunohistochemistry for TGF-beta-Receptor-I/ALK5 and phosphorylated/activated Smad2 confirmed that the Smad-dependent TGF-beta signaling cascade was inactive in these tumors. Although absent in most of the tumor, phosphorylated Smad2 was present in the periphery of tumors. Interestingly, presence of phosphorylated/activated Smad2 correlated with expression of Activin-Receptor-IB/ALK4, suggesting that although Smad-dependent TGF-beta signaling is absent in ErbB2/Neu-induced tumors, Activin signaling may be active at the leading edge of these tumors. Cumulatively, these data indicate that the TGF-beta pathway is intrinsically suppressed in ErbB2/Neu tumors via a mechanism involving loss of TGF-beta-Receptor-I/ALK5.

    Topics: Activin Receptors; Animals; Cell Transformation, Neoplastic; Disease Progression; DNA-Binding Proteins; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Mammary Neoplasms, Animal; Mice; Mice, Transgenic; Oligonucleotide Array Sequence Analysis; Precancerous Conditions; Receptor, ErbB-2; Signal Transduction; Smad2 Protein; Trans-Activators; Transcription, Genetic; Transforming Growth Factor beta; Up-Regulation

2005
Suprabasal alpha 5 beta1 integrin expression stimulates formation of epidermal squamous cell carcinomas without disrupting TGFbeta signaling or inducing spindle cell tumors.
    Molecular carcinogenesis, 2005, Volume: 44, Issue:1

    During epidermal chemical carcinogenesis benign papillomas convert to squamous cell carcinomas, some of which undergo epithelial-mesenchymal conversion to highly malignant spindle cell tumors. TGFbeta inhibits early stages of carcinogenesis but promotes the spindle cell phenotype in later stages. One hallmark of spindle cell tumors is upregulation of the alpha 5 beta 1 integrin fibronectin receptor. To examine the significance of altered alpha 5 beta1 integrin expression, we induced tumors in transgenic mice expressing alpha 5 beta1 in the suprabasal epidermal layers. Invalpha 5 beta1 mice developed threefold more papillomas and squamous cell carcinomas than wild-type (Wt) littermates; however, no spindle cell tumors or increased metastases were observed. Suprabasal expression of the alpha 6 beta 4 integrin increases squamous cell carcinoma formation and decreases TGFbeta sensitivity while alpha 3 beta1 may have the opposite effect. In contrast, nuclear phosphoSmad2 labeling in Invalpha 5 beta1 epidermis and tumors was indistinguishable from Wt, and suprabasal alpha 5 beta1 did not block TGFbeta-induced Smad2/3 translocation or growth inhibition in cultured keratinocytes. We conclude that upregulation of alpha 5 beta1 does not predispose the epidermis to undergo conversion to spindle cell tumors and that the mechanism by which alpha 5 beta1 influences susceptibility to carcinogenesis is independent of perturbed TGFbeta signaling.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Integrin alpha5beta1; Mice; Mice, Transgenic; Phosphorylation; Protein Transport; Signal Transduction; Smad2 Protein; Smad3 Protein; Trans-Activators; Transforming Growth Factor beta

2005
TGF-{beta} maintains dormancy of prostatic stem cells in the proximal region of ducts.
    The Journal of cell biology, 2005, Jul-04, Volume: 170, Issue:1

    We have previously shown that prostatic stem cells are located in the proximal region of mouse prostatic ducts. Here, we show that this region responds differently to transforming growth factor (TGF)-beta than the distal ductal region and that under physiological conditions androgens and TGF-beta are crucial overall regulators of prostatic tissue homeostasis. This conclusion is supported by the observations showing that high levels of TGF-beta signaling are present in the quiescent proximal region of ducts in an androgen-replete animal and that cells in this region overexpress Bcl-2, which protects them from apoptosis. Moreover, androgen ablation reverses the proximal-distal TGF-beta signaling gradient, leading to an increase in TGF-beta signaling in the unprotected distal region (low Bcl-2 expression). This reversal of TGF-beta-mediated signaling accompanies apoptosis of cells in the distal region and gland involution after androgen withdrawal. A physiological TGF-beta signaling gradient (high proximally and low distally) and its functional correlates are restored after androgen replenishment. In addition to highlighting the regulatory role of androgens and TGF-beta, these findings may have important implications for the deregulation of the stem cell compartment in the etiology of proliferative prostatic diseases.

    Topics: Androgens; Animals; Apoptosis; Carcinoma; Cell Differentiation; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Coculture Techniques; Epithelial Cells; Male; Mice; Mice, Inbred C57BL; Prostate; Prostatic Neoplasms; Proto-Oncogene Proteins c-bcl-2; Signal Transduction; Stem Cells; Transforming Growth Factor beta

2005
RAS and TGF-beta exert antagonistic effects on extracellular matrix gene expression and fibroblast transformation.
    Oncogene, 2005, Oct-27, Volume: 24, Issue:47

    Ras, Raf, and Fos function as components in a signal transduction pathway that is constitutively active in many cancers. Many of the changes that underlie cell transformation arise through changes in gene expression. We have used gene expression profiling of 3T3 cells transformed by Ras, Raf, and Fos to define the common and distinct targets of transcriptional control by each of these oncogenes. In this analysis, the most strongly conserved feature of cell transformation at the transcriptional level is the transcriptional repression of genes that encode components of the extracellular matrix (ECM). TGF-beta treatment of fibroblasts is known to increase production of ECM, suggesting that TGF-beta might selectively reverse some of the gene expression changes that occur during cell transformation. Using gene expression profiling of the TGF-beta response, we show that the ability of TGF-beta to reverse the changes in gene expression brought about by cellular transformation is essentially confined to genes that encode components of the ECM and the cytoskeleton. This selective reversal of transformation-induced changes in gene expression is associated with partial reversal of many parameters of cell transformation. The results demonstrate a correlation between gene repression by the Ras/Raf/ERK signaling pathway, gene activation by the TGF-beta signaling pathway, and the transformed phenotype in fibroblasts.

    Topics: Animals; Blotting, Northern; Blotting, Western; Cell Transformation, Neoplastic; Cells, Cultured; Extracellular Matrix; Fibroblasts; Fluorescent Antibody Technique; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Genes, ras; Mice; Oligonucleotide Array Sequence Analysis; Proto-Oncogene Proteins c-raf; RNA, Messenger; Signal Transduction; Transcriptional Activation; Transforming Growth Factor beta

2005
Hepatitis C virus core variants isolated from liver tumor but not from adjacent non-tumor tissue interact with Smad3 and inhibit the TGF-beta pathway.
    Oncogene, 2005, Sep-08, Volume: 24, Issue:40

    Hepatitis C virus (HCV) is a major risk factor for human hepatocellular carcinoma (HCC) but the mechanisms underlying HCV-induced carcinogenesis are still poorly understood. We have hypothesized that viral variants, selected during long-term infection, might contribute to cellular transformation. To address this issue, we have investigated the effect of natural HCV core variants isolated from liver tumors (T), or their non-tumor (NT) counterparts, on the tumor growth factor-beta (TGF-beta) pathway, a major regulator of cellular proliferation, differentiation and apoptosis. We have found a significant reduction in TGF-beta reporter gene activity with the expression of core sequences isolated from liver tumors. In contrast, moderate or no effects were observed with non-tumor mutants or a core reference sequence. The molecular mechanisms have been characterized and involved the inhibition, by tumor-derived cores, of the DNA-binding activity of the Smad3/4 transcription factors complex. This inhibition occurs through a direct interaction between the central domain (amino acids 59-126) of tumor-derived core and the MH1 DNA-binding domain of Smad3, thus preventing its binding to DNA. We have therefore identified a new cell-signaling pathway targeted by HCV core and inhibited by tumor-derived core sequences. These results suggest that during chronic infection, there is selection of viral variants that may promote cell transformation by providing, to clonally expanding cells, resistance to TGF-beta antiproliferative effects.

    Topics: Amino Acid Sequence; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; DNA-Binding Proteins; Hepacivirus; Humans; Liver Neoplasms; Molecular Sequence Data; Mutation; Protein Binding; Protein Structure, Tertiary; Signal Transduction; Smad3 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta; Tumor Cells, Cultured; Viral Core Proteins

2005
[Regulatory effect of Smad7 gene on MAPK signal pathway in malignant transformation of immortalized human bronchial epithelial BEP2D cells].
    Ai zheng = Aizheng = Chinese journal of cancer, 2005, Volume: 24, Issue:9

    Smad7 is an inhibitor of transforming growth factor-beta (TGF-beta) signal pathway. TGF-beta could induce the expression of several genes through activating SMAD and ras/MEK/ERK pathways. This study was to determine whether Smad7 is involved in regulating mitogen-activated protein kinase (MAPK) signal pathway with TGF-beta in malignant transformation of human bronchial epithelial BEP2D cells.. Immortalized BEP2D cells and malignant BERP35T2 cells were co-transfected with full-length Smad7 cDNA constructed pCISmad7.neo or Smad7 siRNA, transactivator vector pTet-Elk or pTet-Jun, and reporter vector pTRE-Luc, and stimulated with TGF-beta. The regulatory effect of Smad7 on MAPK signal pathway was investigated by standard luciferase assay.. In BEP2D cells, when treated with TGF-beta1, phosphorylated activities of Elk and Jun were up-regulated (P(Elk)=0.033, P(Jun)=0.016); after co-transfection of Elk or Jun with pCISmad7.neo, phosphorylated activity of Elk was increased, and that of Jun was decreased (P(Elk)=0.017, P(Jun)=0.028); after co-transfection of Elk or Jun with Smad7 siRNA, phosphorylated activity of Elk was decreased, and that of Jun was increased (P(Elk)=0.018, P(Jun)=0.005). In BERP35T2 cells, when treated with TGF-beta1, phosphorylated activity of Elk was up-regulated (P=0.006); after co-transfection of Elk and Smad7 siRNA, phosphorylated activity of Elk was decreased (P=0.000); no activity of Jun was detected in BERP35T2 cells.. In the process of malignant transformation of BEP2D cells, the intervention of Smad7 in MAPK signal pathway leads to the activity imbalance between extracellular signal-related protein kinase (ERK) and c-Jun NH2-terminal kinase (JNK), which in turn promotes cell proliferation. All these could contribute to further malignant transformation of these cells.

    Topics: Bronchi; Cell Transformation, Neoplastic; Cells, Cultured; Epithelial Cells; ets-Domain Protein Elk-1; Humans; MAP Kinase Signaling System; Mitogen-Activated Protein Kinases; Phosphorylation; Proto-Oncogene Proteins c-jun; RNA, Small Interfering; Smad7 Protein; Transfection; Transforming Growth Factor beta

2005
The use of cystatin C to inhibit epithelial-mesenchymal transition and morphological transformation stimulated by transforming growth factor-beta.
    Breast cancer research : BCR, 2005, Volume: 7, Issue:5

    Transforming growth factor-beta (TGF-beta) is a potent suppressor of mammary epithelial cell (MEC) proliferation and is thus an inhibitor of mammary tumor formation. Malignant MECs typically evolve resistance to TGF-beta-mediated growth arrest, enhancing their proliferation, invasion, and metastasis when stimulated by TGF-beta. Recent findings suggest that therapeutics designed to antagonize TGF-beta signaling may alleviate breast cancer progression, thereby improving the prognosis and treatment of breast cancer patients. We identified the cysteine protease inhibitor cystatin C (CystC) as a novel TGF-beta type II receptor antagonist that inhibits TGF-beta binding and signaling in normal and cancer cells. We hypothesized that the oncogenic activities of TGF-beta, particularly its stimulation of mammary epithelial-mesenchymal transition (EMT), can be prevented by CystC.. Retroviral infection was used to constitutively express CystC or a CystC mutant impaired in its ability to inhibit cathepsin protease activity (namely Delta14CystC) in murine NMuMG MECs and in normal rat kidney (NRK) fibroblasts. The effect of recombinant CystC administration or CystC expression on TGF-beta stimulation of NMuMG cell EMT in vitro was determined with immunofluorescence to monitor rearrangements of actin cytoskeletal architecture and E-cadherin expression. Soft-agar growth assays were performed to determine the effectiveness of CystC in preventing TGF-beta stimulation of morphological transformation and anchorage-independent growth in NRK fibroblasts. Matrigel invasion assays were performed to determine the ability of CystC to inhibit NMuMG and NRK motility stimulated by TGF-beta.. CystC and Delta14CystC both inhibited NMuMG cell EMT and invasion stimulated by TGF-beta by preventing actin cytoskeletal rearrangements and E-cadherin downregulation. Moreover, both CystC molecules completely antagonized TGF-beta-mediated morphological transformation and anchorage-independent growth of NRK cells, and inhibited their invasion through synthetic basement membranes. Both CystC and Delta14CystC also inhibited TGF-beta signaling in two tumorigenic human breast cancer cell lines.. Our findings show that TGF-beta stimulation of initiating metastatic events, including decreased cell polarization, reduced cell-cell contact, and elevated cell invasion and migration, are prevented by CystC treatment. Our findings also suggest that the future development of CystC or its peptide mimetics hold the potential to improve the therapeutic response of human breast cancers regulated by TGF-beta.

    Topics: Animals; Breast Neoplasms; Cell Line; Cell Line, Tumor; Cell Transformation, Neoplastic; Cystatin C; Cystatins; Epithelial Cells; Female; Humans; Kidney; Mesoderm; Protease Inhibitors; Rats; Transforming Growth Factor beta

2005
Somatic acquisition and signaling of TGFBR1*6A in cancer.
    JAMA, 2005, Oct-05, Volume: 294, Issue:13

    TGFBR1*6A is a common polymorphism of the type I transforming growth factor beta receptor (TGFBR1). Epidemiological studies suggest that TGFBR1*6A may act as a tumor susceptibility allele. How TGFBR1*6A contributes to cancer development is largely unknown.. To determine whether TGFBR1*6A is somatically acquired by primary tumors and metastases during cancer development and whether the 3-amino acid deletion that differentiates TGFBR1*6A from TGFBR1 is part of the mature receptor or part of the signal sequence and to investigate TGFBR1*6A signaling in cancer cells.. Tumor and germline tissues from 531 patients with a diagnosis of head and neck, colorectal, or breast cancer recruited from 3 centers in the United States and from 1 center in Spain from June 1, 1994, through June 30, 2004. In vitro translation assays, MCF-7 breast cancer cells stably transfected with TGFBR1*6A, TGFBR1, or the vector alone, DLD-1 colorectal cancer cells that endogenously carry TGFBR1*6A, and SW48 colorectal cancer cells that do not carry TGFBR1*6A.. TGFBR1*6A somatic acquisition in cancer. Determination of the amino terminus of the mature TGFBR1*6A and TGFBR1 receptors. Determination of TGF-beta-dependent cell proliferation.. TGFBR1*6A was somatically acquired in 13 of 44 (29.5%) colorectal cancer metastases, in 4 of 157 (2.5%) of colorectal tumors, in 4 of 226 (1.8%) head and neck primary tumors, and in none of the 104 patients with breast cancer. TGFBR1*6A somatic acquisition is not associated with loss of heterozygosity, microsatellite instability, or a mutator phenotype. The signal sequences of TGFBR1 and TGFBR1*6A are cleaved at the same site resulting in identical mature receptors. TGFBR1*6A may switch TGF-beta growth inhibitory signals into growth stimulatory signals in MCF-7 breast cancer cells and in DLD-1 colorectal cancer cells.. TGFBR1*6A is somatically acquired in 29.5% of liver metastases from colorectal cancer and may bestow cancer cells with a growth advantage in the presence of TGF-beta. The functional consequences of this conversion appear to be mediated by the TGFBR1*6A signal sequence rather than by the mature receptor. The results highlight a new facet of TGF-beta signaling in cancer and suggest that TGFBR1*6A may represent a potential therapeutic target in cancer.

    Topics: Activin Receptors, Type I; Alleles; Amino Acid Sequence; Breast Neoplasms; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Colorectal Neoplasms; Gene Expression Regulation, Neoplastic; Genetic Predisposition to Disease; Genotype; Head and Neck Neoplasms; Humans; Neoplasm Metastasis; Phenotype; Polymorphism, Genetic; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Sequence Deletion; Signal Transduction; Transforming Growth Factor beta

2005
Restoration of TGF-beta signalling reduces tumorigenicity in human lung cancer cells.
    British journal of cancer, 2005, Nov-14, Volume: 93, Issue:10

    Members of the transforming growth factor-beta (TGF-beta) family regulate a wide range of biological processes including cell proliferation, migration, differentiation, apoptosis, and extracellular matrix deposition. Resistance to TGF-beta-mediated tumour suppressor function in human lung cancer may occur through the loss of type II receptor (TbetaRII) expression. In this study, we investigated the expression pattern of TbetaRII in human lung cancer tissues by RT-PCR and Western blot analyses. We observed downregulation of TbetaRII in 30 out of 46 NSCLC samples (65%) by semiquantitative RT-PCR. Western blot analyses with tumour lysates showed reduced expression of TbetaRII in 77% cases. We also determined the effect of TbetaRII expression in lung adenocarcinoma cell line (VMRC-LCD) that is not responsive to TGF-beta due to lack of TbetaRII expression. Stable expression of TbetaRII in these cells restored TGF-beta-mediated effects including Smad2/3 and Smad4 complex formation, TGF-beta-responsive reporter gene activation, inhibition of cell proliferation and increased apoptosis. Clones expressing TbetaRII showed reduced colony formation in soft-agarose assay and significantly reduced tumorigenicity in athymic nude mice. Therefore, these results suggest that reestablishment of TGF-beta signalling in TbetaRII null cells by stable expression of TbetaRII can reverse malignant behaviour of cells and loss of TbetaRII expression may be involved in lung tumour progression.

    Topics: Animals; Apoptosis; Carcinoma, Non-Small-Cell Lung; Cell Proliferation; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; Down-Regulation; Gene Expression Regulation, Neoplastic; Humans; Mice; Mice, Nude; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Transcriptional Activation; Transforming Growth Factor beta; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2005
The ETV6-NTRK3 chimeric tyrosine kinase suppresses TGF-beta signaling by inactivating the TGF-beta type II receptor.
    Proceedings of the National Academy of Sciences of the United States of America, 2005, Nov-08, Volume: 102, Issue:45

    An emerging theme in cancer biology is that although some malignancies occur through the sequential acquisition of different genetic alterations, certain dominantly acting oncoproteins such as those associated with chromosomal translocations have multiple functions and do not require additional mutations for cell transformation. The ETV6-NTRK3 (EN) chimeric tyrosine kinase, a potent oncoprotein expressed in tumors derived from multiple cell lineages, functions as a constitutively active protein tyrosine kinase. Here, we show that EN suppresses TGF-beta signaling by directly binding to the type II TGF-beta receptor, thereby preventing it from interacting with the type I TGF-beta receptor. This activity requires a functional EN protein tyrosine kinase, and type II TGF-beta receptor appears to be a direct target of EN. Our findings provide evidence for a previously undescribed mechanism by which oncogenic tyrosine kinases can block TGF-beta tumor suppressor activity.

    Topics: Activin Receptors, Type I; Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Humans; Mice; Molecular Sequence Data; NIH 3T3 Cells; Oncogene Proteins, Fusion; Phosphorylation; 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; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta

2005
Requirement for the SnoN oncoprotein in transforming growth factor beta-induced oncogenic transformation of fibroblast cells.
    Molecular and cellular biology, 2005, Volume: 25, Issue:24

    Transforming growth factor beta (TGF-beta) was originally identified by virtue of its ability to induce transformation of the AKR-2B and NRK fibroblasts but was later found to be a potent inhibitor of the growth of epithelial, endothelial, and lymphoid cells. Although the growth-inhibitory pathway of TGF-beta mediated by the Smad proteins is well studied, the signaling pathway leading to the transforming activity of TGF-beta in fibroblasts is not well understood. Here we show that SnoN, a member of the Ski family of oncoproteins, is required for TGF-beta-induced proliferation and transformation of AKR-2B and NRK fibroblasts. TGF-beta induces upregulation of snoN expression in both epithelial cells and fibroblasts through a common Smad-dependent mechanism. However, a strong and prolonged activation of snoN transcription that lasts for 8 to 24 h is detected only in these two fibroblast lines. This prolonged induction is mediated by Smad2 and appears to play an important role in the transformation of both AKR-2B and NRK cells. Reduction of snoN expression by small interfering RNA or shortening of the duration of snoN induction by a pharmacological inhibitor impaired TGF-beta-induced anchorage-independent growth of AKR-2B cells. Interestingly, Smad2 and Smad3 play opposite roles in regulating snoN expression in both fibroblasts and epithelial cells. The Smad2/Smad4 complex activates snoN transcription by direct binding to the TGF-beta-responsive element in the snoN promoter, while the Smad3/Smad4 complex inhibits it through a novel Smad inhibitory site. Mutations of Smad4 that render it defective in heterodimerization with Smad3, which are found in many human cancers, convert the activity of Smad3 on the snoN promoter from inhibitory to stimulatory, resulting in increased snoN expression in cancer cells. Thus, we demonstrate a novel role of SnoN in the transforming activity of TGF-beta in fibroblasts and also uncovered a mechanism for the elevated SnoN expression in some human cancer cells.

    Topics: Animals; Base Sequence; Cell Line; Cell Transformation, Neoplastic; Fibroblasts; Gene Expression Regulation, Neoplastic; Humans; Intracellular Signaling Peptides and Proteins; Mice; Molecular Sequence Data; Mutation; Phosphorylation; Proteins; Proto-Oncogene Proteins; Response Elements; RNA, Small Interfering; Smad2 Protein; Smad3 Protein; Smad4 Protein; Transcription, Genetic; Transforming Growth Factor beta

2005
Bone morphogenetic protein-2-induced transformation involves the activation of mammalian target of rapamycin.
    Molecular cancer research : MCR, 2005, Volume: 3, Issue:12

    Bone morphogenetic protein-2 (BMP-2) is an evolutionary conserved protein that is essential for embryonic development. BMP-2 is highly expressed in approximately 98% of human lung carcinomas with little expression in normal lung tissues. BMP-2 has been shown to enhance mobility, invasiveness, and metastasis of cancer cell lines. During development, BMP-2 induces the proto-oncogene phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway to regulate stem cell differentiation. We show that BMP-2 induces the phosphorylation of mTOR in A549 and H1299 lung cancer cell lines, which is attenuated by the PI3K antagonists LY-294002 and wortmannin. p70S6 kinase, which is a direct downstream target of mTOR, is also regulated by BMP-2 in lung cancer cell lines. We find that BMP-2 induces cyclin E in A549 and H1299 cells, which is mediated by the PI3K/mTOR signaling pathway. The regulation of cyclin E by BMP-2 occurs through a Smad 1/5-independent mechanism. Forced expression of BMP-2 in A549 cells (A549/BMP-2) induces transformation as shown by an increase in foci formation. The mTOR antagonist, rapamycin, prevented foci formation of the A549/BMP-2 cells. This study provides evidence that BMP-2-mediated transformation of lung cancer cells involves the activation of the PI3K/mTOR signaling pathway.

    Topics: Androstadienes; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Cell Line, Tumor; Cell Transformation, Neoplastic; Chromones; Cyclin E; Humans; Morpholines; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Phosphorylation; Protein Kinases; Proto-Oncogene Mas; Ribosomal Protein S6 Kinases, 70-kDa; Signal Transduction; Sirolimus; Smad Proteins; TOR Serine-Threonine Kinases; Transforming Growth Factor beta; Up-Regulation; Wortmannin

2005
Cirrhotic hepatocytes exhibit decreased TGFbeta growth inhibition associated with downregulated Smad protein expression.
    Biochemical and biophysical research communications, 2004, Jan-16, Volume: 313, Issue:3

    TGFbeta controls hepatocyte growth through cell cycle arrest and apoptosis, and resistance to TGFbeta is a mechanism of malignant transformation. The aim of this study was to assess differences in TGFbeta-mediated growth inhibition in normal and cirrhotic hepatocytes. Cirrhosis was induced in mice and normal and cirrhotic hepatocytes were isolated by collagenase perfusion and treated with or without TGFbeta (5 ng/ml). DNA synthesis, Smad protein expression, and DNA binding activity were determined. TGFbeta reduced DNA synthesis to a greater degree in normal hepatocytes than in cirrhotic hepatocytes (87% vs. 68%; p<0.05). Smad protein expression was decreased in cirrhotic hepatocytes and Smad 2/3/4 complex formation was suppressed. Furthermore, cirrhotic hepatocytes had decreased DNA binding activity at 120 min following TGFbeta treatment. In conclusion, decreased Smad protein expression may impair TGFbeta-mediated growth inhibition in cirrhotic hepatocytes.

    Topics: Active Transport, Cell Nucleus; Animals; Blotting, Western; Cell Division; Cell Transformation, Neoplastic; Collagenases; DNA; DNA-Binding Proteins; Down-Regulation; Fibrosis; Hepatocytes; Male; Mice; Mice, Inbred BALB C; Protein Binding; Signal Transduction; Smad Proteins; Smad2 Protein; Smad3 Protein; Smad4 Protein; Thymidine; Time Factors; Trans-Activators; Transforming Growth Factor beta

2004
Cancer. Respect thy neighbor!
    Science (New York, N.Y.), 2004, Feb-06, Volume: 303, Issue:5659

    Topics: Animals; Cell Differentiation; Cell Transformation, Neoplastic; Epithelial Cells; Extracellular Matrix; Fibroblasts; Genetic Predisposition to Disease; Humans; Male; Mice; Models, Biological; Neoplasms, Glandular and Epithelial; Prostatic Intraepithelial Neoplasia; Receptors, Transforming Growth Factor beta; Signal Transduction; Stomach Neoplasms; Stromal Cells; Transforming Growth Factor beta

2004
TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia.
    Science (New York, N.Y.), 2004, Feb-06, Volume: 303, Issue:5659

    Stromal cells can have a significant impact on the carcinogenic process in adjacent epithelia. The role of transforming growth factor-beta (TGF-beta) signaling in such epithelial-mesenchymal interactions was determined by conditional inactivation of the TGF-beta type II receptor gene in mouse fibroblasts (Tgfbr2fspKO). The loss of TGF-beta responsiveness in fibroblasts resulted in intraepithelial neoplasia in prostate and invasive squamous cell carcinoma of the forestomach, both associated with an increased abundance of stromal cells. Activation of paracrine hepatocyte growth factor (HGF) signaling was identified as one possible mechanism for stimulation of epithelial proliferation. Thus, TGF-beta signaling in fibroblasts modulates the growth and oncogenic potential of adjacent epithelia in selected tissues.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epithelial Cells; Female; Fibroblasts; Gastric Mucosa; Hepatocyte Growth Factor; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; Neoplasms, Glandular and Epithelial; Prostate; Prostatic Intraepithelial Neoplasia; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-met; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Recombination, Genetic; Signal Transduction; Stomach; Stomach Neoplasms; Stromal Cells; Transforming Growth Factor beta

2004
Tumour-derived TGF-beta1 modulates myofibroblast differentiation and promotes HGF/SF-dependent invasion of squamous carcinoma cells.
    British journal of cancer, 2004, Feb-23, Volume: 90, Issue:4

    The development of an altered stromal microenvironment is a common feature of many tumours including squamous cell carcinoma (SCC), and there is increasing evidence that these changes in the stroma, which include increased expression of proteases and cytokines, may actually promote tumour progression. A common finding is that stromal fibroblasts become 'activated' myofibroblasts, expressing smooth muscle actin and secreting cytokines, proteases and matrix proteins. We show that myofibroblasts are commonly found in the stroma of oral SCC and are often concentrated at the invasive margin of the tumour. Using oral SCC cells and primary oral fibroblasts, we demonstrate that tumour cells directly induce a myofibroblastic phenotype, and that this transdifferentiation is dependent on SCC-derived TGF-beta1. In turn, myofibroblasts secrete significantly higher levels of hepatocyte growth factor/scatter factor compared with fibroblast controls, and this cytokine promotes SCC invasion through Matrigel, a mixture of basement membrane proteins. This is the first time that this double paracrine mechanism has been demonstrated between squamous carcinoma cells and fibroblasts, and emphasises that cancer invasion can be promoted indirectly by the release of tumour-induced host factors from stroma.

    Topics: Carcinoma, Squamous Cell; Cell Differentiation; Cell Transformation, Neoplastic; Fibroblasts; Hepatocyte Growth Factor; Humans; Immunohistochemistry; Mouth Neoplasms; Muscle, Smooth; Neoplasm Invasiveness; Phenotype; Stromal Cells; Transforming Growth Factor beta; Tumor Cells, Cultured

2004
Loss of BMP2, Smad8, and Smad4 expression in prostate cancer progression.
    The Prostate, 2004, May-15, Volume: 59, Issue:3

    The role of the bone morphogenetic protein (BMP) pathway in prostate cancer (PC) is unclear. This study aimed to characterize aspects of the BMP pathway in PC by assessing BMP2, Smad8, and Smad4 expression in normal, hyperplastic, and malignant prostate tissue, and to correlate findings with progression to PC.. Radical prostatectomy (RP) specimens from 74 patients with clinically localized PC (median follow-up 51 months, range 15-152), 44 benign prostatic hypertrophy (BPH) lesions, and 4 normal prostates (NPs) were assessed for BMP2, Smad8, and Smad4 expression using immunohistochemistry.. Both BMP2 (P < 0.001) and nuclear Smad4 (P < 0.0001) expression were significantly decreased in PC compared to benign prostate tissue. Nuclear Smad8 was present in normal/benign prostate tissue but absent in PC and adjacent hyperplasia. Furthermore, loss of BMP2 (P < 0.001) and decreased nuclear Smad4 (P = 0.05) expression correlated with increasing Gleason score.. These data suggest that decreased BMP2, nuclear smad8 and nuclear Smad4 expression are associated with the progression to PC, and in particular loss of BMP2 and Smad4 are related to progression to a more aggressive phenotype.

    Topics: Adult; Aged; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Cell Transformation, Neoplastic; Cohort Studies; Disease Progression; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Humans; Male; Middle Aged; Phenotype; Prognosis; Prostatectomy; Prostatic Hyperplasia; Prostatic Neoplasms; Signal Transduction; Smad4 Protein; Smad8 Protein; Trans-Activators; Transforming Growth Factor beta

2004
Role of transforming growth factor beta in breast carcinogenesis.
    The Lancet. Oncology, 2004, Volume: 5, Issue:4

    Transforming growth factor (TGF) beta is a pre-eminent negative growth regulator that has antiproliferative effects on a range of epithelial cells. This ability has evoked interest in this growth factor as a tumour suppressor with potential clinical significance. In the early stages of breast carcinogenesis, a growth-inhibitory response to TGFbeta is maintained, which depends on an intact TGFbeta signalling pathway. Tumour development and progression of cells along a neoplastic continuum is accompanied by loss of this growth-inhibitory response to TGFbeta, which might instead promote tumour growth indirectly through a combination of permissive effects on stromal tissue, angiogenesis, and the immune system. This review discusses the complexity of functional pleiotropy and the continually changing roles of TGFbeta as a tumour evolves, along with competing therapeutic strategies. The boosting of local endogenous amounts of TGFbeta in conjunction with enhancement of cellular responsiveness might be appropriate in early malignant disease, and anti-TGFbeta approaches could yield a therapeutic gain in metastatic states.

    Topics: Breast Neoplasms; Cell Transformation, Neoplastic; Disease Progression; Female; Humans; Neoplasm Metastasis; Signal Transduction; Transforming Growth Factor beta

2004
Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation.
    Cell, 2004, Apr-16, Volume: 117, Issue:2

    FoxO Forkhead transcription factors are shown here to act as signal transducers at the confluence of Smad, PI3K, and FoxG1 pathways. Smad proteins activated by TGF-beta form a complex with FoxO proteins to turn on the growth inhibitory gene p21Cip1. This process is negatively controlled by the PI3K pathway, a known inhibitor of FoxO localization in the nucleus, and by the telencephalic development factor FoxG1, which we show binds to FoxO-Smad complexes and blocks p21Cip1 expression. We suggest that the activity of this network confers resistance to TGF-beta-mediated cytostasis during the development of the telencephalic neuroepithelium and in glioblastoma brain tumor cells.

    Topics: Animals; Brain Neoplasms; Cell Division; Cell Line, Tumor; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; DNA-Binding Proteins; Fetus; Forkhead Box Protein O1; Forkhead Transcription Factors; Gene Expression Regulation, Developmental; Glioblastoma; Humans; Mice; Mice, Inbred C57BL; Mice, Knockout; Phosphatidylinositol 3-Kinases; Protein Binding; Signal Transduction; Smad Proteins; Stem Cells; Telencephalon; Trans-Activators; Transcription Factors; Transforming Growth Factor beta

2004
ErbB2 and TGF-beta: a cooperative role in mammary tumor progression?
    Cell cycle (Georgetown, Tex.), 2004, Volume: 3, Issue:5

    Amplification and overexpression of ErbB2 (HER2/Neu) is one of the most common alterations associated with breast cancer. Activation of ErbB2 via homodimerization in a non-transformed human mammary epithelial cell line, MCF-10A, in basement membrane cultures leads to formation of proliferative structures that share properties with non-invasive early stage lesions. Recently, we have shown that activation of ErbB2 homodimers combined with expression of transforming growth factor (TGF)-beta induces invasive and migratory activity in MCF-10A cells. In this system, migration requires inputs from numerous cellular pathways. We discuss this data and a model for migration induced by ErbB2 and TGF-beta. Concurrent studies by other groups have also shown that ErbB2 and TGF-beta can cooperate to increase metastatic and invasive behavior in murine mammary tumors. Here we discuss these studies and the potential implications of this research on breast cancer therapeutics.

    Topics: Animals; Breast Neoplasms; Cell Line; Cell Movement; Cell Transformation, Neoplastic; Female; Humans; Mice; Models, Biological; Neoplasm Metastasis; Receptor, ErbB-2; Transforming Growth Factor beta

2004
Raf and RhoA cooperate to transform intestinal epithelial cells and induce growth resistance to transforming growth factor beta.
    Molecular cancer research : MCR, 2004, Volume: 2, Issue:4

    Although unregulated activation of the Ras/Raf/mitogen-activated protein kinase kinase/Erk signaling pathway is believed to be a central mechanism by which many cell types undergo oncogenic transformation, recent studies indicate that activation of Raf kinase by oncogenic Ras is not sufficient to cause tumorigenic transformation in intestinal epithelial cells. Thus, identification of signaling proteins and pathways that interact with Raf to transform intestinal epithelial cells may be critical for understanding aberrant growth control in the intestinal epithelium. Functional interactions between Raf and the small GTPase RhoA were studied in RIE-1 cells overexpressing both activated Raf(22W) and activated RhoA(63L). Double transfectants were morphologically transformed, formed colonies in soft agar, grew in nude mice, overexpressed cyclin D1 and cyclooxygenase-2 (COX-2), and were resistant to growth inhibition by transforming growth factor (TGF) beta. RIE-Raf and RIE-RhoA single transfectants showed none of these characteristics. Expression of a dominant-negative RhoA(N19) construct in RIE-Ras(12V) cells was associated with markedly reduced COX-2 mRNA, COX-2 protein, and prostaglandin E2 levels when compared with RIE-Ras(12V) cells transfected with vector alone. However, no change in transformed morphology, growth in soft agar, cyclin D1 expression, TGFalpha expression, or TGFbeta sensitivity was observed. In summary, coexpression of activated Raf and RhoA induces transformation and TGFbeta resistance in intestinal epithelial cells. Although blockade of RhoA signaling reverses certain well-described characteristics of RIE-Ras cells, it is insufficient to reverse the transformed phenotype and restore TGFbeta sensitivity. Blockade of additional Rho family members or alternate Ras effector pathways may be necessary to fully reverse the Ras phenotype.

    Topics: Alkyl and Aryl Transferases; Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Enzyme Activation; Farnesyltranstransferase; Gene Expression Regulation, Neoplastic; Genes, Dominant; Intestinal Mucosa; Methionine; Mice; Mutation; Proto-Oncogene Proteins c-raf; Proto-Oncogene Proteins p21(ras); Rats; rhoA GTP-Binding Protein; Transfection; Transforming Growth Factor beta; Tumor Stem Cell Assay; Xenograft Model Antitumor Assays

2004
Overexpression of transforming growth factor beta1 in head and neck epithelia results in inflammation, angiogenesis, and epithelial hyperproliferation.
    Cancer research, 2004, Jul-01, Volume: 64, Issue:13

    In the present study, we show that transforming growth factor beta1 (TGF-beta1) was frequently overexpressed in human head and neck squamous cell carcinomas (HNSCCs) and adjacent tissues in comparison with normal head and neck tissues. To determine the role of TGF-beta1 overexpression in HNSCC carcinogenesis, we generated transgenic mice in which TGF-beta1 transgene expression can be induced in head and neck epithelia. TGF-beta1 transgene induction in head and neck epithelia, at levels similar to those in human HNSCCs, caused severe inflammation and angiogenesis. Consequently, TGF-beta1-transgenic epithelia exhibited hyperproliferation. These phenotypes correlated with enhanced Smad signaling in transgenic epithelia and stroma. Our study suggests that TGF-beta1 overexpression at early stages of HNSCC formation provides a tumor promoting microenvironment.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Division; Cell Transformation, Neoplastic; Epithelial Cells; Head and Neck Neoplasms; Humans; Hyperplasia; Inflammation; Mice; Mice, Transgenic; Mouth; Mouth Mucosa; Neovascularization, Pathologic; Oropharynx; Transforming Growth Factor beta; Transforming Growth Factor beta1

2004
Nkx2.1 transcription factor in lung cells and a transforming growth factor-beta1 heterozygous mouse model of lung carcinogenesis.
    Molecular carcinogenesis, 2004, Volume: 40, Issue:4

    The Nkx2.1 homeobox gene and transforming growth factor-beta1 (TGF-beta1) are essential for organogenesis and differentiation of the mouse lung. NKX2.1 is a marker of human lung carcinomas, but it is not known whether this gene participates in early tumorigenesis. Addition of TGF-beta1 to TGF-beta1-responsive nontumorigenic mouse lung cells cotransfected with a NKX2.1Luc luciferase reporter and either a Sp1 or Sp3 plasmid showed a significant increase or decrease, respectively, in NKX2.1Luc transcription. Cotransfection of Sp3 and dominant-negative TGF-beta type II receptor plasmids negated the effect of Sp1. Cotransfected Sp1 plasmid with either dominant-negative Smad2 or Smad3 or Smad4 plasmids significantly decreased NKX2.1Luc transcription. Electrophoretic mobility shift assays revealed binding of Sp1 and Smad4 to the NKX2.1 promoter. With a TGF-beta1 heterozygous mouse model, Nkx2.1 mRNA and protein in lungs of TGF-beta1 heterozygous mice were significantly lower compared to wildtype (WT) littermates. Competitive reverse transcription (RT)-polymerase chain reaction (PCR) and immunostaining showed that Nkx2.1 mRNA and protein decreased significantly in adenomas and adenocarcinomas compared to normal lung tissue. Our in vitro data showed that regulation of Nkx2.1 by TGF-beta1 occurs through TGF-beta type II receptor and Smad signaling, with Sp1 and Sp3 in lung cells. Our in vivo data showed reduced Nkx2.1 in lungs of TGF-beta1 heterozygous mice compared to WT mice, that is detectable in adenomas, and that is further reduced in carcinogenesis, and that correlates with reduction of Sp1, Sp3, and Smads in lung adenocarcinomas. Our findings suggest that reduced Nkx2.1 and TGF-beta1 signaling components may contribute to tumorigenesis in the lungs of TGF-beta1 heterozygous mice.

    Topics: Animals; Blotting, Northern; Cell Transformation, Neoplastic; Disease Models, Animal; Gene Expression Regulation, Neoplastic; Immunohistochemistry; Lung; Lung Neoplasms; Mice; Nuclear Proteins; Receptors, Transforming Growth Factor beta; RNA, Messenger; Thymidine; Thyroid Nuclear Factor 1; Transcription Factors; Transforming Growth Factor beta; Tritium

2004
Chronic exposure of transforming growth factor beta 1 confers a more aggressive tumor phenotype through downregulation of p21(WAF1/CIP1) in conditionally immortalized pancreatic epithelial cells.
    Surgery, 2004, Volume: 136, Issue:2

    Recent studies have demonstrated that transforming growth factor beta 1 (TGF-beta1) expression is markedly enhanced in invasive ductal pancreatic adenocarcinomas, although the precise role of TGF-beta1 in pancreatic carcinogenesis remains unclear. We analyzed TGF-beta1 expression in pancreatic intraepithelial neoplasias (PanINs) and the effects of chronic TGF-beta1 exposure on conditionally immortalized pancreatic epithelial (IMPE) cells.. Sixty-one PanIN lesions were immunohistochemically stained with a polyclonal rabbit antibody against human TGF-beta1. Growth-inhibitory effects of short-term exposure to TGF-beta1 were examined in IMPE cells. IMPE cells resistant to TGF-beta1 (IMPE-Tr cells) were generated by continuous exposure to 1 ng/mL of TGF-beta1 for more than 50 days. Phenotypic alterations of IMPE-Tr cells were examined by soft agar and Matrigel assay and Western blot analysis. IMPE and IMPE-Tr cells were injected subcutaneously into nude mice for an in vivo tumorigenicity assay.. Forty-six percent of PanINs (28/61) were positive for TGF-beta1 expression, whereas all the epithelia of normal pancreatic ducts were negative. TGF-beta1 treatment showed the marked growth-inhibitory effects (>75%) in IMPE cells, whereas its effects were not observed in IMPE-Tr cells. IMPE-Tr cells were more spindle shaped compared with IMPE cells. In soft agar and Matrigel, formations of many colonies were observed in IMPE-Tr cells, but not in IMPE cells. Interestingly, the expression of p21(WAF1/CIP1) was induced by short-term exposure to TGF-beta1 in IMPE cells, whereas the induction was decreased in IMPE-Tr cells. All of the IMPE-Tr cell-injected mice (5/5) had subcutaneous tumors, although no tumor was found in the IMPE cell-injected mice.. TGF-beta1 expression in PanINs and neoplastic transformation of IMPE cells by long-term exposure to TGF-beta1 suggest that TGF-beta1 may act as a tumor promoter in the early stage of pancreatic carcinogenesis.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Collagen; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Down-Regulation; Drug Combinations; Epithelial Cells; Humans; Laminin; Mice; Mice, Nude; Pancreas; Pancreatic Neoplasms; Proteoglycans; Transforming Growth Factor beta; Transforming Growth Factor beta1

2004
[Obstruction of TGF-beta1 signal transduction can decrease the process of hepatocellular carcinoma in mice induced by CCl4/ethanol].
    Zhonghua yi xue za zhi, 2004, Jul-02, Volume: 84, Issue:13

    To study obstruction of the TGF-beta(1) signal transduction by antisense RNA of Smad(4) and its effects on experimental hepatic carcinoma of mice.. We used the mouse model of primary hepatic carcinoma induced by CCl(4)/ethanol, and transferred antisense Smad(4)cDNA with retrovirus-mediated via portal vein infusion into liver. Southern Blot confirmed that the antisense Smad(4)cDNA had been integrated into the liver. The antisense Smad(4) gene could down-regulate the expression of Smad(4) in fibrotic liver observed by Northern and Western Blot.. In the non-therapeutic cirrhotic liver, the expression of Smad(4) mRNA was significantly increased than normal liver. After antisense Smad(4) gene was transferred, the expression of Smad(4) mRNA in the therapeutic liver was significantly decreased compared with non-therapeutic cirrhotic liver. The fibrotic degree of therapeutic liver was alleviated compared with the non-therapeutic fibrotic liver. No significant difference was found between the rates of carcinogenesis of non-therapeutic cirrhotic liver and that of therapeutic cirrhotic liver. But the diameters and numbers of the liver cancers in the therapeutic cirrhotic group were less than that in the non-therapeutic cirrhotic group.. These results indicate that the antisense Smad(4) gene not only can obstruct the progression of liver fibrosis, but also can inhibit the progression of liver cancer, by obstructing the signal transduction of TGF-beta1.

    Topics: Adenoviridae; Animals; Antisense Elements (Genetics); Carbon Tetrachloride; Cell Transformation, Neoplastic; DNA-Binding Proteins; Ethanol; Liver Cirrhosis, Experimental; Liver Neoplasms, Experimental; Male; Mice; Random Allocation; Signal Transduction; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta; Transforming Growth Factor beta1

2004
NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression.
    The Journal of clinical investigation, 2004, Volume: 114, Issue:4

    The transcription factor NF-kappaB is activated in a range of human cancers and is thought to promote tumorigenesis, mainly due to its ability to protect transformed cells from apoptosis. To investigate the role of NF-kappaB in epithelial plasticity and metastasis, we utilized a well-characterized in vitro/in vivo model of mammary carcinogenesis that depends on the collaboration of the Ha-Ras oncoprotein and TGF-beta. We show here that the IKK-2/IkappaBalpha/NF-kappaB pathway is required for the induction and maintenance of epithelial-mesenchymal transition (EMT). Inhibition of NF-kappaB signaling prevented EMT in Ras-transformed epithelial cells, while activation of this pathway promoted the transition to a mesenchymal phenotype even in the absence of TGF-beta. Furthermore, inhibition of NF-kappaB activity in mesenchymal cells caused a reversal of EMT, suggesting that NF-kappaB is essential for both the induction and maintenance of EMT. In line with the importance of EMT for invasion, blocking of NF-kappaB activity abrogated the metastatic potential of mammary epithelial cells in a mouse model system. Collectively, these data provide evidence of an essential role for NF-kappaB during distinct steps of breast cancer progression and suggest that the cooperation of Ras- and TGF-beta-dependent signaling pathways in late-stage tumorigenesis depends critically on NF-kappaB activity.

    Topics: Animals; Apoptosis; Blotting, Western; Breast Neoplasms; Cell Line, Transformed; Cell Line, Tumor; Cell Transformation, Neoplastic; Cell Transformation, Viral; Disease Models, Animal; Epithelial Cells; Gene Expression Regulation, Neoplastic; Immunohistochemistry; Mammary Glands, Animal; Mesoderm; Mice; Mice, Nude; Neoplasm Metastasis; Neoplasm Transplantation; NF-kappa B; Oncogene Protein p21(ras); Retroviridae; Time Factors; Transforming Growth Factor beta

2004
Transforming growth factor-beta pathway serves as a primary tumor suppressor in CD8+ T cell tumorigenesis.
    Cancer research, 2004, Sep-15, Volume: 64, Issue:18

    Tumorigenesis in rodents, as well as in humans, has been shown to be a multistep process, with each step reflecting an altered gene product or gene regulatory process leading to autonomy of cell growth. Initial genetic mutations are often associated with dysfunctional growth regulation, as is demonstrated in several transgenic mouse models. These changes are often followed by alterations in tumor suppressor gene function, allowing unchecked cell cycle progression and, by genomic instability, additional genetic mutations responsible for tumor metastasis. Here we show that reduced transforming growth factor-beta signaling in T lymphocytes leads to a rapid expansion of a CD8+ memory T-cell population and a subsequent transformation to leukemia/lymphoma as shown by multiple criteria, including peripheral blood cell counts histology, T-cell receptor monoclonality, and host transferability. Furthermore, spectral karyotype analysis of the tumors shows that the tumors have various chromosomal aberrations. These results suggest that reduced transforming growth factor-beta signaling acts as a primary carcinogenic event, allowing uncontrolled proliferation with consequent accumulation of genetic defects and leukemic transformation.

    Topics: Animals; CD8-Positive T-Lymphocytes; Cell Transformation, Neoplastic; Chromosome Aberrations; Immunologic Memory; Leukemia, T-Cell; Lymphoproliferative Disorders; Mice; Mice, Inbred C57BL; Mice, Transgenic; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Antigen, T-Cell; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta

2004
Desmoplastic reaction influences pancreatic cancer growth behavior.
    World journal of surgery, 2004, Volume: 28, Issue:8

    Connective tissue growth factor (CTGF), which is regulated by transforming growth factor-ss (TGFss), has recently been implicated in the pathogenesis of fibrotic diseases and tumor stroma. Inasmuch as generation of desmoplastic tissue is characteristic for pancreatic cancer, it is not known whether it gives pancreatic cancer cells a growth advantage or is a reaction of the body to inhibit cancer cell progression. In the present study we analyzed the expression and localization of CTGF and evaluated whether it influences the prognosis of pancreas cancer. Tissue samples were obtained from 25 individuals (6 women, 19 men) undergoing pancreatic resection for pancreatic cancer. Tissue samples from 13 previously healthy organ donors (5 women, 8 men) served as controls. Expression of CTGF was studied by Northern blot analysis. In situ hybridization and immunohistochemistry localized the respective mRNA moieties and proteins in the tissue samples. Northern blot analysis revealed that pancreatic cancer tissue samples exhibited a 46-fold increase in CTGF mRNA expression ( p < 0.001) over that of normal controls. In vitro studies confirmed that pancreatic stellate cells are the major source of CTGF mRNA expression and revealed a large variance in basal and TGFss-induced CTGF expression in cultured pancreatic cancer cells. This could also be confirmed by in situ hybridization, indicating that CTGF mRNA signals were located principally in fibroblasts, with only weak signals in the cancer cells. High CTGF mRNA levels in the tissue samples correlated with better tumor differentiation ( p < 0.03). In addition, patients whose tumors exhibited high CTGF mRNA levels (> onefold increase above normal controls) lived significantly longer than those whose tumors expressed low CTGF mRNA levels (none to onefold) ( p < 0.04 multivariate analysis). Our present data indicate that CTGF, as a downstream mediator of TGFss, is overexpressed in connective tissue cells and to a lesser extent in pancreatic cancer cells. Because patients with high CTGF mRNA expression levels have a better prognosis, our findings indicate that the desmoplastic reaction provides a growth disadvantage for pancreatic cancer cells.

    Topics: Adenocarcinoma; Aged; Cell Division; Cell Transformation, Neoplastic; Connective Tissue Growth Factor; Female; Fibroblasts; Fibrosis; Follow-Up Studies; Humans; Immediate-Early Proteins; Intercellular Signaling Peptides and Proteins; Male; Middle Aged; Neoplasm Staging; Pancreas; Pancreatectomy; Pancreatic Neoplasms; Prognosis; RNA, Messenger; Survival Analysis; Transforming Growth Factor beta; Tumor Cells, Cultured

2004
TGF-beta-induced upregulation of MMP-2 and MMP-9 depends on p38 MAPK, but not ERK signaling in MCF10A human breast epithelial cells.
    International journal of oncology, 2004, Volume: 25, Issue:5

    Transforming growth factor (TGF)-beta has been reported to exert growth inhibitory activity in normal epithelial cells whereas it induces cell proliferation and invasive phenotypes in advanced carcinomas. Our previous study showed that MCF10A, a spontaneously immortalized "normal" breast epithelial cell line, is resistant to TGF-beta-induced growth inhibition, suggesting that conversion of TGF-beta growth inhibitory signaling into an oncogenic pathway may occur at the early stage of tumor development/progression. To address this issue, we investigated the TGF-beta signaling pathway and its role in phenotypic transformation of MCF10A cells. TGF-beta treatment induced changes in the MCF10A cell morphology from cuboidal to an elongated spindle-like shape, accompanied with down-regulation of epithelial cell marker E-cadherin. TGF-beta treatment was sufficient to induce migrative and invasive phenotypes in these cells, an important phenotypic conversion during tumor progression. We also showed that TGF-beta treatment rapidly activated ERK-1/2 and p38 MAPK leading to upregulation of matrix metalloproteinase (MMP)-2 and MMP-9. Using chemical inhibitors and dominant negative mutants of MAPKs, we provide evidence that while both p38 MAPK and ERKs are required for TGF-beta-induced MCF10A cell migration and invasion, TGF-beta-induced MMP-2 and MMP-9 expression depends on p38 MAPK signaling, but is independent of ERK activity. This study demonstrates the roles of TGF-beta signaling pathways for induction of oncogenic signaling in preneoplastic human breast epithelial cells and will deepen our understanding of TGF-beta signaling in the progress of breast cancer.

    Topics: Breast Neoplasms; Cell Movement; Cell Transformation, Neoplastic; Disease Progression; Epithelial Cells; Female; Humans; Matrix Metalloproteinase 2; Matrix Metalloproteinase 9; p38 Mitogen-Activated Protein Kinases; Phenotype; Signal Transduction; Transforming Growth Factor beta; Up-Regulation

2004
Down-regulation of Betaig-h3 gene is involved in the tumorigenesis in human bronchial epithelial cells induced by heavy-ion radiation.
    Radiation research, 2004, Volume: 162, Issue:6

    High-energy (HZE) heavy ions, when compared to low-LET radiation, are highly effective in inducing gene mutation, chromosomal aberrations and neoplastic transformation. However, the underlying molecular mechanisms are not clearly understood. We have recently shown that the down-regulation of Betaig-h3 expression is causally linked to the tumorigenic phenotype of papillomavirus-immortalized human bronchial epithelial (BEP2D) cells treated with high-LET alpha-particle radiation. Using the BEP2D cell culture system, a radiation-induced transformation model has been established by a single 60-cGy dose of (56)Fe heavy-ion radiation. To determine whether the Betaig-h3 gene is involved in (56)Fe ion-induced tumorigenesis, the expression levels of the Betaig-h3 gene in tumorigenic cell lines and the ability of in vivo tumor suppression through the reintroduction of the Betaig-h3 gene in tumorigenic cells were determined. We found that the expression level of this gene is markedly decreased in three tumorigenic cell lines ((56)FeT1-T3) compared with parental BEP2D cells. Ectopic expression of its cDNA in the (56)FeT2 tumorigenic cells significantly suppressed their tumorigenicity. Although biologically active TGFB1 is elevated in two of three tumorigenic cell lines, all these cell lines are resistant to the induction of Betaig-h3 expression by incubating the transformed cells with exogenous TGFB1 relative to control cells. Our data strongly suggest that down-regulation of Betaig-h3 expression results from the defect in the TGFB1 signaling pathway and plays a pivotal role in the tumorigenic process induced by (56)Fe heavy-ion radiation.

    Topics: Bronchi; Bronchial Neoplasms; Cell Transformation, Neoplastic; Cells, Cultured; Down-Regulation; Epithelial Cells; Extracellular Matrix Proteins; Heavy Ions; Humans; Iron; Neoplasms, Radiation-Induced; Transfection; Transforming Growth Factor beta; Transforming Growth Factor beta1

2004
[Effect of overexpression of Smad7 gene on cell proliferation].
    Zhonghua zhong liu za zhi [Chinese journal of oncology], 2004, Volume: 26, Issue:9

    To study the effect of overexpression of Smad7 gene on cell proliferation in human bronchial epithelial cell lines.. Human bronchial epithelial cell lines, BEP2D and BERP35T2 cells, were cotransfected with the mammalian expression vectors PCISmad7.neo and pMyc-SEAP, the latter was ac-myc cis-acting enhancer element fused with alkaline phosphatase (SEAP) reporter gene. Expression of c-myc, p15 and p21 mRNA was detected by RT-PCR before and after stable transfection of Smad7 into BEP2D and BERP35T2 cells in order to study the regulation of TGF-beta-mediated growth inhibition.. After BEP2D and BERP35T2 cells transfected with Smad7, the transcriptional activity of c-myc was significantly increased. Smad7 overexpressing cells showed upregulation of c-myc expression and downregulation of p15 and p21 expression, which contributed to the loss of TGF-beta responses in these cells.. Overexpression of Smad7 may facilitate cell proliferation by antagonizing TGF-beta-mediated antiproliferative gene responses.

    Topics: Bronchi; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p21; Epithelial Cells; Humans; Proto-Oncogene Proteins c-myc; Signal Transduction; Smad7 Protein; Transfection; Transforming Growth Factor beta

2004
Telomerase prolongs the lifespan of normal human ovarian surface epithelial cells without inducing neoplastic phenotype.
    Journal of the Society for Gynecologic Investigation, 2004, Volume: 11, Issue:8

    The aim of this study was to determine the effects of exogenous expression of the catalytic subunit of telomerase (hTERT) on the lifespan, growth characteristics, and tumorigenicity of normal human ovarian surface epithelial (OSE) cells.. Low-passage primary cultures of normal human OSE cells were transfected with hTERT and the resulting cell lines were characterized.. The ectopic expression of hTERT stabilized the telomeres of the OSE cultures above 8 kb. The hTERT-transfected OSE cell lines grew beyond the normal lifespan seen in OSE cells and propagated in culture for more than 40 passages before senescing. Moreover, the hTERT-transfected cells demonstrated extensive proliferative capacity as evidenced by their ability to continuously grow even when seeded at low dilutions. The morphologic features and normal differentiation patterns seen in normal OSE cells were likewise retained by the hTERT-transfected cells. In addition, the cultures remained responsive to physiologic concentrations of epidermal growth factor and transforming growth factor-beta. Changes associated with neoplastic transformation like anchorage-independent growth, tumorigencity and karyotypic instability were not observed.. We were able to show that the ectopic expression of hTERT in normal human OSE: 1) resulted in cultures with greater growth potential and longer lifespan and 2) did not induce a transformed phenotype previously seen in viral oncogene-transfected OSE cells. The established cell lines would not only provide sufficient material for comprehensive studies to investigate the normal physiology of OSE cells, but could also help in the understanding of the early steps of ovarian carcinogenesis.

    Topics: Animals; Cell Differentiation; Cell Division; Cell Survival; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Epidermal Growth Factor; Epithelial Cells; Female; Humans; Immunohistochemistry; Keratins; Mice; Mice, SCID; Ovarian Neoplasms; Ovary; Telomerase; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2004
Rap1 reverses transcriptional repression of TGF-beta type II receptor by a mechanism involving AP-1 in the human pancreatic cancer cell line, UK Pan-1.
    Journal of cellular physiology, 2003, Volume: 194, Issue:1

    The TGF-beta signaling pathway has potent anti-mitogenic effects in epithelial cells and loss of negative growth regulation is often associated with increased tumorigenicity. The human pancreatic ductal adenocarcinoma cell line, UK Pan-1, which expresses DPC4, is not highly responsive to TGF-beta due to transcriptional repression of TGF-beta type II receptor (RII). Here, we show that UK Pan-1 cells transfected with a plasmid to overexpress rap1 protein (UK/rap1) causes an increase in RII transcription and restores sensitivity to TGF-beta growth inhibition. The overexpression of rap1 was associated with diminished ras signaling as measured by ras binding domain (RBD)-binding assays. Electrophoretic mobility shift assays (EMSA) analysis revealed increased binding of nuclear proteins to a previously identified positive regulatory element (PRE1) of the RII promoter in rap1 transfected cells. Competition with an oligo containing the AP-1 consensus site was able to inhibit this binding of nuclear proteins to the PRE1 region. Further EMSA analysis using antibodies to various AP-1 components revealed that junB antibodies partially depleted the increase in binding to the PRE1 seen in UK/rap1 cells while antibodies to other AP-1 constituents such as c-jun, c-fos, and ATF-1 had no effect on binding. Consistent with this data, transient transfection of UK Pan-1 cells with junB resulted in greater RII transcription (twofold) as measured by RII-luciferase assay. Mutation of the AP-1 site inhibited junB-mediated or rap1-mediated increases in RII promoter activity. These data suggest that rap1 signaling may mediate an increase in RII transcription via increased binding of nuclear factors including junB to the PRE1 region of the RII promoter.

    Topics: Carcinoma; Cell Division; Cell Transformation, Neoplastic; Gene Expression Regulation, Neoplastic; Genes, Regulator; Humans; Mitogen-Activated Protein Kinases; Pancreatic Neoplasms; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-fos; Proto-Oncogene Proteins c-jun; rap1 GTP-Binding Proteins; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Repressor Proteins; RNA, Messenger; Signal Transduction; Transcription Factor AP-1; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation

2003
A Nodal- and ALK4-independent signaling pathway activated by Cripto-1 through Glypican-1 and c-Src.
    Cancer research, 2003, Mar-15, Volume: 63, Issue:6

    Human Cripto-1 (CR-1) is a member of the epidermal growth factor-Cripto FRL1 Cryptic family that has been shown to function as a coreceptor with the type I Activin serine-threonine kinase receptor ALK4 for the transforming growth factor beta-related peptide Nodal. However, CR-1 can also activate the mitogen-activated protein kinase and Akt pathways independently of Nodal and ALK4 by an unknown mechanism. Here, we demonstrate that CR-1 specifically binds to Glypican-1, a membrane-associated heparan sulfate proteoglycan, and activates the tyrosine kinase c-Src, triggering the mitogen-activated protein kinase and Akt signaling pathways. Finally, an active Src kinase is necessary for CR-1 to induce in vitro transformation and migration in mouse mammary epithelial cells.

    Topics: Activin Receptors, Type I; Animals; Anti-HIV Agents; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Cell Line; Cell Movement; Cell Transformation, Neoplastic; Chlorocebus aethiops; COS Cells; Enzyme Activation; Enzyme-Linked Immunosorbent Assay; Epidermal Growth Factor; GPI-Linked Proteins; Heparan Sulfate Proteoglycans; HIV Antibodies; Humans; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Mice; Mitogen-Activated Protein Kinases; Neoplasm Proteins; Nodal Protein; Phosphatidylinositol Diacylglycerol-Lyase; Phosphorylation; Polysaccharide-Lyases; Precipitin Tests; Protein Binding; Protein Serine-Threonine Kinases; Proteins; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Signal Transduction; src-Family Kinases; Substrate Specificity; Transforming Growth Factor beta; Type C Phospholipases

2003
PMEPA1, a transforming growth factor-beta-induced marker of terminal colonocyte differentiation whose expression is maintained in primary and metastatic colon cancer.
    Cancer research, 2003, Apr-01, Volume: 63, Issue:7

    To identify potential effectors of transforming growth factor (TGF)-beta-mediated suppression of colon cancer, we used GeneChip expression microarrays to identify TGF-beta-induced genes in VACO 330, a nontransformed TGF-beta-sensitive cell line derived from a human adenomatous colon polyp. PMEPA1 was identified as a gene highly up-regulated by TGF-beta treatment of VACO 330. Northern blot analysis confirmed TGF-beta induction of PMEPA1 in VACO 330, as well as a panel of three other TGF-beta-sensitive colon cell lines. PMEPA1 induction could be detected as early as 2 h after TGF-beta treatment and was not inhibited by pretreatment of cells with cycloheximide, suggesting that PMEPA1 is a direct target of TGF-beta signaling. Wild-type PMEPA1 and an alternative splice variant lacking the putative transmembrane domain were encoded by the PMEPA1 locus and were shown by epitope tagging to encode proteins with differing subcellular localization. Both variants were found to be expressed in normal colonic epithelium, and both were shown to be induced by TGF-beta. Consistent with TGF-beta playing a role in terminal differentiation of colonocytes, in situ hybridization of normal colonic epithelium localized PMEPA1 expression to nonproliferating, terminally differentiated epithelium located at the top of colonic crypts. Intriguingly, in situ hybridization and Northern blot analysis showed that the expression of PMEPA1 was well maintained both in colon cancer primary tumors and in colon cancer liver metastases. PMEPA1 is thus a novel TGF-beta-induced marker of a differentiated crypt cell population. Moreover, as PMEPA1 expression is maintained, presumptively in a TGF-beta-independent manner after malignant transformation and metastasis, it demonstrates that even late colon cancers retain a strong capacity to execute many steps of the normal colonic differentiation program.

    Topics: Alternative Splicing; Amino Acid Sequence; Base Sequence; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Colon; Colonic Neoplasms; Gene Expression Regulation, Neoplastic; Humans; Liver Neoplasms; Membrane Proteins; Molecular Sequence Data; Protein Isoforms; Signal Transduction; Subcellular Fractions; Transfection; Transforming Growth Factor beta; Up-Regulation

2003
The transforming activity of Ski and SnoN is dependent on their ability to repress the activity of Smad proteins.
    The Journal of biological chemistry, 2003, Aug-15, Volume: 278, Issue:33

    The regulation of cell growth and differentiation by transforming growth factor-beta (TGF-beta) is mediated by the Smad proteins. In the nucleus, the Smad proteins are negatively regulated by two closely related nuclear proto-oncoproteins, Ski and SnoN. When overexpressed, Ski and SnoN induce oncogenic transformation of chicken embryo fibroblasts. However, the mechanism of transformation by Ski and SnoN has not been defined. We have previously reported that Ski and SnoN interact directly with Smad2, Smad3, and Smad4 and repress their ability to activate TGF-beta target genes through multiple mechanisms. Because Smad proteins are tumor suppressors, we hypothesized that the ability of Ski and SnoN to inactivate Smad function may be responsible for their transforming activity. Here, we show that the receptor regulated Smad proteins (Smad2 and Smad3) and common mediator Smad (Smad4) bind to different regions in Ski and SnoN. Mutation of both regions, but not each region alone, markedly impaired the ability of Ski and SnoN to repress TGF-beta-induced transcriptional activation and cell cycle arrest. Moreover, when expressed in chicken embryo fibroblasts, mutant Ski or SnoN defective in binding to the Smad proteins failed to induce oncogenic transformation. These results suggest that the ability of Ski and SnoN to repress the growth inhibitory function of the Smad proteins is required for their transforming activity. This may account for the resistance to TGF-beta-induced growth arrest in some human cancer cell lines that express high levels of Ski or SnoN.

    Topics: Animals; Binding Sites; Carcinoma, Hepatocellular; Cell Division; Cell Transformation, Neoplastic; Chick Embryo; Chickens; DNA-Binding Proteins; Fibroblasts; Humans; Intracellular Signaling Peptides and Proteins; Liver Neoplasms; Mutagenesis; Proto-Oncogene Proteins; Smad2 Protein; Smad3 Protein; Smad4 Protein; Suppression, Genetic; Trans-Activators; Transcriptional Activation; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
Intestinal transformation results in transforming growth factor-beta-dependent alteration in tumor cell-cell matrix interactions.
    Surgery, 2003, Volume: 133, Issue:5

    An alteration in the expression of and response to transforming growth factor-beta 1 (TGF-beta 1) appears to be an important event during colorectal carcinogenesis. However, the precise role of TGF-beta 1 in colorectal carcinogenesis is not clear. We have previously described in detail the changes in cell proliferation and differentiation caused by chronic exposure to TGF-beta 1. In this study we sought to better characterize the changes in tumor cell-cell matrix interactions seen during TGF-beta 1-mediated intestinal transformation.. Rat intestinal epithelial cells (RIE) and RIE cells transformed by chronic exposure to TGF-beta 1 (RIE-Tr) were treated with TGF-beta 1 and production of components of the plasmin/plasminogen system measured by ELISA and Western blotting. TGF-beta 1 effects on invasion and adhesion were determined in vitro. The role of urokinase on TGF-beta 1-mediated invasion and adhesion were determined using immunoneutralization. The role of COX-2 was determined using a specific COS-2 inhibitor.. TGF-beta 1 had no effect on RIE-1 adhesion to collagen types I and IV, fibronectin, and laminin, or invasion through collagen types I and IV. However, 5 ng/mL TGF-beta 1 significantly increased the invasiveness and decreased the adhesiveness of RIE-Tr. This effect of TGF-beta 1 on RIE-Tr was associated with a significant increase in plasmin activity secondary to increased expression of uPA. TGF-beta 1 had no effect on either uPA receptor or PAI-1 in this system. Antibodies to uPA completely blocked the TGF-beta 1-mediated invasiveness of the RIE-Tr cells and returned their adhesiveness to basement membrane proteins to baseline. Addition of the selective Cox-2 inhibitor SC-58125 resulted in a dose-dependent decrease in TGF-beta 1-mediated invasion and uPA expression.. This study provides additional evidence for TGF-beta 1 as a tumor promoter during intestinal carcinogenesis and a possible new mechanism for Cox-2-related colon carcinogenesis.

    Topics: Animals; Cell Adhesion; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Cyclooxygenase 2; Cyclooxygenase 2 Inhibitors; Cyclooxygenase Inhibitors; Dose-Response Relationship, Drug; Extracellular Matrix; Fibrinolysin; Intestinal Mucosa; Isoenzymes; Neoplasm Invasiveness; Prostaglandin-Endoperoxide Synthases; Pyrazoles; Rats; Transforming Growth Factor beta; Transforming Growth Factor beta1; Urokinase-Type Plasminogen Activator

2003
IL-2 gene and antisense TGF-beta1 strategies counteract HSV-2 transformed tumor progression.
    Technology in cancer research & treatment, 2003, Volume: 2, Issue:3

    The H238 tumor cells are Herpes simplex virus type 2-transformed BALB/c mouse fibroblasts that constitutively express transforming growth factor (TGF-beta1). TGF-beta can diminish immune capacity, whereas interleukin 2 (IL-2) is stimulatory to the immune system and can counteract the negative effects of TGF-beta1. The H238-BALB/c system provides a syngeneic model to evaluate new strategies with the potential to ameliorate tumor-induced immune depression. Plasmids expressing either antisense TGF-beta1 or murine Il-2 were constructed and stably transformed cells generated (masH238 and H238-IL2, respectively). In vitro measurements (ELISA and RT-PCR) demonstrated a >70% decrease in TGF-beta1 secretion by the masH238 tumor cells, and significant levels of IL-2 production by the H238-IL2 transfected cells when compared to wild type and control plasmid-transfected H238 cells. BALB/c mice injected subcutaneously with the masH238 cells developed significantly smaller tumors than the controls. Mice injected with H238-IL-2 cells developed tumors that failed to progress relative to control tumor growth. The differences in tumor growth in the mice were associated with enhanced immune reactivity and an increased response to T lymphocyte mitogens. Significant differences were also noted in lymphocyte populations and expression of CD25 and CD71 activation markers in the blood and spleens of mice receiving transfected tumor cells. Collectively, the data demonstrate that strategies employing antisense TGF-beta1 and IL-2 expression by transfected tumor cells can counteract the progression of a TGF-beta1-secreting tumor and enhance immune function involving modulation of T lymphocyte populations.

    Topics: Animals; Base Sequence; Cell Division; Cell Transformation, Neoplastic; Female; Flow Cytometry; Herpesvirus 2, Human; Interleukin-2; Kinetics; Lymphocytes; Mice; Mice, Inbred BALB C; Oligodeoxyribonucleotides, Antisense; Spleen; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
Tumor necrosis factor-alpha stimulates the epithelial-to-mesenchymal transition of human colonic organoids.
    Molecular biology of the cell, 2003, Volume: 14, Issue:5

    An epithelial-mesenchymal transition (EMT) characterizes the progression of many carcinomas and it is linked to the acquisition of an invasive phenotype. Given that the tumor microenvironment is an active participant in tumor progression, an important issue is whether a reactive stroma can modulate this process. Using a novel EMT model of colon carcinoma spheroids, we demonstrate that their transforming-growth factor-beta1 (TGF-beta)-induced EMT is accelerated dramatically by the presence of activated macrophages, and we identify tumor necrosis factor-alpha (TNF-alpha) as the critical factor produced by macrophages that accelerates the EMT. A synergy of TNF-alpha and TGF-beta signaling promotes a rapid morphological conversion of the highly organized colonic epithelium to dispersed cells with a mesenchymal phenotype, and this process is dependent on enhanced p38 MAPK activity. Moreover, exposure to TNF-alpha stimulates a rapid burst of ERK activation that results in the autocrine production of this cytokine by the tumor cells themselves. These results establish a novel role for the stroma in influencing EMT in colon carcinoma, and they identify a selective advantage to the stromal presence of infiltrating leukocytes in regulating malignant tumor progression.

    Topics: Cell Transformation, Neoplastic; Colon; Epithelium; Humans; Mesoderm; Mitogen-Activated Protein Kinases; Organoids; p38 Mitogen-Activated Protein Kinases; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

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
Inhibition of Egr-1 expression reverses transformation of prostate cancer cells in vitro and in vivo.
    Oncogene, 2003, Jul-03, Volume: 22, Issue:27

    Transcription factor early growth response-1 (Egr-1) is a crucial regulator of cell growth, differentiation and survival. Several observations suggest that Egr-1 is growth promoting in prostate cancer cells and that blocking its function may impede cancer progression. To test this hypothesis, we developed phosphorothioate antisense oligonucleotides that efficiently inhibit Egr-1 expression without altering the expression of other family members Egr-2, Egr-3 and Egr-4. In TRAMP mouse-derived prostate cancer cell lines, our optimal antisense oligonucleotide decreased the expression of the Egr-1 target gene transforming growth factor-beta1 whereas a control oligonucleotide had no effect, indicating that the antisense blocked Egr-1 function as a transcription factor. The antisense oligonucleotide deregulated cell cycle progression and decreased proliferation of the three TRAMP cell lines by an average of 54+/-3%. Both colony formation and growth in soft agar were inhibited by the antisense oligonucleotide. When TRAMP mice were treated systemically for 10 weeks, the incidence of palpable tumors at 32 weeks of age in untreated mice or mice injected with the control scramble oligonucleotide was 87%, whereas incidence of tumors in antisense-Egr-1-treated mice was significantly reduced to 37% (P=0.026). Thus, Egr-1 plays a functional role in the transformed phenotype and may represent a valid target for prostate cancer therapy.

    Topics: Animals; Blotting, Western; Cell Cycle; Cell Differentiation; Cell Division; Cell Line, Transformed; Cell Membrane; Cell Transformation, Neoplastic; Disease Progression; DNA-Binding Proteins; Early Growth Response Protein 1; Flow Cytometry; Immediate-Early Proteins; In Vitro Techniques; Male; Mice; Oligonucleotides, Antisense; Phenotype; Prostatic Neoplasms; Rats; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Time Factors; Transcription Factors; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
Smad3 regulates senescence and malignant conversion in a mouse multistage skin carcinogenesis model.
    Cancer research, 2003, Jul-01, Volume: 63, Issue:13

    Transforming growth factor beta (TGF-beta) is a growth-inhibitory cytokine for epithelial cells. In the mouse multistage skin carcinogenesis model, defects in TGF-beta 1 signaling reduce senescence in vitro and accelerate malignant progression in vivo. However, the precise postreceptor signaling pathways and specific roles played by Smad proteins in this process have not been defined. Here we show that senescence of v-ras(Ha)-transduced Smad3 null keratinocytes is delayed, whereas overexpression of Smad3, but not Smad2 or Smad4, induced senescence. The TGF-beta 1 target genes c-myc and p15(ink4b) were deregulated in the absence of Smad3. When transplanted to a graft site on nude mice, the v-ras(Ha)-transduced Smad3 null keratinocytes underwent rapid conversion from benign papilloma to malignant carcinoma, whereas wild-type keratinocytes predominantly formed papillomas. These results link Smad3-mediated regulation of growth control genes to senescence in vitro and tumor suppression in vivo.

    Topics: Animals; Base Sequence; Carcinoma; Cell Transformation, Neoplastic; Cells, Cultured; Cellular Senescence; DNA Primers; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Keratinocytes; Mice; Mice, Nude; Papilloma; Promoter Regions, Genetic; Signal Transduction; Skin Neoplasms; Smad3 Protein; Trans-Activators; Transforming Growth Factor beta

2003
Suprabasal alpha6beta4 integrin expression in epidermis results in enhanced tumourigenesis and disruption of TGFbeta signalling.
    Journal of cell science, 2003, Sep-15, Volume: 116, Issue:Pt 18

    Inappropriate alpha6beta4 integrin expression correlates with a high risk of tumour progression in stratified squamous epithelia. Targeted expression of alpha6beta4 in the suprabasal layers of transgenic mouse epidermis dramatically increased the frequency of papillomas, carcinomas and metastases induced by chemical carcinogenesis, independent of the beta4 cytoplasmic domain. Suprabasal alpha6beta4 also perturbed transforming growth factor beta (TGFbeta) signalling as demonstrated by decreased nuclear Smad2 in transgenic epidermis and tumours. In cultured keratinocytes, suprabasal alpha6beta4 relieved TGFbeta-mediated growth inhibition and blocked nuclear translocation of activated Smad2/3. Responsiveness to TGFbeta could be restored by inhibiting cadherin-mediated cell-cell adhesion or phosphoinositide 3-kinase (PI3-K) activity, but not by inhibiting mitogen-activated protein kinase (MAPK) activity. These data suggest that suprabasal alpha6beta4 promotes tumourigenesis by preventing TGFbeta from suppressing clonal expansion of initiated cells in the epidermal basal layer.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Cadherins; Carcinogens; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Enzyme Activation; Epidermis; Integrin alpha6beta4; Keratinocytes; Mice; Mice, Transgenic; Mitogen-Activated Protein Kinases; Neoplasms, Experimental; Phosphatidylinositol 3-Kinases; Signal Transduction; Smad2 Protein; Smad3 Protein; Trans-Activators; Transforming Growth Factor beta

2003
Development and characterization of DP-153, a nontumorigenic prostatic cell line that undergoes malignant transformation by expression of dominant-negative transforming growth factor beta receptor type II.
    Cancer research, 2003, Aug-01, Volume: 63, Issue:15

    We have developed a nontumorigenic epithelial cell line, DP-153, from the dorsal prostate of a Lobund/Wistar rat treated with N-methyl-N-nitrosourea and testosterone propionate. DP-153 cells express cytokeratins 5 and 14, but not cytokeratin 18, consistent with a basal epithelial cell phenotype. Similar to the nontumorigenic NRP-152 prostatic cell line, DP-153 cells do not form tumors in athymic mice and retain many of the properties of normal prostatic cells. They express prostatic acid phosphatase and androgen receptors and require several mitogens (epidermal growth factor, insulin, dexamethasone, and cholera toxin) for sustained growth in culture under serum-containing conditions. DP-153 cells are also growth-stimulated by keratinocyte growth factor and basic fibroblast growth factor and growth-inhibited by all-trans-retinoic acid, 1,25-dihydroxyvitamin D(3), and transforming growth factor (TGF)-beta1. We demonstrate that expression of dominant-negative TGF-beta receptor type II by retroviral transduction of DP-153 cells leads to complete loss of TGF-beta1-induced growth inhibition. When transplanted s.c. in athymic mice, DP-153 cells expressing dominant-negative TGF-beta receptor type II form tumors as early as 4 weeks, in contrast to the vector control and parental cell line, which do not form tumors even 8 months after transplantation, supporting the observation that TGF-beta functions as a tumor suppressor in these cells. Our data further support that DP-153 is a suitable cell line for analysis of normal prostatic growth and carcinogenesis.

    Topics: Animals; Calcitriol; Cell Division; Cell Transformation, Neoplastic; Growth Substances; Isoenzymes; Karyotyping; Keratins; Male; Mice; Mice, Nude; Neoplasm Transplantation; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Rats; Rats, Wistar; Receptor, Transforming Growth Factor-beta Type II; Receptors, Androgen; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Transplantation, Heterologous; Tretinoin; Tumor Cells, Cultured

2003
Antibody blockade of the Cripto CFC domain suppresses tumor cell growth in vivo.
    The Journal of clinical investigation, 2003, Volume: 112, Issue:4

    Cripto, a cell surface-associated protein belonging to the EGF-CFC family of growth factor-like molecules, is overexpressed in many human solid tumors, including 70-80% of breast and colon tumors, yet how it promotes cell transformation is unclear. During embryogenesis, Cripto complexes with Alk4 via its unique cysteine-rich CFC domain to facilitate signaling by the TGF-beta ligand Nodal. We report, for the first time to our knowledge, that Cripto can directly bind to another TGF-beta ligand, Activin B, and that Cripto overexpression blocks Activin B growth inhibition of breast cancer cells. This result suggests a novel mechanism for antagonizing Activin signaling that could promote tumorigenesis by deregulating growth homeostasis. We show that an anti-CFC domain antibody, A8.G3.5, both disrupts Cripto-Nodal signaling and reverses Cripto blockade of Activin B-induced growth suppression by blocking Cripto's association with either Alk4 or Activin B. In two xenograft models, testicular and colon cancer, A8.G3.5 inhibited tumor cell growth by up to 70%. Both Nodal and Activin B expression was found in the xenograft tumor, suggesting that either ligand could be promoting tumorigenesis. These data validate that functional blockade of Cripto inhibits tumor growth and highlight antibodies that block Cripto signaling mediated through its CFC domain as an important class of antibodies for further therapeutic development.

    Topics: Activin Receptors, Type I; Activins; Animals; Antibodies, Monoclonal; Breast Neoplasms; Cell Division; Cell Separation; Cell Transformation, Neoplastic; CHO Cells; Cricetinae; Dose-Response Relationship, Drug; Epidermal Growth Factor; Epitopes; Flow Cytometry; GPI-Linked Proteins; Humans; Immunoblotting; Immunoglobulin G; Immunohistochemistry; Intercellular Signaling Peptides and Proteins; Ligands; Male; Membrane Glycoproteins; Mice; Mice, Nude; Neoplasm Proteins; Neoplasm Transplantation; Nodal Protein; Plasmids; Precipitin Tests; Protein Binding; Protein Structure, Tertiary; Proteins; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Time Factors; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
Tumor suppressor function of Betaig-h3 gene in radiation carcinogenesis.
    Advances in space research : the official journal of the Committee on Space Research (COSPAR), 2003, Volume: 31, Issue:6

    Interaction between cell and extracellular matrix (ECM) plays a crucial role in tumor invasiveness and metastasis. Using an immortalized human bronchial epithelial (BEP2D) cell model, we showed previously that expression of a list of genes including Betaig-h3 (induced by transforming growth factor-beta), DCC (deleted in colorectal cancer), p21(cipl), c-fos, Heat shock protein (HSP27) and cytokeratin 14 were differentially expressed in several independently generated, radiation-induced tumor cell lines (TL1-TL5) relative to parental BEP2D cells. Our previous data further demonstrated that loss of tumor suppressor gene(s) as a likely mechanism of radiation carcinogenesis. In the present study, we chose Betaig-h3 and DCC that were downregulated in tumorigenic cells for further study. Restored expression of Betaig-h3 gene, not DCC gene, by transfecting cDNA into tumor cells resulted in a significant reduction in tumor growth. While integrin receptor alpha 5 beta 1 was overexpressed in tumor cells, its expression was corrected to the level found in control BEP2D cells after Betaig-h3 transfection. These data suggest that Betaig-h3 gene is involved in tumor progression by regulating integrin alpha 5 beta 1 receptor. Furthermore, exogenous TGF- beta 1 induced expression of Betaig-h3 gene and inhibited the growth of both control and tumorigenic BEP2D cells. Therefore, downregulation of Betaig-h3 gene may results from the decreased expression of upstream mediators such as TGF-beta. The findings provide strong evidence that the Betaig-h3 gene has tumor suppressor function in radiation-induced tumorigenic human bronchial epithelial cells and suggest a potential target for interventional therapy.

    Topics: Alpha Particles; Animals; Bronchi; Cell Line, Transformed; Cell Transformation, Neoplastic; Epithelial Cells; Extracellular Matrix; Extracellular Matrix Proteins; Gene Expression; Genes, DCC; Genes, Tumor Suppressor; Humans; Integrin alpha5beta1; Mice; Mice, Nude; Neoplasms, Radiation-Induced; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
TGF-beta switches from tumor suppressor to prometastatic factor in a model of breast cancer progression.
    The Journal of clinical investigation, 2003, Volume: 112, Issue:7

    The TGF-beta signaling network plays a complex role in carcinogenesis because it has the potential to act as either a tumor suppressor or a pro-oncogenic pathway. Currently, it is not known whether TGF-beta can switch from tumor suppressor to pro-oncogenic factor during the course of carcinogenic progression in a single cell lineage with a defined initiating oncogenic event or whether the specific nature of the response is determined by cell type and molecular etiology. To address this question, we have introduced a dominant negative type II TGF-beta receptor into a series of genetically related human breast-derived cell lines representing different stages in the progression process. We show that decreased TGF-beta responsiveness alone cannot initiate tumorigenesis but that it can cooperate with an initiating oncogenic lesion to make a premalignant breast cell tumorigenic and a low-grade tumorigenic cell line histologically and proliferatively more aggressive. In a high-grade tumorigenic cell line, however, reduced TGF-beta responsiveness has no effect on primary tumorigenesis but significantly decreases metastasis. Our results demonstrate a causal role for loss of TGF-beta responsiveness in promoting breast cancer progression up to the stage of advanced, histologically aggressive, but nonmetastatic disease and suggest that at that point TGF-beta switches from tumor suppressor to prometastatic factor.

    Topics: Animals; Antineoplastic Agents; Cell Transformation, Neoplastic; Disease Progression; Female; Humans; Mammary Neoplasms, Experimental; Mice; Neoplasm Metastasis; Neoplasm Transplantation; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Transplantation, Heterologous; Tumor Cells, Cultured

2003
The proto-oncoprotein c-Fos negatively regulates hepatocellular tumorigenesis.
    Oncogene, 2003, Oct-02, Volume: 22, Issue:43

    Hepatocytes adopt an invasive and metastatic phenotype caused by the cooperation of transforming growth factor (TGF)-beta and oncogenic Ha-Ras. In the initial phase of this process, c-Fos is rapidly induced by TGF-beta, but then decreases to undetectable levels. Here, we investigated the functional implications of c-Fos activation and its contribution to hepatocellular tumorigenesis. By employing conditional c-Fos expression, we observed that continuous activation of c-Fos and consequently AP-1 activity leads to depolarization of differentiated murine epithelial hepatocytes. Most remarkably, this change in morphology was associated with inhibition of proliferation and induction of cell death. Coexpression of antiapoptotic Bcl-XL or scavenging of reactive oxygen species was sufficient to prevent the c-Fos-mediated phenotype. In contrast, the cooperation of c-Fos with oncogenic Ha-Ras or a Ras mutant selectively activating the MAPK pathway even enhanced c-Fos-induced effects. Showing the negative role in hepatocellular tumorigenesis, c-Fos repressed oncogenic Ras-driven anchorage-independent growth in vitro and strongly suppressed tumour formation in vivo. Taken together, we demonstrate that c-Fos modulates plasticity of epithelial hepatocytes and acts tumour suppressive in neoplastic hepatocytes by stimulating cell cycle inhibition and cell death.

    Topics: Animals; Apoptosis; bcl-X Protein; Carcinoma, Hepatocellular; CCAAT-Enhancer-Binding Proteins; Cell Death; Cell Division; Cell Line; Cell Survival; Cell Transformation, Neoplastic; Epithelial Cells; Flow Cytometry; Genes, ras; Liver Neoplasms; MAP Kinase Signaling System; Mice; Mice, Inbred BALB C; Phenotype; Proto-Oncogene Proteins c-bcl-2; Proto-Oncogene Proteins c-fos; Reactive Oxygen Species; Signal Transduction; Time Factors; Transcription Factor AP-1; Transcription Factor CHOP; Transcription Factors; Transfection; Transforming Growth Factor beta; Up-Regulation

2003
Cyclic AMP inhibits TGFbeta1-induced cell-scattering and invasiveness in murine-transformed keratinocytes.
    International journal of cancer, 2003, Dec-10, Volume: 107, Issue:5

    Mouse-transformed keratinocytes cultured in the presence of transforming growth factor beta1 (TGF-beta1) acquire an array of morphologic and functional properties that give rise to a migratory phenotype that expresses mesenchymal molecular markers. This cellular conversion involves activation of the Ras-ERK pathway, enhancement of urokinase (uPA) and matrix metalloproteinase-9 (MMP-9) expression and induction of invasiveness. In our present work, we demonstrate that cAMP and forskolin are able to prevent the expression of these mesenchymal properties, probably due to blockade of the Ras-ERK pathway. Our results also show that cAMP and forskolin are able to abolish the TGF-beta1-induced reorganization of the actin cytoskeleton that is characteristic of the mesenchymal phenotype and also inhibits the disruption of the E-cadherin cell to cell interactions. The latter responses seem to depend on the activity of protein kinase A, as demonstrated by the activation of the Ras-ERK pathway by specific protein kinase A inhibitors.

    Topics: Animals; Cell Line, Transformed; Cell Movement; Cell Transformation, Neoplastic; Colforsin; Cyclic AMP; Enzyme Inhibitors; Flavonoids; Gene Expression Regulation, Enzymologic; Humans; Keratinocytes; Matrix Metalloproteinase 9; Mice; Neoplasm Invasiveness; Recombinant Proteins; Transforming Growth Factor beta; Transforming Growth Factor beta1; Urokinase-Type Plasminogen Activator

2003
Smad7 but not Smad6 cooperates with oncogenic ras to cause malignant conversion in a mouse model for squamous cell carcinoma.
    Cancer research, 2003, Nov-15, Volume: 63, Issue:22

    Smad7 and Smad6 are inhibitory Smads that block transforming growth factor-beta (TGF-beta) superfamily signal transduction. Smad7 is overexpressed in chemically induced mouse epidermal tumors, where oncogenic activation of c-ras is a frequent event. To test the role of Smad7 overexpression in tumor progression, we used retroviruses to transduce Smad7 or Smad6 and v-ras(Ha) into primary mouse keratinocytes. By itself, Smad7 transiently enhanced keratinocyte proliferation, blocked normal differentiation, and induced keratin 8, a marker of malignant conversion, but did not cause tumor formation. Smad7 extended the in vitro life span, suppressed senescence, and increased transformation frequency 3-fold of primary keratinocytes coexpressing v-ras(Ha). Smad7/v-ras(Ha) coinfected keratinocytes rapidly progressed to squamous cell carcinomas in vivo, whereas pBabe/v-ras(Ha)- or Smad6/v-ras(Ha)-transduced keratinocytes formed only benign papillomas. Smad7/v-ras(Ha) tumors had elevated proliferation and defective nuclear localizaton of Smad2, Smad3, and Smad5, whereas only Smad5 was altered in Smad6/v-ras(Ha) tumors. Smad7 overexpression in vitro induced epidermal growth factor (EGF)-like growth factors TGF-alpha, heparin binding-EGF, amphiregulin, and EGF receptor tyrosine phosphorylation as well as the EGF-CFC growth factor cripto-1. TGF-alpha and cripto-1 were also overexpressed in Smad7/v-ras(Ha) tumors. These results suggest that Smad7 overexpression accelerates tumor progression through inhibition of TGF-beta superfamily signaling and up-regulation of the EGF-like superfamily of growth factors. This is the first demonstration that Smad7 overexpression can cause malignant conversion in a multistage cancer model and suggests that it may have an important role in the pathogenesis of human cancer.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Disease Models, Animal; DNA-Binding Proteins; Genes, ras; Keratinocytes; Mice; Mice, Inbred BALB C; Mice, Nude; NIH 3T3 Cells; Papilloma; Retroviridae; Signal Transduction; Smad6 Protein; Smad7 Protein; Trans-Activators; Transduction, Genetic; Transforming Growth Factor beta; Up-Regulation

2003
p38 mitogen-activated protein kinase is required for TGFbeta-mediated fibroblastic transdifferentiation and cell migration.
    Journal of cell science, 2002, Aug-01, Volume: 115, Issue:Pt 15

    Transforming growth factor beta (TGFbeta) contributes to tumor progression by inducing an epithelial to mesenchymal transdifferentiation (EMT) and cell migration. We found that TGFbeta-induced EMT was blocked by inhibiting activation of p38 mitogen-activated protein kinase (MAPK) with H-7, a protein kinase C inhibitor, and with SB202190, a direct inhibitor of p38MAPK. Inhibition of the p38MAPK pathway affected TGFbeta-mediated phosphorylation of ATF2, but did not inhibit phosphorylation of Smad2. SB202190 impaired TGFbeta-mediated changes in cell shape and reorganization of the actin cytoskeleton. Forced expression of dominant-negative (DN) MAPK kinase 3 (MKK3) inhibited TGFbeta-mediated activation of p38MAPK and EMT. Expression of DN-p38alpha impaired TGFbeta-induced EMT. Inhibition of p38MAPK blocked TGFbeta-induced migration of non-tumor and tumor mammary epithelial cells. TGFbeta induced activation of the p38MAPK pathway within 15 minutes. Expression of TGFbeta type II (TbetaRII) and type I (TbetaRI/Alk5) kinase-inactive receptors blocked EMT and activation of p38MAPK, whereas expression of constitutively active Alk5-T204D resulted in EMT and phosphorylation of MKK3/6 and p38MAPK. Finally, dominant-negative Rac1N17 blocked TGFbeta-induced activation of the p38MAPK pathway and EMT, suggesting that Rac1 mediates activation of the p38MAPK pathway. These studies suggest that the p38MAPK pathway is required for TGFbeta-mediated EMT and cell migration.

    Topics: Actin Cytoskeleton; Activating Transcription Factor 2; Animals; Cell Differentiation; Cell Movement; Cell Size; Cell Transformation, Neoplastic; Cyclic AMP Response Element-Binding Protein; DNA-Binding Proteins; Enzyme Inhibitors; Epithelial Cells; Fibroblasts; Humans; Mesoderm; Mice; Mitogen-Activated Protein Kinases; Mutation; p38 Mitogen-Activated Protein Kinases; Plant Proteins; rac1 GTP-Binding Protein; Receptors, Transforming Growth Factor beta; Smad Proteins; Trans-Activators; Transcription Factors; Transforming Growth Factor beta; Tumor Cells, Cultured

2002
Transforming growth factor-beta1 modulates matrix metalloproteinase-9 production through the Ras/MAPK signaling pathway in transformed keratinocytes.
    Biochemical and biophysical research communications, 2002, Aug-16, Volume: 296, Issue:2

    Mouse transformed keratinocytes cultured in the presence of transforming growth factor-beta1 (TGF-beta1) acquire a set of morphological and functional properties giving rise to a more motile phenotype that expresses mesenchymal markers. In this work, we present evidence showing that TGF-beta1 stimulates cellular production of MMP-9 (Gelatinase B), a metalloproteinase that plays an important role in tumoral invasion. Our results demonstrate that TGF-beta1stimulates MMP-9 production and MMP-9 promoter activity in a process that depends of the activation of the Ras-ERK1,2 MAP kinase pathway. The latter was demonstrated by cellular transfection of TGF-beta1-sensitive cells with a RasN17 mutant gene, using PD 098059, a MEK 1,2 inhibitor, and treating cells with anti-sense oligodeoxinucleotides. The enhanced MMP-9 production proved to be an important factor in the acquisition of migratory and invasive properties as shown by the use of a specific inhibitor of MMP-9 (GM6001) that inhibits the TGF-beta1-stimulated invasive and migratory properties of these transformed keratinocytes.

    Topics: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Curcumin; Dipeptides; Enzyme Inhibitors; Flavonoids; Genes, Reporter; Humans; Imidazoles; Keratinocytes; MAP Kinase Signaling System; Matrix Metalloproteinase 9; Matrix Metalloproteinase Inhibitors; Mice; Oligodeoxyribonucleotides, Antisense; Promoter Regions, Genetic; Pyridines; ras Proteins; Recombinant Proteins; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured

2002
TGFbeta1 -mediated epithelial to mesenchymal transition is accompanied by invasion in the SiHa cell line.
    European journal of cell biology, 2002, Volume: 81, Issue:8

    It has recently been suggested by several investigators that the epithelial-mesenchymal transition-inducing capacity of TGFbetas contributes to invasive transition of tumors at later stages of carcinogenesis. In the present study, we examined the possibility of TGFbeta1-stimulated epithelial-mesenchymal transition in SiHa cell line, detailed molecular events in the process, and its possible contribution to the invasive transition of tumors. TGFbeta1-induced epithelial-mesenchymal transition of SiHa cells was based on morphological and biochemical criteria; actin stress fiber formation, focal translocalization of integrin alphav, talin, and vinculin, fibronectin-based matrix assembly at the cell periphery, and translocalization and down-regulation of E-cadherin. TGFbeta1 also stimulated surface expression of integrin alphavbeta3 and FAK activation. Focal translocalization of integrin alphav preceded actin reorganization and fibronectin matrix assembly, and functional blocking of the integrin suppressed actin stress fiber formation. Furthermore, induction of actin reorganization and fibronectin matrix assembly by TGFbeta1 were shown to be mutually independent events. These changes were irreversible because 5 minutes pulse exposure to TGFbeta1 was sufficient to stimulate progress of actin reorganization and fibronectin matrix assembly. In further studies with raft culture, TGFbeta1 was found to stimulate invasion of SiHa cells into a type I collagen gel matrix. In conclusion, TGFbeta1 stimulated epithelial-mesenchymal transition of SiHa cells, indicating a positive role in the invasive transition of tumors.

    Topics: Actins; Carcinoma; Cell Adhesion; Cell Size; Cell Transformation, Neoplastic; Cytoskeletal Proteins; Enzyme Inhibitors; Epithelial Cells; Extracellular Matrix Proteins; Female; Fibronectins; Humans; Integrin alphaV; Mesoderm; Neoplasm Invasiveness; Stress Fibers; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Uterine Cervical Neoplasms

2002
Immunohistochemical detection of hepatocyte growth factor, transforming growth factor-beta and their receptors in epithelial odontogenic tumors.
    Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology, 2002, Volume: 31, Issue:9

    Tumors derived from odontogenic epithelium exhibit considerable variation and are classified into several benign and malignant entities. To clarify the role of growth factors in oncogenesis, cytodifferentiation and progression of epithelial odontogenic tumors, expression of hepatocyte growth factor (HGF), transforming growth factor-beta (TGF-beta) and their receptors were analyzed in these tumors as well as in tooth germs.. Specimens of five tooth germs, 34 ameloblastomas, three calcifying epithelial odontogenic tumors (CEOTs), two clear cell odontogenic tumors (CCOTs), five adenomatoid odontogenic tumors (AOTs), six calcifying odontogenic cysts (COCs) and six malignant ameloblastomas were examined immunohistochemically with the use of antibodies against HGF, TGF-beta and their receptors.. In tooth germs and epithelial odontogenic tumors, immunoreactivity for HGF and TGF-beta was detected in both epithelial and mesenchymal cells, while expression of their receptors was found only in epithelial cells. In tooth germs and main types of ameloblastomas, HGF and TGF-beta reactivity was marked in epithelial cells near the basement membrane, and their receptors were diffusely positive in most epithelial cells. In subtypes of ameloblastomas, reduced expression of HGF, c-Met and TGF-beta and increased reactivity for TGF-beta receptors were detected in keratinizing cells in acanthomatous ameloblastomas, and granular cells in granular cell ameloblastomas demonstrated little or no expression of HGF, TGF-beta or their receptors. As compared with main types of ameloblastomas, basal cell ameloblastomas showed high HGF reactivity, and desmoplastic ameloblastomas exhibited elevated reactivity for TGF-beta and its receptors. Neoplastic cells in CEOTs, AOTs and COCs showed reactivity for HGF, TGF-beta and their receptors. Elevated HGF and TGF-beta reactivity was found in pseudoglandular cells in AOTs, and high expression of their receptors was noted in ghost cells in COCs. Metastasizing ameloblastomas showed similar expression patterns of HGF, TGF-beta and their receptors to those of benign ameloblastomas, while CCOTs and ameloblastic carcinomas had increased HGF expression and low reactivity for TGF-beta and its receptors as compared with benign ameloblastomas.. Immunohistochemical localization of HGF, TGF-beta and their receptors in tooth germs and epithelial odontogenic tumors supports the hypothesis that HGF and TGF-beta act on epithelial cells via paracrine and autocrine mechanisms. Altered expression of the agents in these epithelial odontogenic tumors, especially subtypes of ameloblastomas, AOTs and COCs, suggests that HGF and TGF-beta signaling might affect differentiation of neoplastic odontogenic epithelial cells. Activated HGF/c-Met pathway and reduced TGF-beta signaling in CCOTs and ameloblastic carcinomas may be associated with the malignant potential of these epithelial odontogenic tumors.

    Topics: Cell Transformation, Neoplastic; Epithelial Cells; Gene Expression Regulation, Neoplastic; Hepatocyte Growth Factor; Humans; Immunohistochemistry; Odontogenic Tumors; Proto-Oncogene Proteins c-met; Receptors, Transforming Growth Factor beta; Tooth Germ; Transforming Growth Factor beta

2002
Transforming growth factor-beta and epidermal growth factor synergistically stimulate epithelial to mesenchymal transition (EMT) through a MEK-dependent mechanism in primary cultured pig thyrocytes.
    Journal of cell science, 2002, Nov-15, Volume: 115, Issue:Pt 22

    Enhancement of tumor cell growth and invasiveness by transforming growth factor-beta (TGF-beta) requires constitutive activation of the ras/MAPK pathway. Here we have investigated how MEK activation by epidermal growth factor (EGF) influences the response of fully differentiated and growth-arrested pig thyroid epithelial cells in primary culture to TGF-beta1. The epithelial tightness was maintained after single stimulation with EGF or TGF-beta1 (both 10 ng/ml) for 48 hours. In contrast, co-stimulation abolished the transepithelial resistance and increased the paracellular flux of [(3)H]inulin within 24 hours. Reduced levels of the tight junction proteins claudin-1 and occludin accompanied the loss of barrier function. N-cadherin, expressed only in few cells of untreated or single-stimulated cultures, was at the same time increased 30-fold and co-localised with E-cadherin at adherens junctions in all cells. After 48 hours of co-stimulation, both E- and N-cadherin were downregulated and the cells attained a fibroblast-like morphology and formed multilayers. TGF-beta1 only partially inhibited EGF-induced Erk phosphorylation. The MEK inhibitor U0126 prevented residual Erk phosphorylation and abrogated the synergistic responses to TGF-beta1 and EGF. The observations indicate that concomitant growth factor-induced MEK activation is necessary for TGF-beta1 to convert normal thyroid epithelial cells to a mesenchymal phenotype.

    Topics: Adherens Junctions; Animals; Cadherins; Cell Differentiation; Cell Size; Cell Transformation, Neoplastic; Cells, Cultured; Claudin-1; Down-Regulation; Drug Interactions; Enzyme Inhibitors; Epidermal Growth Factor; Epithelial Cells; MAP Kinase Kinase 1; Membrane Proteins; Mesoderm; Microscopy, Electron; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; Neoplasm Invasiveness; Occludin; Protein Serine-Threonine Kinases; Sus scrofa; Thyroid Gland; Tight Junctions; Transforming Growth Factor beta

2002
Rapamycin potentiates transforming growth factor beta-induced growth arrest in nontransformed, oncogene-transformed, and human cancer cells.
    Molecular and cellular biology, 2002, Volume: 22, Issue:23

    Transforming growth factor beta (TGF-beta) induces cell cycle arrest of most nontransformed epithelial cell lines. In contrast, many human carcinomas are refractory to the growth-inhibitory effect of TGF-beta. TGF-beta overexpression inhibits tumorigenesis, and abolition of TGF-beta signaling accelerates tumorigenesis, suggesting that TGF-beta acts as a tumor suppressor in mouse models of cancer. A screen to identify agents that potentiate TGF-beta-induced growth arrest demonstrated that the potential anticancer agent rapamycin cooperated with TGF-beta to induce growth arrest in multiple cell lines. Rapamycin also augmented the ability of TGF-beta to inhibit the proliferation of E2F1-, c-Myc-, and (V12)H-Ras-transformed cells, even though these cells were insensitive to TGF-beta-mediated growth arrest in the absence of rapamycin. Rapamycin potentiation of TGF-beta-induced growth arrest could not be explained by increases in TGF-beta receptor levels or rapamycin-induced dissociation of FKBP12 from the TGF-beta type I receptor. Significantly, TGF-beta and rapamycin cooperated to induce growth inhibition of human carcinoma cells that are resistant to TGF-beta-induced growth arrest, and arrest correlated with a suppression of Cdk2 kinase activity. Inhibition of Cdk2 activity was associated with increased binding of p21 and p27 to Cdk2 and decreased phosphorylation of Cdk2 on Thr(160). Increased p21 and p27 binding to Cdk2 was accompanied by decreased p130, p107, and E2F4 binding to Cdk2. Together, these results indicate that rapamycin and TGF-beta cooperate to inhibit the proliferation of nontransformed cells and cancer cells by acting in concert to inhibit Cdk2 activity.

    Topics: Animals; Antibiotics, Antineoplastic; Carcinoma; CDC2-CDC28 Kinases; Cell Cycle Proteins; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase 2; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinase Inhibitor p27; Cyclin-Dependent Kinases; Cyclins; DNA-Binding Proteins; E2F4 Transcription Factor; Enzyme Inhibitors; Epithelial Cells; Genes, Reporter; Growth Inhibitors; Humans; Nuclear Proteins; Phosphoproteins; Protein Binding; Protein Serine-Threonine Kinases; Proteins; Retinoblastoma Protein; Retinoblastoma-Like Protein p107; Retinoblastoma-Like Protein p130; Signal Transduction; Sirolimus; Tacrolimus Binding Proteins; Transcription Factors; Transforming Growth Factor beta; Tumor Suppressor Proteins

2002
A mechanism in Epstein-Barr virus oncogenesis: inhibition of transforming growth factor-beta 1-mediated induction of MAPK/p21 by LMP1.
    Virology, 2002, Oct-25, Volume: 302, Issue:2

    Epstein-Barr virus (EBV)-infected, gastric epithelial cell line GT38 is resistant to TGF-beta 1-mediated growth inhibition and apoptosis, although TGF-beta 1 partially induces EBV reactivation in the cells. These findings indicate that abnormalities exist in these cells in the TGF-beta 1-mediated signaling pathway, influencing growth inhibition and apoptosis. In order to characterize the steps with abnormalities, we analyzed the TGF-beta 1/MAPK/p21 pathway in the cells. TGF-beta 1 activated MAPK (ERK 1/2) and p21 in the TGF-beta 1-susceptible cell line HSC-39 but not in GT38 cells. GT38 cells had higher constitutive levels of ERK 1/2 phosphorylation and p21 expression than did HSC-39 cells. U0126, a specific inhibitor of MEK, suppressed TGF-beta 1-mediated ERK 1/2 phosphorylation and p21 induction in HSC-39 cells and constitutive ERK 1/2 phosphorylation in GT38 cells. EBV latent membrane protein 1 (LMP1) induced constitutive ERK 1/2 phosphorylation and NF-kappa B activation in LMP1-transfected HSC-39 cells, which then became resistant to TGF-beta 1-mediated growth inhibition, TGF-beta 1-mediated ERK 1/2 phosphorylation, and p21 induction, and proliferated in low-serum medium. These results are consistent with the conclusion that the TGF-beta 1/MAPK/p21 pathway is required for TGF-beta 1-mediated growth inhibition, and that the resistance to TGF in GT38 cells is derived from constitutive MAPK phosphorylation induced by LMP1.

    Topics: Cell Line, Transformed; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Gene Expression Regulation; Herpesvirus 4, Human; Humans; Mitogen-Activated Protein Kinases; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Viral Matrix Proteins

2002
[Regulation of Smad7 gene by TGF-beta 1 in process of malignant transformation].
    Ai zheng = Aizheng = Chinese journal of cancer, 2002, Volume: 21, Issue:2

    Escape from transforming growth factor-beta(TGF-beta)-induced inhibition of growth and proliferation may contribute to tumorigenesis. Smad7 is inhibitory Smads of TGF-beta s signal transduction pathway and prevents TGF-beta signaling. The disorder of Smad7 may lead to the perturbation of TGF-beta signal pathway. In this study, The authors analyzed the expression of Smad7 mRNA and the regulation of Smad7 gene by TGF-beta 1 in the process of malignant transformation of BEP2D cells to investigate the mechanism of cells malignant transformation.. Cells were cultured and stimulated with TGF-beta 1 followed by RNA extraction. Purified total RNA from TGF-beta 1 treated cells and untreated controls and performed an expression analysis with a human Smad7-specific probe applying Northern blot. As a loading control for the Northern experiment, the membrane was hybridized with a human glyceraldehyde-3-phosphate dehydrogenase(GAPDH) probe. Proteins were extracted from BEP2D and BERP35T-2 cells, then perform Western blot to examine the expression level of TGF-beta 1.. Before stimulation with TGF-beta 1, the expression level of Smad7 in the BERP35T-2 cells were higher than that in the BEP2D cells. When stimulated with TGF-beta 1, Smad7 expression levels was upregulated evidently in BEP2D cells, but not significant in BERP35T-2 cells. The expression level of endogenetic TGF-beta 1, BERP35T-2 cells was a little higher than BEP2D cells.. Over expression of Smad7 mRNA and down-regulation of the cells' responsiveness to TGF-beta 1 in human lung cancer cell line which induced by alpha-particles should be one of the mechanism of radiation induced lung cancer.

    Topics: Blotting, Western; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Gene Expression Regulation; Humans; RNA, Messenger; Smad7 Protein; Trans-Activators; Transforming Growth Factor beta; Transforming Growth Factor beta1

2002
Release of transforming growth factor beta-1 in a vestibular schwannoma cell line.
    Acta oto-laryngologica, 2002, Volume: 122, Issue:7

    Vestibular schwannoma (VS) often displays an irregular growth pattern due to factors which remain unknown. In this study, VS cell lines from tissue removed during surgery were cultured in order to assess the presence of transforming growth factor beta-1 (TGFbeta-1) a multifunctional polypeptide that has different pleiotropic effects on various tissues. The antibody-binding inhibition assay technique was used. The proliferation rate of VS cell lines in vitro was assessed by [3H]thymidine incorporation, with determinations carried out on days 0, 1, 2 and 4 after removal of fetal calf serum (FCS) from the medium. After 48 h of FCS deprivation, cell growth decreased. TGFbeta-1 release also decreased progressively 24 h after removing FCS from the medium, reaching an overall decrease of 50% after 3 days. The cell culturing procedure appeared suitable for VS cells, in that VS cultures were viable for at least 4 months. TGFbeta-1 was initially shown to be released in significant amounts, but also to decrease progressively to values five times lower after 4 days. These data thus support the possibility of a significant association between TGFbeta-1 release and VS cell replication.

    Topics: Antigen-Antibody Complex; Cell Movement; Cell Transformation, Neoplastic; Culture Techniques; Enzyme-Linked Immunosorbent Assay; Humans; Neuroma, Acoustic; Time Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1

2002
cDNA array analysis of SPARC-modulated changes in glioma gene expression.
    Journal of neuro-oncology, 2002, Volume: 60, Issue:3

    We have demonstrated that secreted protein acidic and rich in cysteine (SPARC) is highly expressed in human gliomas and it promotes glioma invasion and delays tumor growth in vitro and in vivo. cDNA array analyses were performed to determine whether SPARC, which interacts at the cell surface, has an impact on intracellular signaling and downstream gene expression changes, which might account for some of its effects on invasion and growth. Using a doxycycline (dox)-controlled gene expression system, two cDNA array analyses were performed using a parental U87T2 clone (-SPARC) transfected with the dox-controlled transactivator and a U87T2 parental-derived SPARC-transfected clone, A2b2 (+SPARC). Array analysis performed between the parental and the SPARC-transfected clone (-dox) identified 13 upregulated genes and 14 downregulated genes. With the exception of PAI-1 and MMP2, the identified genes are novel with respect to SPARC's mechanism of action. Array analysis performed using the SPARC-transfected clone ( +/- dox) identified 2 types of gene regulation; one reversible upon SPARC suppression, the other irreversible. Two of the SPARC-induced genes, BIGH3 (irreversible by dox) and PAI-1 (reversible by dox) were further studied in additional SPARC-transfected clones, human astrocytoma tissues, and human glioma cell lines by RT-PCR and Northern blot analyses. The results indicate that: (1) the array results were validated, (2) the dox regulation was validated, and (3) the differential expression identified by the array analyses was present between normal brain and in human astrocytoma tissues and cell lines. Therefore, we conclude that these cDNA array analyses provide candidate genes involved in SPARC-mediated effects on glioma cell cycle progression, signaling, and migration, and that SPARC may induce reversible and irreversible gene expression changes. Further investigation of these candidates may shed insights into SPARC's role in glioma cell proliferation and invasion, and potential use as a therapeutic target.

    Topics: Anti-Bacterial Agents; Astrocytes; Astrocytoma; Blotting, Northern; Brain; Cell Transformation, Neoplastic; Doxycycline; Extracellular Matrix Proteins; Fetus; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Glioma; Humans; Neoplasm Invasiveness; Neoplasm Proteins; Oligonucleotide Array Sequence Analysis; Osteonectin; Reverse Transcriptase Polymerase Chain Reaction; RNA-Binding Proteins; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2002
Transforming growth factor-beta and Ras regulate the VEGF/VEGF-receptor system during tumor angiogenesis.
    International journal of cancer, 2002, Jan-10, Volume: 97, Issue:2

    The formation of new microvasculature by capillary sprouting, or angiogenesis, is a prerequisite for solid tumor growth. The genetic alterations required to activate the angiogenic program in tumor angiogenesis are still only vaguely known, but dominantly acting oncoproteins may have a much greater impact than previously realized. Here we have studied the consequences of oncogenic transformation on tumor angiogenesis in a mouse mammary carcinoma model. We provide evidence that the expression of vascular endothelial growth factor (VEGF) and of the VEGF receptor-2 (Flk-1), a signaling system centrally involved in tumor angiogenesis, occurs efficiently in tumors formed by Ras-transformed mammary epithelial cells and that both TGF-beta1 and hypoxia are potent inducers of VEGF expression in these cells. VEGF induction in the tumor periphery is mainly triggered by TGF-beta1, whereas VEGF expression in perinecrotic areas is regulated by both hypoxia and TGF-beta1. As the Ras-transformed tumor cells convert into migrating, fibroblastoid cells that start to produce TGF-beta during tumor progression, the TGF-beta effect on VEGF expression becomes propagated throughout the tumor tissue. Thus, in progressed tumors, areas of TGF-beta1 activation and hypoxia may overlap and hence cooperate to induce VEGF expression and angiogenesis. Nevertheless, the overexpression of VEGF in non-Ras-transformed mouse mammary epithelial cells was not sufficient to promote vascularization in vivo. Based on these findings, we conclude that amongst the multiple mutations that render a normal cell tumorigenic, oncogenic Ras is a major player that in conjunction with the tumor's micro-environment sets the stage for tumor cell invasion and angiogenesis.

    Topics: Animals; Cell Movement; Cell Transformation, Neoplastic; Cells, Cultured; Endothelial Growth Factors; Endothelium, Vascular; Epithelial Cells; Fibroblasts; Glioma; Humans; Hypoxia; Immunoblotting; Immunoenzyme Techniques; In Situ Hybridization; Lymphokines; Mammary Neoplasms, Experimental; Mice; Mice, Inbred BALB C; Neoplasm Invasiveness; Neovascularization, Pathologic; Oncogene Protein p21(ras); Rats; Receptor Protein-Tyrosine Kinases; Receptors, Growth Factor; Receptors, Vascular Endothelial Growth Factor; RNA, Messenger; Transforming Growth Factor beta; Umbilical Veins; Up-Regulation; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

2002
Inhibition of transforming growth factor beta signaling and Smad-dependent activation of transcription by the Latent Membrane Protein 1 of Epstein-Barr virus.
    The Journal of biological chemistry, 2002, Mar-15, Volume: 277, Issue:11

    Inhibition of transforming growth factor beta (TGFbeta) signaling by the Epstein-Barr virus Latent Membrane Protein 1 (LMP1) may account, at least in part, for the oncogenic activity of LMP1. We found that LMP1 is a potent inhibitor of TGFbeta signaling and Smad-dependent activation of transcription in 293 epithelial cells and COS-7 fibroblasts. LMP1 strongly inhibited the uninduced and the Smad-inducible activity of the promoters of the human p21/WAF1/Cip1 gene and the mouse Smad7 gene. Inhibition of TGFbeta signaling and Smad-dependent activation of transcription by LMP1 was greatly reduced by deletion of both C-terminal activating regions 1 and 2 of LMP1 as well as by overexpression of a non-degradable form of IkappaB. In contrast, specific inhibitors of p38 kinase or MEK kinase did not reverse the inhibitory activity of LMP1. TGFbeta signaling was enhanced by overexpression of dominant negative forms of the LMP1 effectors TRAF2, NIK, and IKKbeta and was abolished by overexpression of p65/RelA or a p50/p65 fusion protein. Deletion of the transactivation domain of p65 abolished its inhibitory activity. Immunoblotting and immunofluorescence microscopy indicated that suppression of TGFbeta signaling and Smad transcriptional activity by LMP1 was not due to Smad degradation or cytoplasmic retention suggesting that LMP1 affects the nuclear function of Smad proteins. Our data are consistent with an essential role of NF-kappaB activation by LMP1 in the inhibition of TGFbeta signaling and Smad-mediated transcriptional responses.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Fungal Proteins; Humans; NF-kappa B; Saccharomyces cerevisiae Proteins; Smad Proteins; Smad3 Protein; Trans-Activators; Transcription Factors; Transcriptional Activation; Transforming Growth Factor beta; Viral Matrix Proteins

2002
Ras and TGF[beta] cooperatively regulate epithelial cell plasticity and metastasis: dissection of Ras signaling pathways.
    The Journal of cell biology, 2002, Jan-21, Volume: 156, Issue:2

    Multistep carcinogenesis involves more than six discrete events also important in normal development and cell behavior. Of these, local invasion and metastasis cause most cancer deaths but are the least well understood molecularly. We employed a combined in vitro/in vivo carcinogenesis model, that is, polarized Ha-Ras-transformed mammary epithelial cells (EpRas), to dissect the role of Ras downstream signaling pathways in epithelial cell plasticity, tumorigenesis, and metastasis. Ha-Ras cooperates with transforming growth factor beta (TGFbeta) to cause epithelial mesenchymal transition (EMT) characterized by spindle-like cell morphology, loss of epithelial markers, and induction of mesenchymal markers. EMT requires continuous TGFbeta receptor (TGFbeta-R) and oncogenic Ras signaling and is stabilized by autocrine TGFbeta production. In contrast, fibroblast growth factors, hepatocyte growth factor/scatter factor, or TGFbeta alone induce scattering, a spindle-like cell phenotype fully reversible after factor withdrawal, which does not involve sustained marker changes. Using specific inhibitors and effector-specific Ras mutants, we show that a hyperactive Raf/mitogen-activated protein kinase (MAPK) is required for EMT, whereas activation of phosphatidylinositol 3-kinase (PI3K) causes scattering and protects from TGFbeta-induced apoptosis. Hyperactivation of the PI3K pathway or the Raf/MAPK pathway are sufficient for tumorigenesis, whereas EMT in vivo and metastasis required a hyperactive Raf/MAPK pathway. Thus, EMT seems to be a close in vitro correlate of metastasis, both requiring synergism between TGFbeta-R and Raf/MAPK signaling.

    Topics: Animals; Apoptosis; Blotting, Western; Cell Line; Cell Movement; Cell Transformation, Neoplastic; Epithelial Cells; Hepatocyte Growth Factor; Mammary Glands, Animal; MAP Kinase Signaling System; Mesoderm; Mice; Microscopy, Confocal; Microscopy, Fluorescence; Mitogen-Activated Protein Kinase Kinases; Molecular Weight; Mutation; Neoplasm Metastasis; Oncogene Protein p21(ras); Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Signal Transduction; Transforming Growth Factor beta

2002
Genetics of adenocarcinomas of the small intestine: frequent deletions at chromosome 18q and mutations of the SMAD4 gene.
    Oncogene, 2002, Jan-03, Volume: 21, Issue:1

    The small intestinal mucosa makes up about 90% of the total surface of the gastrointestinal tract. However, adenocarcinomas arise rarely in this location. To elucidate genetic alterations underlying tumour development in the small intestine we investigated 17 sporadic adenocarcinomas. By comparative genomic hybridization recurrent gains of chromosomal material were found at chromosomes 7, 8, 13q, and 20 (5/17, each), while non-random losses were seen at 8p, 17p (4/17, each), and 18 (8/17 cases). Deletions at 5q, the location of the APC tumour suppressor gene, were seen in three cases. Microsatellite analysis with markers on chromosomal arms 1p, 5q, 8p, 17p, 18q, 19p, and 22q revealed a microsatellite instable phenotype in two cases and a high frequency of loss at 18q21-q22 (80%). Given the high incidence of 18q21-q22 deletions, we performed sequencing analysis of SMAD4, a downstream component of the TGFbeta-pathway, located at 18q21. Four tumours displayed mutations in highly conserved domains of the gene indicating disruption of TGFbeta-signalling. Our data reveal complex genetic alterations in sporadic small intestinal carcinomas. However, most tumours share deletions of 18q21-q22, which frequently target SMAD4. This indicates that disruption of TGFbeta-signalling plays a critical role in small intestinal tumorigenesis.

    Topics: Adenocarcinoma; Adult; Aged; Aged, 80 and over; Alleles; Amino Acid Substitution; Cell Transformation, Neoplastic; Chromosome Aberrations; Chromosomes, Human, Pair 18; Chromosomes, Human, Pair 5; Codon; DNA Mutational Analysis; DNA-Binding Proteins; DNA, Neoplasm; Female; Humans; Intestinal Mucosa; Intestinal Neoplasms; Intestine, Small; Loss of Heterozygosity; Male; Microsatellite Repeats; Middle Aged; Mutation, Missense; Neoplasm Proteins; Nucleic Acid Hybridization; Point Mutation; Retrospective Studies; Sequence Deletion; Signal Transduction; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2002
Synergy between truncated c-Met (cyto-Met) and c-Myc in liver oncogenesis: importance of TGF-beta signalling in the control of liver homeostasis and transformation.
    Oncogene, 2002, Feb-21, Volume: 21, Issue:9

    The c-Met tyrosine kinase receptor and its ligand, Hepatocyte Growth Factor/ Scatter Factor, have been implicated in human cancer. We have previously described that the transgenic expression of a truncated form of human c-Met (cyto-Met) in the liver confers resistance to several apoptotic stimuli. Here we show the impact of cyto-Met expression on liver proliferation and transformation. Despite a sixfold increase of hepatocyte proliferation, adult transgenic livers displayed normal size and architecture. We present evidence showing that activation of TGF-beta1 signalling controls the liver mass in cyto-Met mice. The oncogenic potential of cyto-Met was further assessed in the context of c-Myc-induced hepatocarcinogenesis, using WHV/c-Myc transgenic mice. Co-expression of cyto-Met and c-Myc further enhanced hepatocyte proliferation and caused a dramatic acceleration of the Myc-induced tumorigenesis, leading to the emergence of hepatocarcinomas in 3-4-month-old animals. Importantly, the TGF-beta receptor type II expression was strongly downregulated in most tumours, indicating that impairment of TGF-beta1-mediated growth inhibition plays a major role in accelerated neoplastic development. The strong potential of cyto-Met for oncogenic cooperation without direct transforming activity designates cyto-Met mice as an ideal tool for studying the early steps of multistage hepatocarcinogenesis and for identification of prognostic markers of transformation.

    Topics: Animals; Apoptosis; Blotting, Western; Cell Division; Cell Transformation, Neoplastic; Down-Regulation; Gene Expression Regulation, Neoplastic; Hepatitis B Virus, Woodchuck; Hepatocytes; Homeostasis; Humans; Hyperplasia; Liver Neoplasms; Mice; Mice, Transgenic; Organ Size; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-met; Proto-Oncogene Proteins c-myc; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Messenger; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factor beta1; Transgenes

2002
Hepatocytes convert to a fibroblastoid phenotype through the cooperation of TGF-beta1 and Ha-Ras: steps towards invasiveness.
    Journal of cell science, 2002, Mar-15, Volume: 115, Issue:Pt 6

    In hepatocarcinogenesis, it is an open question whether transforming growth factor (TGF)-beta1 provides a tumor-suppressive or a tumor-promoting role. To address this question, we employed immortalized murine hepatocytes, which display a high degree of differentiation and, expectedly, arrest in the G1 phase under exposure to TGF-beta1. These hepatocytes maintain epithelial polarization upon expression of oncogenic Ha-Ras. However, Ras-transformed hepatocytes rapidly convert to a spindle-shaped, fibroblastoid morphology upon treatment with TGF-beta1, which no longer inhibits proliferation. This epithelial to fibroblastoid conversion (EFC) is accompanied by disruption of intercellular contacts and remodeling of the cytoskeletal framework. Fibroblastoid derivatives form elongated branching cords in collagen gels and grow to severely vascularized tumors in vivo, indicating their increased malignancy and even invasive phenotype. Additionally, fibroblastoid cells secrete strongly enhanced levels of TGF-beta1, suggesting an autocrine regulation of TGF-beta signaling. Expression profiling further revealed that the loss of the adhesion component E-cadherin correlates with the upregulation of its transcriptional repressor Snail in fibroblastoid cells. Moreover, the phosphoinositide 3-OH (PI3) kinase pathway was required for the maintenance of EFC, as inhibition of PI3 kinase reverted fibroblastoid cells to an epithelial-like phenotype. Taken together, these data indicate a dual role of TGF-beta1 in hepatocytes: it induces proliferation arrest but provides a crucial function in promoting late malignant events in collaboration with activated Ha-Ras.

    Topics: Autocrine Communication; Cadherins; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epithelium; Fibroblasts; Hepatocytes; MAP Kinase Kinase 1; MAP Kinase Kinase 2; Mitogen-Activated Protein Kinase Kinases; Neoplasm Invasiveness; Neoplasms, Experimental; Phenotype; Phosphatidylinositol 3-Kinases; Protein Serine-Threonine Kinases; Protein-Tyrosine Kinases; Proto-Oncogene Proteins p21(ras); Transforming Growth Factor beta; Transforming Growth Factor beta1; Up-Regulation

2002
Causal relationship between the loss of RUNX3 expression and gastric cancer.
    Cell, 2002, Apr-05, Volume: 109, Issue:1

    Runx3/Pebp2alphaC null mouse gastric mucosa exhibits hyperplasias due to stimulated proliferation and suppressed apoptosis in epithelial cells, and the cells are resistant to growth-inhibitory and apoptosis-inducing action of TGF-beta, indicating that Runx3 is a major growth regulator of gastric epithelial cells. Between 45% and 60% of human gastric cancer cells do not significantly express RUNX3 due to hemizygous deletion and hypermethylation of the RUNX3 promoter region. Tumorigenicity of human gastric cancer cell lines in nude mice was inversely related to their level of RUNX3 expression, and a mutation (R122C) occurring within the conserved Runt domain abolished the tumor-suppressive effect of RUNX3, suggesting that a lack of RUNX3 function is causally related to the genesis and progression of human gastric cancer.

    Topics: Animals; Apoptosis; Cell Division; Cell Transformation, Neoplastic; Core Binding Factor Alpha 3 Subunit; DNA Methylation; DNA-Binding Proteins; Epithelium; Exons; Female; Gastric Mucosa; Gene Deletion; Gene Expression Regulation, Neoplastic; Gene Targeting; Humans; Hyperplasia; Male; Mice; Mice, Knockout; Protein Structure, Tertiary; Stomach; Stomach Neoplasms; Transcription Factors; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Proteins

2002
Changes of TGFbeta1 and TGFbetaRII expression in esophageal precancerous and cancerous lesions: a study of a high-risk population in Henan, northern China.
    Diseases of the esophagus : official journal of the International Society for Diseases of the Esophagus, 2002, Volume: 15, Issue:1

    The level of transforming growth factor beta1 (TGFbeta1) and transforming growth factor betaII receptor (TGFbetaRII) was determined immunohistochemically in normal tissues and tissues with different severities of lesions (basal cell hyperplasia, BCH; dysplasia, DYS; carcinoma in situ, CIS; and squamous cell carcinoma, SCC) from surgically resected human esophagi and esophageal biopsies of symptom-free subjects. The samples were from an area with high esophageal cancer incidence in northern China (Linzhou, formerly Linxian, and nearby county Huixian in Henan Province). Peroxidase immunostain (ABC) and conventional hematoxylin and eosin stain were used. The tissue sections were incubated with antibodies of TGFbeta1 and TGFbetaRII overnight. The immunoreactivity was observed in cytoplasm of the esophageal specimen. From normal to BCH to DYS to CIS and to SCC, the positive immunostaining rates for TGFbeta1 increased significantly (P < 0.05). A linear correlation between the positive immunostaining rates of TGFbeta1 and the different lesions was observed (P < 0.05). From well- to moderately- and poorly differentiated SCC, the positive immunostaining rates for TGFbeta1 decreased gradually, but the difference was not significant (P > 0.05). In contrast, with the lesions progressing from normal to BCH to DYS to CIS and to SCC, the positive immunostaining rates for TGFbetaRII decreased significantly (P < 0.05). From well- to moderately- and poorly differentiated SCC, the positive immunostaining rates for TGFbetaRII decreased significantly (P < 0.05). There was a linear correlation between the positive rates of TGFbetaRII and different lesions and SCC differentiation (P < 0.05). The present results indicated that the alterations of TGFbeta1 and TGFbetaRII is a frequent event in esophageal multistage carcinogenesis, the absent or lower expression of TGFbetaRII may lead to the loss of cell proliferation control by TGFbeta1 and the overexpression of TGFbeta1 may be a negative feedback response caused by the lower expression of TGFbetaRII protein.

    Topics: Adult; Aged; Biomarkers, Tumor; Carcinoma; Cell Transformation, Neoplastic; Culture Techniques; Esophageal Neoplasms; Esophagectomy; Female; Humans; Immunohistochemistry; Male; Middle Aged; Precancerous Conditions; Probability; Prospective Studies; Receptors, Transforming Growth Factor beta; Sampling Studies; Sensitivity and Specificity; Statistics, Nonparametric; Transforming Growth Factor beta; Transforming Growth Factor beta1

2002
Overexpression of Betaig-h3 gene downregulates integrin alpha5beta1 and suppresses tumorigenicity in radiation-induced tumorigenic human bronchial epithelial cells.
    British journal of cancer, 2002, Jun-17, Volume: 86, Issue:12

    Interaction between cell and extracellular matrix plays a crucial role in tumour invasion and metastasis. Using an immortalised human bronchial epithelial (BEP2D) cell model, the study here shows that expression of Betaig-h3 gene, which encodes a secreted adhesion molecule induced by transforming growth factor-beta, is markedly decreased in several independently generated, radiation-induced tumour cell lines (TL1-TL5) relative to parental BEP2D cells. Transfection of Betaig-h3 gene into tumour cells resulted in a significant reduction in tumour growth. While integrin receptor alpha5beta1 was overexpressed in tumour cells, its expression was corrected to the level found in control BEP2D cells after Betaig-h3 transfection. These data suggest that Betaig-h3 gene is involved in tumour progression by regulating integrin receptor alpha5beta1. The findings provide strong evidence that the Betaig-h3 gene has tumour suppressor function in human BEP2D cell model and suggest a potential target for interventional therapy.

    Topics: Animals; Blotting, Northern; Blotting, Western; Bronchi; Cell Transformation, Neoplastic; Cloning, Molecular; DNA Primers; Down-Regulation; Epithelial Cells; Extracellular Matrix Proteins; Gene Expression; Genes, Tumor Suppressor; Humans; Mice; Mice, Nude; Neoplasm Proteins; Oligonucleotide Array Sequence Analysis; Polymerase Chain Reaction; Receptors, Fibronectin; Transfection; Transforming Growth Factor beta

2002
Mutations of the Smad4 gene in acute myelogeneous leukemia and their functional implications in leukemogenesis.
    Oncogene, 2001, Jan-04, Volume: 20, Issue:1

    The Smad family proteins are critical components of the transforming growth factor (TGF)-beta signaling pathway. TGF-beta is a multipotent cytokine that elicits many biological functions. In particular, TGF-beta exhibits effects on the cell cycle manifested by G1-phase arrest, differentiation, or apoptosis of several target cells, suggesting that disruption of TGF-beta signaling pathway could be involved in cancer formation. Here we show one missense mutation of the Smad4 gene in the MH1 domain (P102L) and one frame shift mutation resulting in termination in the MH2 domain (Delta(483 - 552)) in acute myelogeneous leukemia. Both of the mutated Smad4 proteins lack transcriptional activities. Concomitant expression of the P102L mutant with wild-type Smad4 inactivates wild-type Smad4 through inhibiting its DNA-binding ability. The Delta(483 - 552) mutant blocks nuclear translocation of wild-type Smad4 and thus disrupts TGF-beta signaling. This is the first report showing that mutations in the Smad4 gene are associated with the pathogenesis of acute myelogeneous leukemia and the obtained results should provide useful insights into the mechanism whereby disruption of TGF-beta signaling pathway could lead to acute myelogeneous leukemia. Oncogene (2001) 20, 88 - 96.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; COS Cells; DNA; DNA-Binding Proteins; Enzyme Activation; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Genetic Vectors; Growth Inhibitors; HL-60 Cells; Humans; Jurkat Cells; Leukemia, Myeloid, Acute; Mutation; Plasminogen Activator Inhibitor 1; Promoter Regions, Genetic; Protein Binding; Recombinant Fusion Proteins; Serine Proteinase Inhibitors; Signal Transduction; Smad3 Protein; Smad4 Protein; Subcellular Fractions; Trans-Activators; Transcriptional Activation; Transforming Growth Factor beta; Tumor Cells, Cultured

2001
Human T-cell leukemia virus type I oncoprotein Tax represses Smad-dependent transforming growth factor beta signaling through interaction with CREB-binding protein/p300.
    Blood, 2001, Apr-01, Volume: 97, Issue:7

    Human T-cell leukemia virus type I (HTLV-I) Tax is a potent transcriptional regulator that can activate or repress specific cellular genes and that has been proposed to contribute to leukemogenesis in adult T-cell leukemia. Previously, HTLV-I- infected T-cell clones were found to be resistant to growth inhibition by transforming growth factor (TGF)-beta. Here it is shown that Tax can perturb Smad-dependent TGF-beta signaling even though no direct interaction of Tax and Smad proteins could be detected. Importantly, a mutant Tax of CREB-binding protein (CBP)/p300 binding site, could not repress the Smad transactivation function, suggesting that the CBP/p300 binding domain of Tax is essential for the suppression of Smad function. Because both Tax and Smad are known to interact with CBP/p300 for the potentiation of their transcriptional activities, the effect of CBP/p300 on suppression of Smad-mediated transactivation by Tax was examined. Overexpression of CBP/p300 reversed Tax-mediated inhibition of Smad transactivation. Furthermore, Smad could repress Tax transcriptional activation, indicating reciprocal repression between Tax and Smad. These results suggest that Tax interferes with the recruitment of CBP/p300 into transcription initiation complexes on TGF-beta-responsive elements through its binding to CBP/p300. The novel function of Tax as a repressor of TGF-beta signaling may contribute to HTLV-I leukemogenesis. (Blood. 2001;97:2137-2144)

    Topics: Activin Receptors, Type I; Animals; Binding Sites; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Chlorocebus aethiops; COS Cells; DNA-Binding Proteins; Gene Products, tax; Genes, pX; Humans; Liver Neoplasms; Lung; Macromolecular Substances; Mink; Nerve Growth Factors; Nuclear Proteins; Protein Binding; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Recombinant Fusion Proteins; Regulatory Sequences, Nucleic Acid; Signal Transduction; Smad Proteins; Smad2 Protein; Smad3 Protein; Smad4 Protein; Trans-Activators; Transcriptional Activation; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured; Xenopus laevis; Xenopus Proteins

2001
Landscaper seeks remunerative position.
    Gut, 2001, Volume: 48, Issue:5

    Topics: Adenomatous Polyposis Coli; Alleles; Cell Transformation, Neoplastic; DNA-Binding Proteins; Humans; In Situ Hybridization, Fluorescence; Intestinal Mucosa; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta

2001
The corepressor CtBP interacts with Evi-1 to repress transforming growth factor beta signaling.
    Blood, 2001, May-01, Volume: 97, Issue:9

    Evi-1 is a zinc finger nuclear protein whose inappropriate expression leads to leukemic transformation of hematopoietic cells in mice and humans. This was previously shown to block the antiproliferative effect of transforming growth factor beta (TGF-beta). Evi-1 represses TGF-beta signaling by direct interaction with Smad3 through its first zinc finger motif. Here, it is demonstrated that Evi-1 represses Smad-induced transcription by recruiting C-terminal binding protein (CtBP) as a corepressor. Evi-1 associates with CtBP1 through one of the consensus binding motifs, and this association is required for efficient inhibition of TGF-beta signaling. A specific inhibitor for histone deacetylase (HDAc) alleviates Evi-1-mediated repression of TGF-beta signaling, suggesting that HDAc is involved in the transcriptional repression by Evi-1. This identifies a novel function of Evi-1 as a member of corepressor complexes and suggests that aberrant recruitment of corepressors is one of the mechanisms for Evi-1-induced leukemogenesis.

    Topics: Alcohol Oxidoreductases; Animals; Cell Transformation, Neoplastic; COS Cells; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Humans; Leukemia; MDS1 and EVI1 Complex Locus Protein; Mice; Phosphoproteins; Proto-Oncogenes; Repressor Proteins; Signal Transduction; Transcription Factors; Transforming Growth Factor beta; Zinc Fingers

2001
Mechanism for mutational inactivation of the tumor suppressor Smad2.
    Molecular and cellular biology, 2001, Volume: 21, Issue:10

    Transforming growth factor beta (TGF-beta) is a potent natural antiproliferative agent that plays an important role in suppressing tumorigenicity. In numerous tumors, loss of TGF-beta responsiveness is associated with inactivating mutations that can occur in components of this signaling pathway, such as the tumor suppressor Smad2. Although a general framework for how Smads transduce TGF-beta signals has been proposed, the physiological relevance of alterations of Smad2 functions in promoting tumorigenesis is still unknown. Here, we show that expression of Smad2.P445H, a tumor-derived mutation of Smad2 found in human cancer, suppresses the ability of the Smads to mediate TGF-beta-induced growth arrest and transcriptional responses. Smad2.P445H is phosphorylated by the activated TGF-beta receptor at the carboxy-terminal serine residues and associates with Smad3 and Smad4 but is unable to dissociate from the receptor. Upon ligand-induced phosphorylation, Smad2.P445H interacts stably with wild-type Smad2, thereby blocking TGF-beta-induced nuclear accumulation of wild-type Smad2 and Smad2-dependent transcription. The ability of the Smad2.P445H to block the nuclear accumulation of wild-type Smad2 protein reveals a new mechanism for loss of sensitivity to the growth-inhibitory functions of TGF-beta in tumor development.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Mutation; Smad2 Protein; Trans-Activators; Transforming Growth Factor beta

2001
Smads mediate signaling of the TGFbeta superfamily in normal keratinocytes but are lost during skin chemical carcinogenesis.
    Oncogene, 2001, Jan-25, Volume: 20, Issue:4

    The Smads are the signaling mediators of the TGFbeta superfamily. In the present study, we examined Smad expression in mouse epidermis and chemically-induced skin tumors. Mutations in Smad2 and -4 genes were also screened. Transcripts of Smad1 through -5 were constantly expressed in the epidermis regardless of changes in TGFbeta signaling, state of differentiation and stages of carcinogenesis. Smad7 transcripts were barely detectable in keratinocytes, but were induced by TGFbeta1 treatment and in chemically-induced skin tumors. At the protein level, Smad1 was detected throughout the epidermis, whereas Smad2 through -5 exhibited greater levels in suprabasal layers than basal keratinocytes. In cultured keratinocytes, Smad2, -3 and -4 underwent nuclear translocation upon TGFbeta1 treatment. Furthermore, nuclear translocation of Smads correlated with decreased BrdU labeling in proliferative keratinocytes. Although no mutations were detected in the Smad2 and -4 genes in tumors, proteins of Smad1 through -5 were partially or completely lost in carcinomas. These data document that Smads are expressed at high levels in the epidermis and mediate signaling of the TGFbeta superfamily. During skin carcinogenesis, loss of Smad1 through -5 and overexpression of Smad7 may contribute to the loss of growth inhibition mediated by TGFbeta superfamily members, thus resulting in tumor progression.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Carcinogens; Cell Nucleus; Cell Transformation, Neoplastic; Cells, Cultured; Keratinocytes; Mice; Mice, Inbred ICR; Protein Transport; RNA, Messenger; RNA, Neoplasm; Signal Transduction; Skin Neoplasms; Transcription Factors; Transforming Growth Factor beta

2001
Oncogenic ras represses transforming growth factor-beta /Smad signaling by degrading tumor suppressor Smad4.
    The Journal of biological chemistry, 2001, Aug-03, Volume: 276, Issue:31

    The loss of growth-inhibitory responses to transforming growth factor-beta (TGF-beta) is a frequent consequence of malignant transformation. Smad2, Smad3, and Smad4 proteins are important mediators of the antiproliferative responses to TGF-beta and may become inactivated in some human cancers. Epithelial cells harboring oncogenic Ras mutations often exhibit a loss of TGF-beta antiproliferative responses. To further investigate the effect of oncogenic Ras in TGF-beta signaling, we used an isopropyl-1-thio-beta-d-galactopyranoside-inducible expression system to express Ha-Ras(Val-12) in intestinal epithelial cells. Induction of Ha-Ras(Val-12) caused a decrease in the level of Smad4 expression, inhibited TGF-beta-induced complex formation between Smad2/Smad3 and Smad4, blocked Smad4 nuclear translocation, inhibited the TGF-beta-mediated decrease in [(3)H]thymidine incorporation, and repressed TGF-beta-activated transcriptional responses. The withdrawal of isopropyl-1-thio-beta-d-galactopyranoside or the addition of an inhibitor of the ubiquitin-proteasome pathway restored the Smad4 level and TGF-beta-induced Smad complex formation. Forced expression of Smad4 resulted in partial recovery of the TGF-beta-mediated growth inhibition and transcriptional responses in the presence of oncogenic Ras. Further, PD98059, a specific inhibitor of the MEK/ERK/mitogen-activated protein kinase pathway prevented the Ras-induced decrease in Smad4 expression and complex formation. Our results suggest a novel mechanism by which oncogenic Ras represses TGF-beta signaling by mitogen-activated protein kinase-dependent down-regulation of Smad4, thereby subverting the tumor suppressor function of TGF-beta.

    Topics: Animals; Cell Division; Cell Line, Transformed; Cell Nucleus; Cell Transformation, Neoplastic; DNA-Binding Proteins; Gene Expression Regulation; Genes, ras; Genes, Tumor Suppressor; Isopropyl Thiogalactoside; Kinetics; Mutation; Protein Transport; Signal Transduction; Thymidine; Trans-Activators; Transcription, Genetic; Transforming Growth Factor beta

2001
SMAD4 mutations in colorectal cancer probably occur before chromosomal instability, but after divergence of the microsatellite instability pathway.
    Proceedings of the National Academy of Sciences of the United States of America, 2001, Aug-14, Volume: 98, Issue:17

    Loss of chromosome 18q21 is well documented in colorectal cancer, and it has been suggested that this loss targets the DCC, DPC4/SMAD4, and SMAD2 genes. Recently, the importance of SMAD4, a downstream regulator in the TGF-beta signaling pathway, in colorectal cancer has been highlighted, although the frequency of SMAD4 mutations appears much lower than that of 18q21 loss. We set out to investigate allele loss, mutations, protein expression, and cytogenetics of chromosome 18 copy number in a collection of 44 colorectal cancer cell lines of known status with respect to microsatellite instability (MSI). Fourteen of thirty-two MSI(-) lines showed loss of SMAD4 protein expression; usually, one allele was lost and the other was mutated in one of a number of ways, including deletions of various sizes, splice site changes, and missense and nonsense point mutations (although no frameshifts). Of the 18 MSI(-) cancers with retained SMAD4 expression, four harbored missense mutations in the 3' part of the gene and showed allele loss. The remaining 14 MSI(-) lines had no detectable SMAD4 mutation, but all showed allele loss at SMAD4 and/or DCC. SMAD4 mutations can therefore account for about 50-60% of the 18q21 allele loss in colorectal cancer. No MSI(+) cancer showed loss of SMAD4 protein or SMAD4 mutation, and very few had allelic loss at SMAD4 or DCC, although many of these MSI(+) lines did carry TGFBIIR changes. Although SMAD4 mutations have been associated with late-stage or metastatic disease, our combined molecular and cytogenetic data best fit a model in which SMAD4 mutations occur before colorectal cancers become aneuploid/polyploid, but after the MSI(+) and MSI(-) pathways diverge. Thus, MSI(+) cancers may diverge first, followed by CIN(+) (chromosomal instability) cancers, leaving other cancers to follow a CIN(-)MSI(-) pathway.

    Topics: Adenocarcinoma; Blotting, Western; Cell Transformation, Neoplastic; Chromosome Deletion; Chromosomes, Human, Pair 18; Colorectal Neoplasms; DNA Mutational Analysis; DNA-Binding Proteins; DNA, Neoplasm; Gene Deletion; Gene Expression Profiling; Genes, DCC; Humans; Loss of Heterozygosity; Microsatellite Repeats; Mutation; Neoplasm Proteins; Ploidies; Polymorphism, Single-Stranded Conformational; Signal Transduction; Smad4 Protein; Time Factors; Trans-Activators; Transforming Growth Factor beta; Tumor Cells, Cultured

2001
ras oncogene expression determines sensitivity for intercellular induction of apoptosis.
    Carcinogenesis, 2001, Volume: 22, Issue:9

    Fibroblasts carrying an inducible ras oncogene acquire the transformed phenotype after oncogene induction. As a consequence, the transformed cells become sensitive to intercellular induction of apoptosis, a novel regulatory process directed by non-transformed fibroblasts against their transformed descendants. The causal relationship between oncogene expression and sensitivity to intercellular induction of apoptosis is based on extracellular superoxide anion production by oncogene-expressing cells. Superoxide anions (after dismutation to hydrogen peroxide) thereby foster HOCl synthesis and at the same time direct the selectivity of apoptosis induction through hydroxyl generation from HOCl. In parallel, ras expression enhances the sensitivity of fibroblasts for apoptosis-inducing stimuli like cycloheximide, ceramide and mitomycin C. This sensitization seems to be based on a decreased concentration of short lived endogenous apoptosis inhibitors. TGF-beta, like ras induction, decreases the concentration of endogenous apoptosis inhibitors, but does not induce the transformed phenotype. Therefore, TGF-beta treatment alone is not sufficient to render fibroblasts sensitive for intercellular induction of apoptosis, but TGF-beta treatment in parallel with ras activation enhances intercellular induction of apoptosis. Our findings demonstrate that Ras-mediated superoxide anion production determines sensitivity to intercellular induction of apoptosis, whereas the parallel decrease in endogenous apoptosis inhibitors modulates the kinetics of apoptosis induction.

    Topics: Animals; Apoptosis; Cattle; Cell Communication; Cell Transformation, Neoplastic; Cycloheximide; Fibroblasts; Gene Expression Regulation; Genes, ras; Humans; Mice; Mice, Inbred C3H; Protein Synthesis Inhibitors; Superoxides; Transforming Growth Factor beta; Transforming Growth Factor beta1

2001
A novel mechanism for regulating transforming growth factor beta (TGF-beta) signaling. Functional modulation of type III TGF-beta receptor expression through interaction with the PDZ domain protein, GIPC.
    The Journal of biological chemistry, 2001, Oct-26, Volume: 276, Issue:43

    Transforming growth factor beta (TGF-beta) mediates its biological effects through three high-affinity cell surface receptors, the TGF-beta type I, type II, and type III receptors, and the Smad family of transcription factors. Although the functions of the type II and type I receptors are well established, the precise role of the type III receptor in TGF-beta signaling remains to be established. While expression cloning signaling molecules downstream of TGF-beta, we cloned GIPC (GAIP-interacting protein, C terminus), a PDZ domain-containing protein. GIPC binds a Class I PDZ binding motif in the cytoplasmic domain of the type III receptor resulting in regulation of expression of the type III receptor at the cell surface. Increased expression of the type III receptor mediated by GIPC enhanced cellular responsiveness to TGF-beta both in terms of inhibition of proliferation and in plasminogen-activating inhibitor (PAI)-based promoter gene induction assays. In all cases, deletion of the Class I PDZ binding motif of the type III receptor prevented the type III receptor from binding to GIPC and abrogated the effects of GIPC on type III receptor expressing cells. These results establish, for the first time, a protein that interacts with the cytoplasmic domain of the type III receptor, determine that expression of the type III receptor is regulated at the protein level and that increased expression of the type III receptor is sufficient to enhance TGF-beta signaling. These results further support an essential, non-redundant role for the type III receptor in TGF-beta signaling.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Binding Sites; Carrier Proteins; Cell Transformation, Neoplastic; Cloning, Molecular; Cysteine Endopeptidases; Gene Expression Regulation; Gene Library; Mice; Multienzyme Complexes; Muscles; Neuropeptides; Plasminogen Activator Inhibitor 1; Proteasome Endopeptidase Complex; Protein Binding; Protein Structure, Tertiary; Proteoglycans; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Signal Transduction; Stem Cells; Tissue Distribution; Transcriptional Activation; Transforming Growth Factor beta; Two-Hybrid System Techniques

2001
Heterozygous mice for the transforming growth factor-beta type II receptor gene have increased susceptibility to hepatocellular carcinogenesis.
    Cancer research, 2001, Sep-15, Volume: 61, Issue:18

    The transforming growth factor-beta (TGF-beta) receptor complex and its downstream signaling intermediates constitute a tumor suppressor pathway. In many cancers, expression of TGF-beta type II receptor (TbetaR-II) is markedly decreased. In the present study, we show that the hepatocytes isolated from 15-day-old, but not 9-month-old, mice heterozygous for the deletion of the TbetaR-II gene are slightly less sensitive to the growth-inhibitory effect of TGF-beta when compared with wild-type littermates of same age. In addition, the proliferation index of hepatocytes as indicated by bromodeoxyuridine incorporation is mildly increased in the heterozygous mice. These subtle changes in cellular phenotype did not result in either gross or microscopic abnormality of the liver. The treatment of these mice with the chemical carcinogen, diethylnitrosamine, results in a significantly enhanced tumorigenesis in the liver when compared with the wild-type littermates. Our results demonstrate the gene-dosage effect of TbetaR-II and indicate that the reduced expression of TbetaR-II in mice increases susceptibility to tumorigenesis in the liver.

    Topics: Animals; Carcinogens; Cell Transformation, Neoplastic; Diethylnitrosamine; Female; Gene Dosage; Genes, cdc; Genetic Predisposition to Disease; Heterozygote; Liver; Liver Neoplasms, Experimental; Male; Mice; Mice, Inbred C57BL; Phenobarbital; Pregnancy; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Messenger; Transforming Growth Factor beta; Transforming Growth Factor beta1

2001
Extracellular signaling through the microenvironment: a hypothesis relating carcinogenesis, bystander effects, and genomic instability.
    Radiation research, 2001, Volume: 156, Issue:5 Pt 2

    Cell growth, differentiation and death are directed in large part by extracellular signaling through the interactions of cells with other cells and with the extracellular matrix; these interactions are in turn modulated by cytokines and growth factors, i.e. the microenvironment. Here we discuss the idea that extracellular signaling integrates multicellular damage responses that are important deterrents to the development of cancer through mechanisms that eliminate abnormal cells and inhibit neoplastic behavior. As an example, we discuss the action of transforming growth factor beta (TGFB1) as an extracellular sensor of damage. We propose that radiation-induced bystander effects and genomic instability are, respectively, positive and negative manifestations of this homeostatic process. Bystander effects exhibited predominantly after a low-dose or a nonhomogeneous radiation exposure are extracellular signaling pathways that modulate cellular repair and death programs. Persistent disruption of extracellular signaling after exposure to relatively high doses of ionizing radiation may lead to the accumulation of aberrant cells that are genomically unstable. Understanding radiation effects in terms of coordinated multicellular responses that affect decisions regarding the fate of a cell may necessitate re-evaluation of radiation dose and risk concepts and provide avenues for intervention.

    Topics: Bystander Effect; Cell Transformation, Neoplastic; Cells, Cultured; Extracellular Matrix Proteins; Genome; Humans; Neoplasm Proteins; Neoplasms, Radiation-Induced; Phenotype; Radiation; Signal Transduction; Transforming Growth Factor beta

2001
A novel human cancer culture model for the study of prostate cancer.
    Oncogene, 2001, Nov-29, Volume: 20, Issue:55

    Research into molecular and genetic mechanisms underlying prostate carcinogenesis would be greatly advanced by in vitro models of prostate tumors representing primary tumors. We have successfully established an immortalized human prostate epithelial (HPE) cell culture derived from a primary tumor with telomerase. The actively proliferating early passaged RC-58T cells were transduced through infection with a retrovirus vector expressing the human telomerase catalytic subunit (hTERT). A high level of telomerase was detected in RC-58T/hTERT cells but not RC-58T cells. RC-58T/hTERT cells are currently growing well at passage 50, whereas RC-58T cells senesced at passage 7. RC-58T/hTERT cells exhibit transformed morphology. More importantly, these immortalized cells showed anchorage-independent growth as they formed colonies in soft agar and grew above the agar layer. Expression of androgen-regulated prostate specific gene NKX3.1 and epithelial specific cytokeratin 8 (CK8) but not prostate specific antigen (PSA) and androgen receptor was detected in RC-58T/hTERT cells. Prostate stem cell antigen (PSCA) and p16 were also expressed in this cell line. RC-58T/hTERT cells showed growth inhibition when exposed to retinoic acid and transforming growth factor (TGF)-beta1 known potent inhibitors of prostate epithelial cell growth. A number of chromosome alterations were observed including the loss of chromosomes Y, 3p, 10p, 17p, 18q and the gain of chromosomes 16 and 20. These results demonstrate that this primary tumor-derived HPE cell line retained its transformed phenotypes and should allow studies to elucidate molecular and genetic alterations involved in prostate cancer. This is the first documented case of an established human prostate cancer cell line from a primary tumor of a prostate cancer patient with telomerase.

    Topics: Agar; Cell Culture Techniques; Cell Division; Cell Line, Transformed; Cell Size; Cell Transformation, Neoplastic; Chromosome Aberrations; DNA-Binding Proteins; Epithelial Cells; Humans; Karyotyping; Male; Models, Biological; Prostatic Neoplasms; Retroviridae; Reverse Transcriptase Polymerase Chain Reaction; RNA, Neoplasm; Telomerase; Transduction, Genetic; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tretinoin; Tumor Cells, Cultured

2001
The regulation and function of the Id proteins in lymphocyte development.
    Oncogene, 2001, Dec-20, Volume: 20, Issue:58

    Helix-loop-helix (HLH) proteins are essential factors for lymphocyte development and function. One class of HLH proteins, the E-proteins, regulate many aspects of lymphocyte maturation, survival, proliferation, and differentiation. E-proteins are negatively regulated by another class of HLH proteins known as the Id proteins. The Id proteins function as dominant negative inhibitors of E-proteins by inhibiting their ability to bind DNA. Here we discuss the function and regulation of the Id proteins in lymphocyte development.

    Topics: Animals; Cell Differentiation; Cell Division; Cell Lineage; Cell Transformation, Neoplastic; Clonal Deletion; DNA; DNA-Binding Proteins; Female; Gene Expression Regulation, Developmental; H-Y Antigen; Helix-Loop-Helix Motifs; Hematopoiesis; Humans; Inhibitor of Differentiation Protein 1; Inhibitor of Differentiation Protein 2; Inhibitor of Differentiation Proteins; Lymphocyte Activation; Lymphocyte Subsets; Lymphoma; Male; MAP Kinase Signaling System; Mice; Mice, Knockout; Mice, Transgenic; Models, Biological; Multigene Family; Neoplasm Proteins; Protein Kinases; Repressor Proteins; TCF Transcription Factors; Transcription Factor 7-Like 1 Protein; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta

2001
Expression of a decorin-like moleculein human melanoma.
    Pathology oncology research : POR, 2001, Volume: 7, Issue:4

    Decorin, a member of the family of small leucin-rich proteoglycans, has originally been described as a secreted proteoglycan component of the connective tissues, and has been implicated in the negative regulation of cell proliferation directly or via interactions with TGF-beta. It was reported to be generally absent from tumor cells. Here we show that human melanoma cell lines express a decorin-like molecule. We detected decorin mRNA by RT-PCR in 7 out 7 human melanoma lines characterized by various metastatic potential. Using polyclonal antiserum against the core protein of decorin, the typical 80-120 kD glycanated form as well as a high molecular weight aberrant version (200-210 kD) of decorin were demonstrated by Western blot technique in the culture supernatants as well as in lysates of human melanoma cells. Finally, decorin epitope was also demonstrated immunohistochemically in human melanoma xenografts, as well as in tumor cells of surgically resected melanomas but not in melanocytes of nevi. The expression of this aberrant decorin did not inhibit the in vitroor in vivogrowth of human melanoma cells, and it was independent of their metastatic potential. Human melanoma cell lines expressing aberrant decorin retained sensitivity to the antiproliferative and gelatinase-stimulatory effects of exogenous TGF-beta.

    Topics: Animals; Blotting, Southern; Cell Division; Cell Transformation, Neoplastic; Collagenases; Decorin; DNA Primers; Extracellular Matrix Proteins; Flow Cytometry; Immunoenzyme Techniques; Melanoma; Mice; Mice, SCID; Neoplasm Metastasis; Polymerase Chain Reaction; Proteoglycans; RNA, Messenger; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured

2001
CTGF modulates cell cycle progression in cAMP-arrested NRK fibroblasts.
    Journal of cellular physiology, 2000, Volume: 182, Issue:1

    Connective tissue growth factor (CTGF) is a 38-kDa cysteine-rich peptide, whose synthesis and secretion are selectively induced by transforming growth factor beta (TGF-beta) in connective tissue cells. Previous studies have demonstrated that CTGF functions as a downstream mediator of TGF-beta mitogenic activity, where it controls cell cycle progression through late G1 and S-phase entry of NRK fibroblast suspension cultures. Here we report that CTGF induces this S-phase entry by upregulating cyclin A levels. The molecular mechanism for cyclin A induction appears to be via reduction of p27(Kip1) levels, which results in hyperphosphorylation of pRb and release of E2F, a known modulator of cyclin A gene transcription. These data indicate that CTGF acts as a mediator of TGF-beta-induced fibroblast proliferation in suspension cultures by regulating cdk activities.

    Topics: 8-Bromo Cyclic Adenosine Monophosphate; Animals; Cell Cycle; Cell Cycle Proteins; Cell Line; Cell Transformation, Neoplastic; Connective Tissue Growth Factor; Cyclic AMP; Cyclin A; Cyclin-Dependent Kinase Inhibitor p27; Cyclin-Dependent Kinases; Fibroblasts; Growth Substances; Immediate-Early Proteins; Intercellular Signaling Peptides and Proteins; Kidney; Microtubule-Associated Proteins; Mitogens; Phosphorylation; Promoter Regions, Genetic; Rats; Retinoblastoma Protein; Transforming Growth Factor beta; Tumor Suppressor Proteins; Up-Regulation

2000
Inhibition of collagenase-3 (MMP-13) expression in transformed human keratinocytes by interferon-gamma is associated with activation of extracellular signal-regulated kinase-1,2 and STAT1.
    Oncogene, 2000, Jan-13, Volume: 19, Issue:2

    Collagenase-3 (MMP-13) is characterized by an exceptionally wide substrate specificity and restricted expression. MMP-13 is specifically expressed by transformed human keratinocytes in squamous cell carcinomas in vivo and its expression correlates with their invasion capacity. Here, we show, that interferon-gamma (IFN-gamma) markedly inhibits expression of MMP-13 by human cutaneous SCC cells (UT-SCC-7) and by ras-transformed human epidermal keratinocytes (A-5 cells) at the transcriptional level. In addition, IFN-gamma inhibits collagenase-1 (MMP-1) expression in these cells. IFN-gamma abolished the enhancement of MMP-13 and MMP-1 expression by transforming growth factor-beta (TGF-beta) and tumor necrosis factor-alpha (TNF-alpha), and inhibited invasion of A-5 cells through type I collagen. IFN-gamma also rapidly and transiently activates extracellular signal-regulated kinase 1,2 (ERK1,2) and blocking ERK1,2 pathway (Raf/MEK1,2/ERK1,2) by specific MEK1,2 inhibitor PD98059 partially (by 50%) prevents Ser-727 phosphorylation of STAT1 and suppression of MMP-13 expression by IFN-gamma. Furthermore, Ser-727 phosphorylation of STAT1 by ERK1,2, or independently of ERK1,2 activation is associated with marked reduction in MMP-13 expression. These observations identify a novel role for IFN-gamma as a potent inhibitor of collagenolytic activity and invasion of transformed squamous epithelial cells, and show that inhibition of MMP-13 expression by IFN-gamma involves activation of ERK1,2 and STAT1.

    Topics: Cell Line, Transformed; Cell Survival; Cell Transformation, Neoplastic; Collagenases; DNA-Binding Proteins; Enzyme Activation; Enzyme Inhibitors; Humans; Interferon-gamma; Keratinocytes; Matrix Metalloproteinase 13; Matrix Metalloproteinase Inhibitors; Matrix Metalloproteinases; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Organ Specificity; Recombinant Proteins; STAT1 Transcription Factor; Trans-Activators; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

2000
Role of TGF-beta in EGF-induced transformation of NRK cells is sustaining high-level EGF-signaling.
    FEBS letters, 2000, Jan-21, Volume: 466, Issue:1

    We have been isolating and analyzing NRK cell mutants, which fail to transform by epidermal growth factor (EGF) and transforming growth factor (TGF)-beta. One such mutant, R14, can respond to the growth inhibitory signal of TGF-beta to the same extent as parental NRK but fail to respond to the growth stimulatory signal of EGF. This mutant has a defect in EGF receptor (EGFR) expression. When R14 mutant expressed a high level of EGFR, however, EGF not only induced proliferation in this mutant but also induced transformation without the aid of TGF-beta. These findings suggest that the major role of TGF-beta in this transformation system should be to counteract the ligand-dependent down-regulation of EGFR, thereby sustaining high-level EGF-signaling.

    Topics: Animals; Base Sequence; Cell Line; Cell Transformation, Neoplastic; DNA Primers; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Molecular Sequence Data; Mutation; Promoter Regions, Genetic; Rats; Signal Transduction; Transforming Growth Factor beta

2000
TGF-beta isoforms are differentially expressed in increasing malignant grades of HaCaT keratinocytes, suggesting separate roles in skin carcinogenesis.
    The Journal of pathology, 2000, Volume: 190, Issue:5

    The three mammalian isoforms of transforming growth factor-beta (TGF-beta1, -beta2, and -beta3) are potent regulators of cell growth, differentiation, and extracellular matrix deposition. To study their role in skin carcinogenesis, normal human keratinocytes, early (31) and late (310) passage immortalized keratinocytes (HaCaT cells), and five HaCaT-ras clones exhibiting benign (A-5, I-7), malignant (II-4, A-5 RT1), and highly aggressive (A-5 RT3) tumourigenic phenotypes were examined for the expression of TGF-beta isoforms, by immunohistochemistry. This was performed under in vivo conditions, in surface transplants and subcutaneously growing tumours in nude mice. Generally, all tissues that formed keratinized epithelia demonstrated an immunostaining pattern similar to normal human skin. TGF-beta1 was localized to the upper differentiated layers, the stratum granulosum and corneum, in a perimembranous pattern, whereas TGF-beta2 and, weaker, TGF-beta3 immunostaining was present in all suprabasal layers of normal keratinizing epithelia. In contrast, non-keratinizing transplants of non-tumourigenic or highly aggressive cells showed little to no immunoreactivity for TGF-beta1. Whereas TGF-beta2 expression was moderate in the upper layers of non-tumourigenic epithelia, large tumour cells of the malignant HaCaT-ras clones, particularly at the invasion front, were strongly positive for TGF-beta2. TGF-beta3 immunostaining was most pronounced in the stroma of malignant tumours, implying its paracrine induction by the malignant tumour transplants. These results suggest differential functions for each TGF-beta isoform in epidermal carcinogenesis, such that TGF-beta1 is associated with the more differentiated state, TGF-beta2 with highly malignant and invading cells, and TGF-beta3 with tumour stroma formation and angiogenesis. Furthermore, the expression of TGF-betas by both early- and late-stage tumours implies that the isoforms may have distinct functions at different stages of malignancy, supporting their dual role in skin carcinogenesis.

    Topics: Animals; Cell Differentiation; Cell Line; Cell Transformation, Neoplastic; Humans; Immunoenzyme Techniques; Keratinocytes; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Transplantation; Protein Isoforms; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured

2000
Transforming growth factor beta2 promotes glucose carbon incorporation into nucleic acid ribose through the nonoxidative pentose cycle in lung epithelial carcinoma cells.
    Cancer research, 2000, Mar-01, Volume: 60, Issue:5

    The invasive transformation of A-459 lung epithelial carcinoma cells has been linked to the autocrine regulation of malignant phenotypic changes by transforming growth factor beta (TGF-beta). Here we demonstrate, using stable 13C glucose isotopes, that the transformed phenotype is characterized by decreased CO2 production via direct glucose oxidation but increased nucleic acid ribose synthesis through the nonoxidative reactions of the pentose cycle. Increased nucleic acid synthesis through the nonoxidative pentose cycle imparts the metabolic adaptation of nontransformed cells to the invasive phenotype that potentially explains the fundamental metabolic disturbance in tumor cells: highly increased nucleic acid synthesis despite hypoxia and decreased glucose oxidation.

    Topics: Adenocarcinoma; Cell Transformation, Neoplastic; Glucose; Humans; Lung Neoplasms; Oxidation-Reduction; Pentose Phosphate Pathway; Ribose; Transforming Growth Factor beta; Tumor Cells, Cultured

2000
Interleukin-2 gene-transduced human leukemic cells induce major histocompatibility complex-restricted and -unrestricted anti-leukemic effectors in mixed lymphocyte-tumor cultures.
    Cancer gene therapy, 2000, Volume: 7, Issue:2

    To explore the feasibility of designing vaccination protocols in acute leukemia patients with cytokine gene-transduced leukemic cells, we studied in vitro the growth potential of human leukemic cells transduced with the interleukin-2 (IL-2), IL-7, or IL-7 plus IL-2 genes, as well as the capacity of generating both autologous and allogeneic cytotoxic lymphocytes directed against the parental cells. A lymphoblastic T-cell line, ST4, obtained from a patient in long-lasting complete remission, was retrovirally engineered with the IL-2, IL-7, and IL-7 plus IL-2 genes; in addition, clones releasing different amounts of the cytokines were obtained by limiting dilution. Mixed lymphocyte-tumor cultures (MLTCs) were set up with parental or transduced leukemic cells as stimulators and with autologous or allogeneic lymphocytes as responders. When nonirradiated ST4 parental cells or clones producing <50 international units (IU)/mL/10(6) cells/72 hours of IL-2 were used as stimulators, leukemic overgrowth was observed in MLTCs within 16 days of culture. When clones producing >80 IU/mL/10(6) cells/72 hours of IL-2 were used as stimulators, the proliferation of leukemic cells was blocked and the transduced leukemic cells were completely cleared from the cultures by day 16; repeated restimulations with IL-2-producing leukemic cells were required to sustain long-term lymphocyte survival. On the contrary, when IL-7- or IL-7-IL-2-producing cells were used as stimulators, only a delay in leukemic cell overgrowth was observed, and lymphocytes were completely cleared from the cultures after day 60. IL-7 production by the different clones ranged between 11 and 36 ng/mL/10(6) cells/72 hours, whereas the highest IL-2-producing IL-7-IL-2 clone released 50 IU/mL/10(6) cells/72 hours of IL-2. When the stimulator efficacy of the highest IL-2-producing clone (ST4/IL-2#A7) was compared with that of exogenous IL-2 plus parental cells, a 7-fold higher amount of exogenous IL-2 was required to achieve the same results obtained with IL-2-producing leukemic cells. Autologous and allogeneic long-term MLTCs (up to 35 days) with ST4/IL-2#A7 as the stimulator were capable of generating cytotoxic effectors equally endowed with both major histocompatibility complex (MHC) class I-unrestricted and -restricted activity against parental ST4 cells. By day 18 of both autologous and allogeneic cultures, a substantial proportion of CD56+ cells was consistently recorded; this was coupled to a predominantly

    Topics: Cancer Vaccines; Cell Division; Cell Transformation, Neoplastic; Cytotoxicity, Immunologic; Gene Transfer Techniques; Genetic Therapy; Humans; Interleukin-2; K562 Cells; Leukemia, T-Cell; Lymphocyte Culture Test, Mixed; Major Histocompatibility Complex; Transforming Growth Factor beta; Tumor Cells, Cultured; Vaccines, DNA

2000
Differential cell cycle response of nontumorigenic and tumorigenic human papillomavirus-positive keratinocytes towards transforming growth factor-beta1.
    Journal of molecular medicine (Berlin, Germany), 2000, Volume: 78, Issue:2

    Human papillomaviruses (HPVs) are causative agents of a number of malignancies in humans, including cervical cancer. Their tumorigenic potential is linked to expression of the viral E6/E7 genes which can interfere with normal cell cycle control by targeting p53, p21WAF1, p27KIP1, and pRb. We show here that nontumorigenic and tumorigenic HPV-positive keratinocytes (HPK) exhibit striking differences in the response of cell cycle regulatory genes towards transforming growth factor beta-beta1. Treatment with this agent led to an efficient induction of p53 and the growth-inhibitory p15INK4 and p21WAF1 genes only in nontumorigenic HPKs and was linked to an efficient reduction in viral E6/E7 oncogene expression. This was associated with increased pRb levels, exhibiting sustained hypophosphorylation, and a permanent growth arrest in the G1 phase of the cell cycle. In contrast, tumorigenic HPKs exhibited only a modest rise in p53 protein levels and a substantially reduced induction of the p15INK4 and p21WAF1 genes, which was linked to a lesser degree of viral oncogene repression. In addition, tumorigenic HPKs rapidly resumed cell growth after a transient G1 arrest, concomitantly with the reappearance of hyperphosphorylated pRb. These results support the notion that the progression of HPV-positive cells to a malignant phenotype is associated with increased resistance to growth inhibition by transforming growth factor-beta1. This is linked in the tumorigenic cells to a lack of persistent G1 arrest, inefficient induction of several cell cycle control genes involved in growth inhibition, and inefficient repression of the growth-promoting viral E6/E7 oncogenes.

    Topics: Blotting, Northern; Blotting, Western; Cell Cycle; Cell Line, Transformed; Cell Transformation, Neoplastic; Cell Transformation, Viral; Gene Expression Regulation; Humans; Keratinocytes; Papillomaviridae; Transforming Growth Factor beta

2000
Role of the IkappaB kinase complex in oncogenic Ras- and Raf-mediated transformation of rat liver epithelial cells.
    Molecular and cellular biology, 2000, Volume: 20, Issue:15

    NF-kappaB/Rel factors have been implicated in the regulation of liver cell death during development, after partial hepatectomy, and in hepatocytes in culture. Rat liver epithelial cells (RLEs) display many biochemical and ultrastructural characteristics of oval cells, which are multipotent cells that can differentiate into mature hepatocytes. While untransformed RLEs undergo growth arrest and apoptosis in response to transforming growth factor beta1 (TGF-beta1) treatment, oncogenic Ras- or Raf-transformed RLEs are insensitive to TGF-beta1-mediated growth arrest. Here we have tested the hypothesis that Ras- or Raf-transformed RLEs have altered NF-kappaB regulation, leading to this resistance to TGF-beta1. We show that classical NF-kappaB is aberrantly activated in Ras- or Raf-transformed RLEs, due to increased phosphorylation and degradation of IkappaB-alpha protein. Inhibition of NF-kappaB activity with a dominant negative form of IkappaB-alpha restored TGF-beta1-mediated cell killing of transformed RLEs. IKK activity mediates this hyperphosphorylation of IkappaB-alpha protein. As judged by kinase assays and transfection of dominant negative IKK-1 and IKK-2 expression vectors, NF-kappaB activation by Ras appeared to be mediated by both IKK-1 and IKK-2, while Raf-induced NF-kappaB activation was mediated by IKK-2. NF-kappaB activation in the Ras-transformed cells was mediated by both the Raf and phosphatidylinositol 3-kinase pathways, while in the Raf-transformed cells, NF-kappaB induction was mediated by the mitogen-activated protein kinase cascade. Last, inhibition of either IKK-1 or IKK-2 reduced focus-forming activity in Ras-transformed RLEs. Overall, these studies elucidate a mechanism that contributes to the process of transformation of liver cells by oncogene Ras and Raf through the IkappaB kinase complex leading to constitutive activation of NF-kappaB.

    Topics: Animals; Apoptosis; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Epithelium; Genes, ras; I-kappa B Proteins; Liver; NF-kappa B; Oncogene Proteins v-raf; Phosphatidylinositol 3-Kinases; Phosphorylation; Rats; Retroviridae Proteins, Oncogenic; Transforming Growth Factor beta

2000
Endocervical cancer is associated with an increase in the ligands and receptors for transforming growth factor-beta and a contrasting decrease in p27(Kip1).
    Gynecologic oncology, 2000, Volume: 78, Issue:2

    The aim of this study was to investigate the relationship between the expression of the TGF-beta ligands and TGF-beta receptors to the expression of p27(Kip1), a TGF-beta-regulated gene, in endocervical cancer.. To examine the expression of TGF-beta and p27(Kip1) in malignant transformation of the uterine endocervix, a panel of 23 formalin-fixed and paraffin-embedded human cervical specimens, including 8 with benign endocervical glands, 8 with cervical adenocarcinoma in situ, and 7 with cervical adenocarcinomas, was used. Tissues were immunostained with polyclonal antibodies that react specifically with TGF-beta 1, TGF-beta 2, TGF-beta 3, TGF-beta RI, TGF-beta RII, and p27(Kip1).. Immunostaining for TGF-beta 1, TGF-beta 2, TGF-beta 3, TGF-beta RI, TGF-beta RII, and p27(Kip1) was detected in normal endocervix, with the TGF-betas showing weak cytoplasmic staining, while p27(Kip1) showed strong nuclear staining. Expression of TGF-beta increased significantly upon neoplastic transformation with the TGF-beta ligands and receptors showing strong cytoplasmic staining in adenocarcinoma in situ compared to normal endocervix. Interestingly, expression of TGF-beta was lower in adenocarcinoma than in adenocarcinoma in situ, but still significantly higher than in normal endocervix. TGF-beta 2 and TGF-beta 3 showed higher levels of immunostaining than TGF-beta 1 in adenocarcinomas. In contrast, p27(Kip1) protein expression decreased with progressive malignancy, with lower p27(Kip1) protein levels detected in adenocarcinoma than in adenocarcinoma in situ, while normal endocervix showed the highest level of p27(Kip1) protein expression.. Elevated expression of the TGF-beta ligands and receptors is found in both cervical adenocarcinoma in situ and adenocarcinoma compared to normal endocervix. In contrast, a progressive decrease in p27(Kip1) occurs upon neoplastic transformation of the normal endocervix to cervical adenocarcinoma. These results suggest that neoplastic transformation of the endocervix may be related to dysregulation of TGF-beta and p27(Kip1) seen as an elevation of TGF-beta and a reduction of p27(Kip1) expression that may lead to loss of cell cycle control.

    Topics: Activin Receptors, Type I; Adenocarcinoma; Carcinoma in Situ; Cell Cycle Proteins; Cell Transformation, Neoplastic; Cervix Uteri; Cyclin-Dependent Kinase Inhibitor p27; Eosine Yellowish-(YS); Female; Genes, Tumor Suppressor; Hematoxylin; Humans; Immunohistochemistry; Ligands; Microtubule-Associated Proteins; Neoplasm Staging; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Staining and Labeling; Transforming Growth Factor beta; Tumor Suppressor Proteins; Uterine Cervical Neoplasms

2000
Transforming growth factor beta1 selectively regulates ferritin gene expression in malignant H-ras-transformed fibrosarcoma cell lines.
    Biochemistry and cell biology = Biochimie et biologie cellulaire, 2000, Volume: 78, Issue:4

    Transforming growth factor beta1 is an important growth regulator in many cell types, usually exerting a negative effect on cellular growth. Inhibition of DNA synthesis and cell proliferation is frequently lost during malignant transformation, and in some cases, tumor cell proliferation is actually stimulated by TGF-beta1. The present study demonstrates a novel link between alterations in TGF-beta1 regulation during malignant conversion, and the expression of ferritin, an important activity involved in a number of biological functions including iron homeostasis and cell-growth control. A series of H-ras-transformed mouse 10 T 1/2 cell lines, exhibiting increasing malignant potential, was investigated for possible TGF-beta1-mediated changes in ferritin gene expression. Selective induction of gene expression was observed, since only H-ras-transformed cells with malignant potential exhibited marked elevations in ferritin gene expression, in particular, alterations in H-ferritin gene expression. The regulation of H-ferritin gene expression in response to TGF-beta1 did not involve alterations in transcription, but occurred through mechanisms of post-transcriptional stabilization of the H-ferritin mRNA. Additionally, evidence was obtained for a cycloheximide-sensitive regulator of H-ferritin gene expression, since the presence of this protein synthesis inhibitor increased H-ferritin message levels, and in combination with TGF-beta1, cooperated in an additive manner to augment H-ferritin gene expression. These results show for the first time that TGF-beta1 can regulate ferritin gene expression in malignant H-ras transformed cells, and suggest a mechanism for growth factor stimulation of malignant cells, in which early alterations in the control of H-ferritin gene expression are important.

    Topics: Animals; Blotting, Northern; Cell Line, Transformed; Cell Transformation, Neoplastic; Culture Media, Serum-Free; Cycloheximide; Dactinomycin; Ferritins; Gene Expression Regulation; Genes, ras; Mice; Nucleic Acid Synthesis Inhibitors; Protein Synthesis Inhibitors; Ribonucleotide Reductases; RNA, Messenger; Transfection; Transforming Growth Factor beta; Transforming Growth Factor beta1

2000
Mechanisms of cell transformation in the Syrian hamster embryo (SHE) cell transformation system.
    Annals of the New York Academy of Sciences, 2000, Volume: 919

    The Syrian hamster embryo (SHE) cell transformation system has been used for investigational studies of basic mechanisms of neoplastic transformation, as well as determining the carcinogenic potential of chemical, physical, and biological agents. Many of these investigations utilize an intermediate step in the SHE cell neoplastic transformation process, known as morphological transformation, as an indicator that the cells have acquired an increased potential to progress to malignancy. While the nature of the morphologically transformed phenotype is not completely understood, it is believed to result from a block in the cellular differentiation of stem cells present within the SHE cell population. In terms of determination of the transforming potential of biological/chemical/physical agents, more than 500 agents have been tested in the SHE cell transformation assay with an 80-90% correlation between MT and carcinogenic potential. As such, the SHE cell transformation assay has utility as a test to provide short-term information on the carcinogenic potential of chemicals. One class of agents of current interest with regard to SHE cell transformation assay utilization consists of growth and differentiation factors (GDFs). Analysis of the SHE cell transformation potential of the GDFs, epidermal growth factor (EGF), fibroblast growth factor 4 (FGF-4), platelet-derived growth factor AA (PDGF AA), PDGF AB, PDGF BB, and the antimitogenic GDF, transforming growth factor beta one (TGF-beta1), was performed. All GDFs, with the exception of TGF-beta1, induced SHE cell transformation. However, an interesting difference between the GDFs was observed--PDGF A/B and PDGF B/B, but not PDGF A/A, EGF, or FGF-4, induced transformation after both a transient 1-day exposure and a continuous 7-day exposure, while continuous 7-day exposure was required for transformation by PDGF A/A, EGF, and FGF-4. Interestingly, both transient 1-day and continuous 7-day TGF-beta1 exposure resulted in suppression of transformation induced by a variety of transforming agents including growth factors, Ames assay-positive carcinogens, Ames assay-negative carcinogens, and spontaneous transformation. Interestingly TGF-beta1 was not able to suppress transformation by the tumor promoter, TPA. Together, these data demonstrate the utility of the Syrian hamster embryo cell transformation system for analyzing the transforming potential of GDFs and for characterizing differences in transforming mechanisms be

    Topics: Animals; Carcinogens; Cell Division; Cell Line, Transformed; Cell Size; Cell Transformation, Neoplastic; Cricetinae; Growth Inhibitors; Growth Substances; Mesocricetus; Mitogens; Phenotype; Transforming Growth Factor beta; Transforming Growth Factor beta1

2000
Inhibition of farnesyltransferase increases TGFbeta type II receptor expression and enhances the responsiveness of human cancer cells to TGFbeta.
    Oncogene, 2000, Nov-16, Volume: 19, Issue:48

    Several small GTPases of the Ras superfamily have been shown to antagonize TGFbeta signaling in human tumor cell lines. Some of these GTPases are post-translationally modified by farnesylation, a lipid modification catalyzed by farnesyltransferase and required for the proteins to attach to membranes and to function. In this study, we investigated the effect of the farnesyltransferase inhibitor FTI-277 on TGFbeta-regulated cell growth and transcription. Treatment of the human pancreatic tumor cell line, Panc-1, with FTI-277 enhanced the ability of TGFbeta to inhibit both anchorage-dependent and -independent tumor cell growth. FTI-277 also enhanced the ability of TGFbeta to induce transcription, as measured by p3TP-lux reporter activity and collagen synthesis. The enhancement of TGFbeta responses by FTI-277 correlated with the stimulation of transcription and protein expression of type II TGFbeta receptor (TbetaRII). Consequently, FTI-277-treated cells exhibited a higher level of TGFbeta binding to its receptor. Thus, inhibition of protein farnesylation stimulates TbetaRII expression, which leads to increased TGFbeta receptor binding and signaling as well as inhibition of tumor cell growth and transformation.

    Topics: 3T3 Cells; Alkyl and Aryl Transferases; Animals; Cell Division; Cell Transformation, Neoplastic; Drug Synergism; Enzyme Inhibitors; Farnesyltranstransferase; Humans; Methionine; Mice; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Transcription, Genetic; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured

2000
The transcription factor EGR-1 suppresses transformation of human fibrosarcoma HT1080 cells by coordinated induction of transforming growth factor-beta1, fibronectin, and plasminogen activator inhibitor-1.
    The Journal of biological chemistry, 1999, Feb-12, Volume: 274, Issue:7

    Re-expression of EGR-1 in fibrosarcoma HT1080 suppresses transformation including tumorigenicity (Huang, R.-P., Liu, C., Fan, Y., Mercola, D., and Adamson, E. (1995) Cancer Res. 55, 5054-5062) owing in part to up-regulation of the transforming growth factor (TGF)-beta1 promoter by EGR-1 which suppresses growth by an autocrine mechanism (Liu, C., Adamson, E., and Mercola, D. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 11831-11836). Here we show that enhanced cell attachment contributes to the suppression via increased secretion of fibronectin (FN) and also of plasminogen activator inhibitor-1 (PAI-1). The secretion of FN and PAI-1 is strongly correlated with EGR-1 expression (RPEARSON = 0.971 and 0. 985, respectively). Addition of authentic TGF-beta1 to parental cells greatly stimulated secretion of PAI-1 but not FN, whereas addition of TGF-beta antibody or lipofection with specific antisense TGF-beta1 oligonucleotides to EGR-1-regulated cells completely inhibits the secretion of PAI-1 but not FN. However, in gel mobility shift assays pure EGR-1 or nuclear extracts of EGR-1-regulated cells specifically bind to two GC-rich elements of the human FN promoter at positions -75/-52 and -4/+18, indicating that the increased secretion of FN is likely due to direct up-regulation by EGR-1. Moreover, adhesion was greatly enhanced in EGR-1-regulated cells and was reversed by treatment with Arg-Gly-Asp (RGD) or PAI-1 antibody indicating that the secreted proteins are functional. We conclude that EGR-1 regulates the coordinated expression of gene products important for cell attachment ("oikis" factor) and normal growth control.

    Topics: Cell Adhesion; Cell Transformation, Neoplastic; DNA-Binding Proteins; Early Growth Response Protein 1; Fibronectins; Fibrosarcoma; Gene Expression Regulation, Neoplastic; Humans; Immediate-Early Proteins; Oligodeoxyribonucleotides, Antisense; Oligopeptides; Plasminogen Activator Inhibitor 1; Promoter Regions, Genetic; Transcription Factors; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured; Zinc Fingers

1999
A malignant transformation of human cells by 2,3,7,8-tetrachlorodibenzo-p-dioxin exhibits altered expressions of growth regulatory factors.
    Carcinogenesis, 1999, Volume: 20, Issue:1

    The neoplastic transformation of human cells in culture with exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has recently been reported. In this study, expressions of growth regulatory factors were analyzed to examine their possible roles in TCDD-induced malignant transformation of human cells. Reverse transcription-polymerase chain reaction (RT-PCR) and immunoblot analysis were performed to detect altered expressions of genes associated with dioxin responses. The RT-PCR analysis showed that expressions of the growth regulatory factors, such as transforming growth factor-beta1 (TGF-beta1), plasminogen activator inhibitor-2 (PAI-2) and tumor necrosis factor-alpha (TNF-alpha), were significantly changed in the transformed cells as compared with the parental cells. Whereas parental cells showed a dose-dependent increase of PAI-2 mRNA levels following TCDD exposure, the transformed cells did not show any significant induction. In addition, constitutive levels of PAI-2 mRNA were 25 times lower in the transformed cells than in the parental cells. The mRNA stability assay suggests that downregulation of PAI-2 mRNA in the transformed cells may be associated with the posttranscriptional control. Expression of TGF-beta1 mRNA in the transformed cells was also four times lower than the parental cells. However, levels of TNF-alpha mRNA in the transformed cells were increased 3-fold. These results suggest that dysregulation of growth regulatory factors may be involved in TCDD-induced cellular transformation. Whereas plenty of studies demonstrated a number of immediate toxic effects by TCDD, this study revealed an initial evidence that altered expression of growth regulatory genes, such as PAI-2, TGF-beta1 or TNF-alpha, are some of the genetic events fixed in the genome following the successive cell divisions of TCDD-damaged cells. It is suggested that these changes may be associated with TCDD-induced malignant transformation of human cells.

    Topics: Aryl Hydrocarbon Receptor Nuclear Translocator; Cell Transformation, Neoplastic; Cells, Cultured; Clone Cells; Cytochrome P-450 CYP1A1; DNA-Binding Proteins; Gene Expression Regulation; Humans; Keratinocytes; Plasminogen Activator Inhibitor 2; Polychlorinated Dibenzodioxins; Receptors, Aryl Hydrocarbon; RNA, Messenger; Transcription Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

1999
Overexpression of transforming growth factor beta-type II receptor reduces tumorigenicity and metastastic potential of K-ras-transformed thyroid cells.
    International journal of cancer, 1999, Jan-05, Volume: 80, Issue:1

    Expression of type II receptor of transforming growth factor beta (TbetaRII) is necessary for this factor to inhibit the growth of thyroid epithelial cells. In rat thyroid transformed cells, the resistance to transforming growth factor beta (TGFbeta) is associated with a decreased expression of TbetaRII mRNA and protein. Reduced TbetaRII expression has also been found in human thyroid differentiated and undifferentiated carcinomas. To investigate the role of TbetaRII in modulating the tumorigenic potential of k-ras-transformed thyroid cells, we transfected these cells with an expression vector carrying the human TbetaRII gene, regulated by an inducible promoter. Isolated clones, overexpressing TbetaRII, showed a reduction in the anchorage-dependent and -independent cell growth, compared with control k-ras-transformed cells. When transplanted in athymic nude mice, the transfected clones presented a decrease in tumorigenicity with respect to the highly malignant parental cells. Moreover, the diminished tumorigenic ability of the clones studied was accompanied by a statistically significant reduction in spontaneous and lung artificial metastases. Taken together, our data demonstrate that TbetaRII acts as a potent tumor suppressor gene when overexpressed in malignant thyroid cells.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Collagen; Gene Expression Regulation; Genes, ras; Humans; Kinetics; Mice; Mice, Nude; Neoplasm Metastasis; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; Rats; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Recombinant Proteins; Thyroid Neoplasms; Time Factors; Transfection; Transforming Growth Factor beta

1999
Transformation of intestinal epithelial cells by chronic TGF-beta1 treatment results in downregulation of the type II TGF-beta receptor and induction of cyclooxygenase-2.
    Oncogene, 1999, Jan-28, Volume: 18, Issue:4

    The precise role of TGF-beta in colorectal carcinogenesis is not clear. The purpose of this study was to determine the phenotypic alterations caused by chronic exposure to TGF-beta in non-transformed intestinal epithelial (RIE-1) cells. Growth of RIE-1 cells was inhibited by >75% following TGF-beta1 treatment for 7 days, after which the cells resumed a normal growth despite the presence of TGF-beta1. These 'TGF-beta-resistant' cells (RIE-Tr) were continuously exposed to TGF-beta for >50 days. Unlike the parental RIE cells, RIE-Tr cells lost contact inhibition, formed foci in culture, grew in soft agarose. RIE-Tr cells demonstrated TGF-beta-dependent invasive potential in an in vitro assay and were resistant to Matrigel and Na-butyrate-induced apoptosis. The RIE-Tr cells were also tumorigenic in nude mice. The transformed phenotype of RIE-Tr cells was associated with a 95% decrease in the level of the type II TGF-beta receptor (TbetaRII) protein, a 40-fold increase in cyclooxygenase-2 (COX-2) protein, and 5.9-fold increase in the production of prostacyclin. Most RIE-Tr subclones that expressed low levels of TbetaRII and high levels of COX-2 were tumorigenic. Those subclones that express abundant TbetaRII and low levels of COX-2 were not tumorigenic in nude mice. A selective COX-2 inhibitor inhibited RIE-Tr cell growth in culture and tumor growth in nude mice. The reduced expression of TbetaRII, increased expression of COX-2, and the ability to form colonies in Matrigel were all reversible upon withdrawal of exogenous TGF-beta1 for the RIE-Tr cells.

    Topics: Animals; Apoptosis; Cell Count; Cell Division; Cell Transformation, Neoplastic; Cyclooxygenase 2; Down-Regulation; Drug Resistance; Enzyme Induction; Epithelial Cells; Intestines; Isoenzymes; Phenotype; Prostaglandin-Endoperoxide Synthases; Protein Serine-Threonine Kinases; Rats; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

1999
Cyclosporine induces cancer progression by a cell-autonomous mechanism.
    Nature, 1999, Feb-11, Volume: 397, Issue:6719

    Malignancy is a common and dreaded complication following organ transplantation. The high incidence of neoplasm and its aggressive progression, which are associated with immunosuppressive therapy, are thought to be due to the resulting impairment of the organ recipient's immune-surveillance system. Here we report a mechanism for the heightened malignancy that is independent of host immunity. We show that cyclosporine (cyclosporin A), an immunosuppressant that has had a major impact on improving patient outcome following organ transplantation, induces phenotypic changes, including invasiveness of non-transformed cells, by a cell-autonomous mechanism. Our studies show that cyclosporine treatment of adenocarcinoma cells results in striking morphological alterations, including membrane ruffling and numerous pseudopodial protrusions, increased cell motility, and anchorage-independent (invasive) growth. These changes are prevented by treatment with monoclonal antibodies directed at transforming growth factor-beta (TGF-beta). In vivo, cyclosporine enhances tumour growth in immunodeficient SCID-beige mice; anti-TGF-beta monoclonal antibodies but not control antibodies prevent the cyclosporine-induced increase in the number of metastases. Our findings suggest that immunosuppressants like cyclosporine can promote cancer progression by a direct cellular effect that is independent of its effect on the host's immune cells, and that cyclosporine-induced TGF-beta production is involved in this.

    Topics: Animals; Antibodies; Carcinogens; Carcinoma, Renal Cell; Cell Adhesion; Cell Division; Cell Movement; Cell Size; Cell Transformation, Neoplastic; Cyclosporine; Humans; Immunosuppressive Agents; Male; Mice; Mice, SCID; Microscopy, Electron, Scanning; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasm Transplantation; Phenotype; Pseudopodia; Transforming Growth Factor beta; Tumor Cells, Cultured

1999
Desensitization of melanoma cells to autocrine TGF-beta isoforms.
    Journal of cellular physiology, 1999, Volume: 178, Issue:2

    Previous studies have suggested that transforming growth factor-beta 1 (TGF-beta1) acts as an autocrine growth inhibitor on normal human melanocytes, while melanoma cells may not respond to this stimulus. The role of other TGF-beta isoforms such as TGF-beta2 and TGF-beta3 remained less well characterized. In the present study, the mRNA and protein levels of all three isoforms of TGF-beta were analyzed in a panel of human melanoma cell lines and in cultures of normal human melanocytes in vitro. Northern analysis showed that the degree of TGF-beta1, -beta2, -beta3 mRNA expression varied considerably in melanoma cells, whereas TGF-beta expression was very low in melanocytes. In melanoma cells, secreted amounts of TGF-beta1 and TGF-beta3 were found increased in comparison to normal melanocytes: 615 pg/ml vs. 118 pg/ml and 193 pg/ml vs. 30 pg/ml (mean values). In addition, low levels of TGF-beta2 were detected (mean value: 28 pg/ml). Although TGF-beta secretion increased, the proliferation of melanoma cells was found to be only moderately inhibited by TGF-beta isoforms, in contrast to its strong antiproliferative effect on normal human melanocytes: - 15%, -11%, and -18% vs. -52%, -46%, and -50% average inhibition at 0.5 ng/ml TGF-beta1, -beta2, and -beta3, respectively. The different efficacy of TGF-beta on melanocyte and melanoma cells was highly significant (P<0.0001); in addition, TGF-beta-dependent growth inhibition of melanoma cells from primary tumors vs. cells from metastases showed a trend for further decreased response for the metastatic populations (P< or = 0.075). Measurements of DNA synthesis revealed even more pronounced differences between melanocytes (-86%, -78%, and -80% inhibition, respectively, for TGF-beta1, -beta2, and -beta3) and melanoma cells (no inhibition). Our data show loss of responsiveness of melanoma cells to the growth-inhibitory function of TGF-beta isoforms but not of melanocytes. Although melanoma cells are not growth-inhibited by all three TGF-beta isoforms, they secrete significantly higher levels of TGF-beta, as compared to melanocytes. The reduced response indicates their escape from TGF-beta surveillance with ongoing tumor progression.

    Topics: Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; DNA; DNA, Neoplasm; Gene Expression; Growth Inhibitors; Humans; Melanocytes; Melanoma; Neoplasm Metastasis; Recombinant Proteins; RNA, Messenger; RNA, Neoplasm; 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 actin organisation by TGF-beta in H-ras-transformed fibroblasts.
    Journal of cell science, 1999, Volume: 112 ( Pt 8)

    The actin cytoskeleton undergoes architectural changes during the processes of cell transformation and tumourigenesis. Transforming growth factors beta arrest cell cycle progression, regulate differentiation and modulate the onset of oncogenesis and tumourigenesis. Here, we investigated the direct role of transforming growth factor beta-1 in altering the transformed phenotype and regulating the actin organisation of oncogenic fibroblasts that constitutively or inducibly express the H-ras oncogene. Following transforming growth factor beta-1 treatment, these transformed fibroblasts undergo a dramatic morphological alteration that includes a discrete reorganisation of their actin cytoskeleton and focal adhesions. Quantitative biochemical analysis demonstrated that transforming growth factor beta-1 potently induced polymerisation of globular to filamentous actin, thus corroborating the morphological analysis. The effect of transforming growth factor beta-1 on the cytoskeleton correlates with the ability of this cytokine to suppress anchorage-independent growth of the transformed fibroblasts. Furthermore, transforming growth factor beta-1 upregulates considerably the levels of the RhoB small GTPase and less the RhoA levels. Finally, The beta GTPase inhibitor, C3 exotransferase, blocks the ability of TGF-beta1 to induce cytoskeletal reorganisation. These findings indicate that transforming growth factor beta can regulate cell morphology and growth in a concerted manner possibly via mechanisms that control the actin cytoskeleton.

    Topics: 3T3 Cells; Actins; Animals; Blotting, Western; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Cytoskeleton; Dexamethasone; Fibroblasts; Fluorescent Antibody Technique, Indirect; GTP Phosphohydrolases; Mammary Tumor Virus, Mouse; Mice; Microscopy, Confocal; ras Proteins; Rats; Transforming Growth Factor beta; Up-Regulation

1999
Restoration of transforming growth factor beta signaling pathway in human prostate cancer cells suppresses tumorigenicity via induction of caspase-1-mediated apoptosis.
    Cancer research, 1999, Mar-15, Volume: 59, Issue:6

    Previous studies (Y. Guo and N. Kyprianou, Cell Growth Diff., 9: 185-193, 1998) have demonstrated that overexpression of transforming growth factor (TGF) beta type II receptor (TbetaRII) gene in human prostate cancer cells LNCaP, which are refractory to TGF-beta1 and lack TbetaRII receptor expression, can restore TGF-beta1 sensitivity and suppress in vitro tumorigenic growth by inhibiting cell proliferation. In the present study, we investigated the effect of TbetaRII receptor overexpression in LNCaP cells on apoptosis induction and tumorigenicity. The ability of LNCaP cells that overexpress TbetaRII to undergo apoptosis in response to TGF-beta1 was examined by DNA fragmentation and terminal transferase-mediated dUTP-biotin end labeling analysis. To explore the potential apoptotic nature of TGF-beta1-mediated antitumor effect against human prostate cancer cells, the expression of apoptotic proteins bcl-2 and bax was examined by Western blot analyses. The significance of caspase 1 in TGF-beta1-mediated apoptosis was also determined by examining the expression and activation of caspase 1 by reverse transcription-PCR and Western blot analyses, respectively. Comparative analysis of tumorigenicity of the parental LNCaP and TbetaRII-overexpressing clones in severely combined immunodeficient mice revealed a significant suppression of tumor growth in TbetaRII transfectant clones compared with parental LNCaP cells and neomycin-control clones (P < 0.05). A significantly higher incidence of endogenous apoptosis was observed in TbetaRII clone-61-derived tumor compared with the parental LNCaP tumors. This induction of apoptosis in the LNCaP tumors with restored TGF-beta1 signaling was associated with decreased bcl-2 expression, increased bax, and caspase-1 immunoreactivty. Moreover, an increased expression of the cyclin-dependent kinase inhibitor p27Kip1 was detected in TbetaRII-overexpressing tumors compared with the parental tumors. LNCaP TbetaRII transfectant cells exhibited a marked induction of apoptosis, paralleled with a decreased bcl-2 expression in response to TGF-beta1 treatment in vitro. This TGF-beta1-mediated apoptosis induction in TbetaRII transfectant cells was significantly protected by the caspase-1 inhibitor (zVAD-fmk) in a dose-dependent manner. Furthermore, a significant temporal induction of caspase-1 mRNA and protein expression was detected in TbetaRII cells in response to TGF-beta1 treatment. Our findings suggest that restoration of TGF-beta1 si

    Topics: Animals; Apoptosis; Caspase 1; Cell Transformation, Neoplastic; DNA Fragmentation; Enzyme Induction; Humans; Male; Mice; Mice, SCID; Neoplasm Transplantation; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

1999
A mechanism of repression of TGFbeta/ Smad signaling by oncogenic Ras.
    Genes & development, 1999, Apr-01, Volume: 13, Issue:7

    TGFbeta can override the proliferative effects of EGF and other Ras-activating mitogens in normal epithelial cells. However, epithelial cells harboring oncogenic Ras mutations often show a loss of TGFbeta antimitogenic responses. Here we report that oncogenic Ras inhibits TGFbeta signaling in mammary and lung epithelial cells by negatively regulating the TGFbeta mediators Smad2 and Smad3. Oncogenically activated Ras inhibits the TGFbeta-induced nuclear accumulation of Smad2 and Smad3 and Smad-dependent transcription. Ras acting via Erk MAP kinases causes phosphorylation of Smad2 and Smad3 at specific sites in the region linking the DNA-binding domain and the transcriptional activation domain. These sites are separate from the TGFbeta receptor phosphorylation sites that activate Smad nuclear translocation. Mutation of these MAP kinase sites in Smad3 yields a Ras-resistant form that can rescue the growth inhibitory response to TGFbeta in Ras-transformed cells. EGF, which is weaker than oncogenic mutations at activating Ras, induces a less extensive phosphorylation and cytoplasmic retention of Smad2 and Smad3. Our results suggest a mechanism for the counterbalanced regulation of Smad2/Smad3 by TGFbeta and Ras signals in normal cells, and for the silencing of antimitogenic TGFbeta functions by hyperactive Ras in cancer cells.

    Topics: Animals; Cell Line; Cell Nucleus; Cell Transformation, Neoplastic; Colonic Neoplasms; COS Cells; DNA-Binding Proteins; Dose-Response Relationship, Drug; Humans; Luciferases; MAP Kinase Kinase 1; Mitogen-Activated Protein Kinase Kinases; Models, Genetic; Phosphorylation; Protein Serine-Threonine Kinases; Protein-Tyrosine Kinases; ras Proteins; Signal Transduction; Smad2 Protein; Smad3 Protein; Time Factors; Trans-Activators; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

1999
Transforming growth factor beta1 suppresses nonmetastatic colon cancer at an early stage of tumorigenesis.
    Cancer research, 1999, Jul-15, Volume: 59, Issue:14

    The transforming growth factor beta (TGF-beta) pathway is known to play an important role in both human and urine colon cancer. However, the staging, ligand specificity, and mechanism underlying the tumor suppressive activity of this pathway are unknown. We developed a mouse model for colon cancer that identifies an early role for TGF-beta1 in tumor suppression and implicates TGF-beta2 or TGF-beta3 in the prevention of metastasis. Analysis of the development of colon cancer in TGF-beta1 knockout mice pinpoints the defect to the hyperplasty/adenoma transition and reveals that the mechanism involves an inability to maintain epithelial tissue organization and not a loss of growth control, increased inflammatory activity, or increased genetic instability. These mice provide a unique opportunity to investigate the specific role of TGF-beta1 at this critical transition in the development of colon cancer.

    Topics: Adenocarcinoma; Adenoma; Adenomatous Polyposis Coli Protein; Animals; Apoptosis; beta Catenin; Biomarkers; Cecum; Cell Division; Cell Transformation, Neoplastic; Colon; Colonic Neoplasms; Crosses, Genetic; Cytoskeletal Proteins; Disease Progression; DNA; DNA-Binding Proteins; DNA, Neoplasm; Genes, APC; Genetic Predisposition to Disease; Humans; Hyperplasia; Inflammation; Intestinal Mucosa; Mice; Mice, Knockout; Microsatellite Repeats; Neoplasm Metastasis; Nuclear Proteins; Specific Pathogen-Free Organisms; Trans-Activators; Transforming Growth Factor beta

1999
p53 gene mutations and p21 protein expression induced independently of p53, by TGF-beta and gamma-rays in squamous cell carcinoma cells.
    European journal of cancer (Oxford, England : 1990), 1999, Volume: 35, Issue:2

    p53 gene mutation and the influence of TGF-beta and gamma-rays on p21 promoter activity, p21 mRNA and protein expression were investigated in nine cell lines (OSC-1 to -9) established from metastatic squamous cell carcinomas (SCC) of the cervical lymph nodes. The direct DNA sequence analysis of exons 2 to 11 of the p53 gene revealed 16 point mutations in all cell lines, but neither deletions nor additions were observed. TGF-beta upregulated p21 promoter activity by approximately 2-fold of the control and concurrently increased p21 mRNA expression, except in OSC-8 and -9. However, gamma-rays suppressed p21 promoter activity, although p21 mRNA expression in irradiated cells was increased except for OSC-8 and -9. In parallel with the messenger expression, p21 protein expression was strongly increased by TGF-beta, but only weakly increased by gamma-rays. These results indicate that point mutation of the p53 gene is frequent in metastatic SCC cells and p21 mRNA and its protein expression is p53-independently induced by both TGF-beta and gamma-rays, although the mechanism of induction by TGF-beta and gamma-rays is different.

    Topics: Apoptosis; Blotting, Western; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Gamma Rays; Genes, p53; Humans; Mutation; Reverse Transcriptase Polymerase Chain Reaction; Transforming Growth Factor beta; Tumor Cells, Cultured

1999
Dysregulation of apoptosis by c-myc in transgenic hepatocytes and effects of growth factors and nongenotoxic carcinogens.
    Molecular carcinogenesis, 1999, Volume: 25, Issue:4

    Regulation of apoptosis is an important component of multistage hepatocarcinogenesis. The proto-oncogene c-myc has been shown to be important in apoptosis regulation and to be amplified and overexpressed in human and rodent liver neoplasia. The objectives of the study reported here were to determine whether apoptosis regulation is altered in transgenic hepatocytes that overexpress c-myc and whether growth factors or nongenotoxic carcinogens alter apoptosis regulation in c-myc versus wild-type hepatocytes. Hepatocytes isolated from c-myc transgenic mice had four fold more c-myc RNA and protein (at 12-48 h) in addition to increased apoptosis levels compared with wild-type hepatocytes. The increased apoptosis in c-myc hepatocytes was accompanied by increased p53, bax, and bak and decreased bcl-2 protein levels. Hepatocytes overexpressing c-myc were more sensitive to apoptosis induced by bleomycin but less sensitive to apoptosis induced by transforming growth factor (TGF)-beta. Phenobarbital, a potent liver tumor promoter, inhibited apoptosis in c-myc hepatocytes but not in wild-type hepatocytes, decreased p53 and bax, and increased bcl-2 protein levels. Nafenopin inhibited apoptosis in both c-myc and wild-type hepatocytes, whereas 2,3,7,8-tetrachlorodibenzo-pdioxin did not inhibit apoptosis in either wild-type or c-myc hepatocytes. TGF-alpha inhibited apoptosis and increased bcl-X(L) and decreased bak protein levels in c-myc hepatocytes but not in wild-type hepatocytes. Insulin-like growth factor-II did not affect apoptosis in c-myc or wild-type hepatocytes. In this study, overexpression of c-myc altered the response to apoptotic stimuli in transgenic hepatocytes. Furthermore, phenobarbital and TGF-alpha inhibited c-myc-induced apoptosis, which may have resulted in a selective growth advantage for an initiated cell population and which may be a mechanism for tumor promotion.

    Topics: Animals; Animals, Genetically Modified; Apoptosis; Bleomycin; Carcinogens; Cell Transformation, Neoplastic; Cells, Cultured; Genes, myc; Humans; Liver; Male; Mice; Mice, Inbred C57BL; Mice, Inbred CBA; Phenobarbital; Proto-Oncogene Mas; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Suppressor Protein p53

1999
Perturbation of EGF-induced MAP kinase activation by TGF-beta 1.
    Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas, 1999, Volume: 32, Issue:7

    TGF-beta 1 regulates both cellular growth and phenotypic plasticity important for maintaining a growth advantage and increased invasiveness in progressively malignant cells. Recent studies indicate that TGF-beta-1 stimulates the conversion of epitheliod to fibroblastoid phenotype which presumably leads to the inactivation of growth-inhibitory effects by TGF-beta 1 (Portella et al. (1998) Cell Growth and Differentiation, 9: 393-404). Therefore, the investigation of TGF-beta 1 signaling that leads to altered growth and migration may provide novel targets for the prevention of increased cell growth and invasion. Although much attention has been paid to TGF-beta 1 responses in epithelial cells, the above studies suggest that examination of signal transduction pathways in fibroblasts are important as well. Data from our laboratory are consistent with the concept that TGF-beta 1 can act as a regulatory switch in density-dependent C3H 10T1/2 fibroblasts capable of either promoting or delaying G1 traverse. The regulation of this switch is proposed to occur prior to pRb phosphorylation, namely prior to activation of cyclin-dependent kinases. The current study is concerned with the evaluation of a key cyclin (cyclin D1) which activates cdk4 and p27KIP1 which in turn inhibit cdk2 in the proliferative responses of epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) and their modulation by TGF-beta 1. Although the molecular events that lead to elevation of cyclin D1 are not completely understood, it appears likely that activation of p42/p44MAPK kinases is involved in its transcriptional regulation. TGF-beta 1 delayed EGF- or PDGF-induced cyclin D1 expression and blocked the induction of active p42/p44MAPK. The mechanism by which TGF-beta 1 induces a block in p42/p44MAPK activation is being examined and the possibility that TGF-beta 1 regulates phosphatase activity is being tested.

    Topics: Cell Transformation, Neoplastic; Cyclin D1; Enzyme Activation; Epidermal Growth Factor; Humans; Mitogen-Activated Protein Kinases; Transforming Growth Factor beta

1999
Tumorigenicity of mouse BALB/c 3T3 fibroblast cells which express Epstein-Barr virus-encoded LMP1 and show normal growth phenotypes in vitro is correlated with loss of transforming growth factor-beta 1-mediated growth inhibition.
    Archives of virology, 1999, Volume: 144, Issue:2

    Latent membrane protein 1 (LMP1) encoded by the Epstein-Barr virus (EBV) genome is known to induce loss of contact inhibition and the anchorage-independent growth in rodent fibroblasts and increased expression of cell-surface activation markers and cell adhesion molecules in human B lymphocytes. To analyze the role of LMP1 in tumorigenicity, we prepared BALB/c 3T3 clones (B3LP) expressing LMP1. These B3LP cells showed non-transformed phenotypes in vitro which were characterized by normal cell morphology, contact inhibition in growth and anchorage-dependent growth. The activity of NF-kappa B induced generally in several cell lines after transfer of the LMP1 gene was not detected in B3LP cells. However, B3LP expressing LMP1 at moderate levels lost sensitivity to growth arrest by transforming growth factor-beta 1 (TGF-beta 1) and formed tumors in severe combined immune deficiency mice. Cells expressing the truncated form of LMP1 and expressing LMP1 at low level were sensitive to TGF-beta 1-mediated growth arrest and did not form tumors in mice. Therefore, cells expressing LMP1 at moderate but not at low levels formed tumors in mice and lost sensitivity to TGF-beta 1. Our results suggest that loss of TGF-beta 1-mediated growth inhibition is an important event for the tumorigenicity of LMP1-expressing cells.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cell Transformation, Viral; Fibroblasts; Gene Expression; Genes, myc; Herpesvirus 4, Human; Humans; Mice; Mice, Inbred BALB C; Mice, SCID; Neoplasms, Experimental; NF-kappa B; Phenotype; Transforming Growth Factor beta; Viral Matrix Proteins

1999
Loss of responsiveness to transforming growth factor beta induces malignant transformation of nontumorigenic rat prostate epithelial cells.
    Cancer research, 1999, Oct-01, Volume: 59, Issue:19

    Transforming growth factor (TGF)-betas are multifunctional growth factors, the properties of which include the potent inhibition of epithelial cell growth. Expression patterns of TGF-betas and TGF-beta receptors in the normal prostate indicate that these growth regulators play key roles in prostatic development and proliferative homeostasis. Importantly, TGF-beta receptor levels are frequently diminished in malignant human prostate tissue. To test the hypothesis that loss of TGF-beta responsiveness is causally involved in the tumorigenic process, we have used retroviral transduction to introduce a dominant-negative mutant type II TGF-beta receptor (DNR) into the premalignant rat prostatic epithelial cell line, NRP-152. High-level expression of the DNR abolished the ability of TGF-beta to inhibit cell growth, to promote cell differentiation, and to induce apoptosis, and it partially blocked the induction of extracellular matrix gene expression. When injected into nude mice, NRP-152-DNR cells formed carcinomas at 13 of 34 sites, compared with 0 of 30 sites for parental and control cells (P = 0.0001). We conclude that the type II TGF-beta receptor is an important tumor suppressor in the prostate, and furthermore, that loss of TGF-beta responsiveness can contribute early in the tumorigenic process by causing the malignant transformation of preneoplastic cells.

    Topics: Animals; Apoptosis; Cell Differentiation; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epithelial Cells; Extracellular Matrix Proteins; Gene Expression Regulation; Humans; Male; Mice; Mice, Nude; Prostate; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Rats; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Recombinant Proteins; Transfection; Transforming Growth Factor beta; Transplantation, Heterologous

1999
Scarring in papillary carcinoma of the thyroid: report of two new cases with exuberant nodular fasciitis-like stroma.
    Histopathology, 1999, Volume: 35, Issue:5

    To describe two new cases of papillary carcinoma of the thyroid with exuberant nodular fasciitis-like stroma, one of which was characterized by previously unreported transformation into a poorly differentiated lesion. Moreover, we explore the presence of TGF-beta to help to clarify the pathogenesis of the collagen formation.. The case characterized by an aggressive behaviour exhibited areas of transformation into a poorly differentiated (insular) carcinoma of the thyroid. In both cases, as revealed by immunohistochemistry, neoplastic cells produced and secreted high amounts of TGF-beta. On the contrary, TGF-beta immunoreaction was never present in the normal thyroid or in papillary carcinomas without collagen bundles, while a weak, exclusively intracellular reaction was present in a patchy manner in cases showing intratumoral fibrous bundles.. The rare variant of papillary thyroid carcinoma characterized by exuberant stroma may give rise to more aggressive lesions, as do other histotypes of differentiated thyroid carcinomas. TGF-beta, the fundamental cytokine which mediates scarring and activation of myofibroblasts, most probably induces the exuberant stroma.

    Topics: Adult; Aged; Carcinoma, Papillary; Cell Transformation, Neoplastic; Female; Humans; Immunoenzyme Techniques; Keloid; Male; Stromal Cells; Thyroid Gland; Thyroid Neoplasms; Transforming Growth Factor beta

1999
Antitumor efficacy of hypothemycin, a new Ras-signaling inhibitor.
    Japanese journal of cancer research : Gann, 1999, Volume: 90, Issue:10

    We have devised a new drug screening assay to discover anti-cancer drugs which inhibit Ras-mediated cellular signals, by utilizing a Ras-responsive element (RRE)-driven reporter gene system. We found that hypothemycin, an anti-bacterial, reduces RRE-dependent transcription. Treatment of tumor cells with hypothemycin resulted in reduced expression of Ras-inducible genes, including MMP (matrix metalloproteinase)-1, MMP-9, transforming growth factor-beta (TGF-beta), and vascular endothelial growth factor (VEGF), but not that of the constitutively expressed gene, MMP-2. The results of zymography demonstrated that hypothemycin reduced the production of MMP-9 and MMP-3, another Ras-inducible MMP, in the culture medium. Hypothemycin selectively inhibits anchorage-independent growth of Ras-transformed cells in comparison with anchorage-dependent growth. These findings suggest that hypothemycin inhibits Ras-mediated cellular signaling. Daily treatment of tumor-bearing mice with hypothemycin resulted in significant inhibition of tumor growth. Since MMP-1, MMP-3 and MMP-9 play important roles in tumor invasion and TGF-beta and VEGF are involved in tumor angiogenesis, hypothemycin is considered to be an example of a new class of antitumor drugs, whose antitumor efficacy can be at least partly attributed to inhibition of Ras-inducible genes.

    Topics: 3T3 Cells; Adenocarcinoma; Animals; Antineoplastic Agents; Cell Transformation, Neoplastic; Colonic Neoplasms; Endothelial Growth Factors; Female; Gene Expression Regulation, Neoplastic; Genes, ras; Genes, Reporter; Humans; Lymphokines; Matrix Metalloproteinases; Mice; Mice, Inbred BALB C; Mice, Nude; ras Proteins; Signal Transduction; Transcription, Genetic; Transfection; Transforming Growth Factor beta; Transplantation, Heterologous; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors; Zearalenone

1999
Defects in transforming growth factor-beta signaling cooperate with a Ras oncogene to cause rapid aneuploidy and malignant transformation of mouse keratinocytes.
    Proceedings of the National Academy of Sciences of the United States of America, 1999, Dec-21, Volume: 96, Issue:26

    Genetic inactivation of the transforming growth factor-beta (TGF-beta) signaling pathway can accelerate tumor progression in the mouse epidermal model of multistage carcinogenesis. By using an in vitro model of keratinocyte transformation that parallels in vivo malignant conversion to squamous cell carcinoma, we show that v-ras(Ha) transduced primary TGF-beta1-/- keratinocytes and keratinocytes expressing a TGF-beta type II dominant-negative receptor transgene have significantly higher frequencies of spontaneous transformation than control genotypes. Malignant transformation in the TGF-beta1-/- keratinocytes is preceded by aneuploidy and accumulation of chromosomal aberrations. Similarly, transient inactivation of TGF-beta signaling with a type II dominant-negative receptor adenovirus causes rapid changes in ploidy. Exogenous TGF-beta1 can suppress aneuploidy, chromosome breaks, and malignant transformation of the TGF-beta1-/- keratinocytes at concentrations that do not significantly arrest cell proliferation. These results point to genomic instability as a mechanism by which defects in TGF-beta signaling could accelerate tumor progression in mouse multistage carcinogenesis.

    Topics: Aneuploidy; Animals; Calcium; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Dose-Response Relationship, Drug; Drug Resistance; Genes, ras; Keratinocytes; Mice; Mice, Mutant Strains; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Transduction, Genetic; Transforming Growth Factor beta

1999
Sex hormone-induced mammary carcinogenesis in female Noble rats: expression of TGF-beta1 and its receptors, TGF-alpha, and EGF-R in mammary carcinogenesis.
    Breast cancer research and treatment, 1999, Volume: 58, Issue:3

    We have established a Noble rat model to explore the mechanisms of hormonal mammary carcinogenesis, in which the role of androgen in promoting mammary carcinogenesis was highlighted. We have also established that stromal-epithelial interactions may be responsible for the promotional effects of testosterone in mammary carcinogenesis. Based on these understandings, in the present study we examined the expression of transforming growth factor beta-1 (TGF-beta1) and its receptors (TGF-beta RI, TGF-beta RII), transforming growth factor alpha (TGF-alpha), and epidermal growth factor receptor (EGF-R) in 'pre-malignant' mammary glands treated with different protocols of sex hormones, as well as in mammary cancers. We observed that TGF-beta1 was strongly expressed in most mammary tumors, whereas TGF-beta RI and TGF-beta RII were negative in most mammary tumor cells. The results from comparative study of 'pre-malignant' glands further showed that when the animals were treated with testosterone, either alone or in combination with 17beta-estradiol, the mammary gland epithelial cells expressed high levels of TGF-beta1. This over-expression of TGF-beta1 can be blocked by flutamide, indicating that testosterone may be responsible for the expression of TGF-beta1 in mammary glands. TGF-beta RI and TGF-beta RII were also expressed strongly in testosterone-treated mammary epithelial cells and only weakly detectable in 17beta-estradiol treated and control mammary epithelial cells. Furthermore, TGF-beta RI and TGF-beta RII were also expressed in stromal cells, both in mammary tumors and in hormone-treated mammary glands. These observations indicate that the mechanism of testosterone in mammary carcinogenesis may be through its regulation of expression of TGF-beta1 and its receptors. On the other hand, TGF-alpha was also expressed in all 39 mammary cancers, while only 81% of the cancers were EGF-R positive. TGF-alpha was also strongly expressed in stromal cells in all three experimental groups, but only moderately expressed in epithelial cells when treated with a combination of testosterone and 17beta-estradiol. By contrast, EGF-R was strongly expressed in epithelial cells in the three experimental groups but negative in stromal cells. Flutamide or tamoxifen was unable to block the expression of TGF-alpha induced by the combined sex hormone treatment. However, they were effective in blocking the expression of TGF-alpha when the animals were treated with testosterone or 17beta

    Topics: Animals; Cell Transformation, Neoplastic; Epithelial Cells; ErbB Receptors; Estradiol; Female; Mammary Glands, Animal; Mammary Neoplasms, Animal; Rats; Testosterone; Transforming Growth Factor alpha; Transforming Growth Factor beta

1999
Conversion of normal human oral keratinocytes to tumorigenic cells is associated with the acquisition of resistance to TGF-beta.
    International journal of oncology, 1998, Volume: 12, Issue:4

    Normal human epithelial cells cannot proliferate and undergo apoptosis in the presence of transforming growth factor-beta (TGF-beta) in vitro, but many human epidermoid cancer cells are resistant to TGF-beta. Resistance to TGF-beta may thus, in part, be responsible for uncontrolled proliferation of cancer cells. Though detailed mechanisms for the resistance of cancer cells to TGF-beta remain unknown, resistance may be due to decreased expression of TGF-beta receptors from cancer cells. To investigate this possibility, we determined the expression of TGF-beta and type II TGF-beta receptor in primary normal human oral keratinocytes (NHOK), human papillomavirus-immortalized human oral keratinocytes (HOK-16B) and two tumor cell lines derived from HOK-16B (CTHOK-16B-BaP and CTHOK-16B-DMBA). Our results show that (1) the cellular and secretory TGF-beta levels in immortalized and tumor cells were notably lower than in NHOK and (2) the level of type II TGF-beta receptor of the tested cells was similar to each other. Taken together, the conversion of NHOK to tumorigenic cells may, in part, be due to the acquisition of NHOK resistance to TGF-beta through underexpression of this cytokine.

    Topics: Apoptosis; Cell Division; Cell Transformation, Neoplastic; Humans; Keratinocytes; Mouth Mucosa; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

1998
Characterization of the MADH2/Smad2 gene, a human Mad homolog responsible for the transforming growth factor-beta and activin signal transduction pathway.
    Genomics, 1998, Feb-15, Volume: 48, Issue:1

    The transforming growth factor beta (TGF-beta) superfamily is a family of multifunctional cytokines that transduce signals via serine/threonine kinase receptors. Recent studies revealed that Mothers against dpp (Mad) in Drosophila and its homologs play important roles in the intracellular signal transduction of the serine/threonine kinase receptors. In mammals, one of the Mad homologs, MADH2 (also termed Smad2), was reported to be a mediator of TGF-beta and activin signaling and was found mutated in some of the colon and lung cancer cases. We describe here the genomic organization of the human MADH2 gene. The gene is composed of 12 exons; 2 exons 1, i.e., exon 1a and 1b, are used separately or in conjunction to form exon 1a-exon 1b-exon 2 alternatively spliced mRNA. The 2 exons 1 are closely located, and the MADH2 mRNAs are transcribed from two promoters in one CpG island. The promoter activity in the 5' upstream sequence was confirmed by the luciferase assay. The 3' end of the mRNA is heterogenous, and we found several polyadenylation signals. Northern blot analysis revealed high expression of the MADH2 mRNA, e.g., in skeletal muscle, heart, and placenta. RT-PCR assay using primers in exons 2 and 4 and direct nucleotide sequencing proved that exon 3 is spliced out in about 10% of MADH2 in human placenta. These data will be valuable for studying the MADH2 function in both normal cells and cancer cells.

    Topics: Activins; Amino Acid Sequence; Animals; Base Sequence; Basic Helix-Loop-Helix Leucine Zipper Transcription Factors; Blotting, Northern; Cell Transformation, Neoplastic; COS Cells; DNA-Binding Proteins; Exons; Humans; Inhibins; Introns; Molecular Sequence Data; Polymerase Chain Reaction; Promoter Regions, Genetic; Repressor Proteins; RNA, Messenger; Sequence Homology, Amino Acid; Signal Transduction; Smad2 Protein; Trans-Activators; Transforming Growth Factor beta

1998
Topical retinoic acid reduces skin papilloma formation but resistant papillomas are at high risk for malignant conversion.
    Cancer research, 1998, Apr-01, Volume: 58, Issue:7

    Retinoic acid (RA) was topically applied to the skin of Sencar mice during the promotion phase of specific tumor induction protocols that produce papillomas at low (12-O-tetradecanoylphorbol-13-acetate promoted, TPA) or high (mezerein-promoted) risk for premalignant progression and malignant conversion. RA consistently reduced the yield of papillomas and carcinomas in both protocols, but the frequency of malignant conversion in papillomas that emerged during RA treatment was not reduced. When TPA was reapplied after cessation of RA treatment, the number of papillomas increased 2-fold, suggesting that RA had not eliminated initiated cells. In vitro, RA prevented the emergence of transformed keratinocytes in an assay that mimics malignant conversion, suggesting that RA can suppress conversion if applied during the stage of premalignant progression. Examination of tumor markers at weeks 14 and 22 of the tumor-induction experiments in vivo indicated that papillomas evolving during RA treatment exhibited a phenotype of high progression risk, even in the TPA-promoted groups. In the majority of these tumors, the alpha6beta4 integrin and retinoid X receptor alpha transcripts were detected suprabasally, indicating an advanced state of premalignant progression. RA-treated tumors also expressed higher levels of transcripts for transforming growth factor (TGF)-beta1 and localized TGF-beta1 peptide in the basal portions of the tumor fronds. Because up-regulated expression of TGF-beta1 suppresses papilloma formation, these studies suggest a mechanism whereby RA can prevent papilloma eruption via a TGF-beta intermediate, but papillomas resistant to RA may have altered TGF-beta signaling and progress to carcinomas at an increased frequency.

    Topics: Administration, Topical; Animals; Anticarcinogenic Agents; Antineoplastic Agents; Biomarkers, Tumor; Carcinogens; Carcinoma, Basal Cell; Cell Transformation, Neoplastic; Disease Progression; Diterpenes; Female; Immunohistochemistry; Mice; Mice, Inbred BALB C; Mice, Inbred SENCAR; Papilloma; Phenotype; Precancerous Conditions; Receptors, Retinoic Acid; Retinoid X Receptors; Risk Factors; Skin Neoplasms; Terpenes; Tetradecanoylphorbol Acetate; Transcription Factors; Transforming Growth Factor beta; Tretinoin

1998
Overexpression of PKCepsilon in R6 fibroblasts causes increased production of active TGFbeta.
    Journal of cellular physiology, 1998, Volume: 175, Issue:3

    In previous studies, our laboratory demonstrated that Rat 6 (R6) fibroblasts which stably overproduce high levels of PKCepsilon display abnormalities in growth control that are characteristic of malignant transformation (Cacace et al., 1993, Oncogene, 8:2095-2104). The R6-PKCepsilon overproducing cell lines also exhibited a decreased growth factor requirement. The present study demonstrates that conditioned medium (CM) from two individual clones, R6-PKCepsilon 10 and 30, stimulates DNA synthesis in control R6-C1 cells. Maximal DNA synthesis and morphologic transformation was achieved in control cells when they were treated with medium from R6-PKCepsilon cells grown in the presence of TPA (TPA-CM). Size fractionation of the TPA-CM from PKCepsilon 30 cells revealed that this activity is due to a factor(s) that has an apparent molecular weight in the range of 10-30 kD and is heat and acid stable. This factor, like TGFbeta1, stimulated anchorage-independent growth of NRK cells. Western blot analysis (under nonreducing conditions) of the TPA-CM from R6-PKCepsilon 30 and R6-PKCepsilon 10 cells revealed the presence of the 25 kD active forms of TGFbeta2 and 3. These active forms of TGFbeta were not found in the CM of control R6 cells, or R6 cells that overexpress PKCalpha or PKCbeta1. The addition of a pan-specific TGFbeta antibody to NRK cells treated with the 10-30 kD fraction of TPA-CM from PKCepsilon 30 cells blocked the ability of this material to stimulate thymidine incorporation. Taken together, these studies suggest that the oncogenic activity of PKCepsilon in R6 cells is due, at least in part, to its ability to induce production of the active forms of TGFbeta2 and 3.

    Topics: Animals; Cell Adhesion; Cell Division; Cell Line; Cell Transformation, Neoplastic; Culture Media, Conditioned; DNA; Fibroblasts; Isoenzymes; Mitogens; Molecular Weight; Protein Kinase C; Protein Kinase C-epsilon; Rats; RNA, Messenger; Tetradecanoylphorbol Acetate; Transforming Growth Factor beta

1998
Reduction in transforming growth factor-beta type II receptor in mouse lung carcinogenesis.
    Molecular carcinogenesis, 1998, Volume: 22, Issue:1

    Transforming growth factor-beta (TGF-beta) is a growth modulator that inhibits the proliferation of many epithelial cells through interaction with its receptors, the type I and type II receptors (TGF-beta RI and RII) by activating their serine/threonine kinase activities. Loss of growth inhibition by TGF-beta is thought to contribute to the development of many types of tumors. To examine the roles of TGF-beta1, -beta2, and -beta3 and TGF-beta RI and RII in chemically induced mouse lung tumorigenesis, we used immunohistochemical and in situ hybridization analyses to measure the expression of their proteins and mRNAs in A/J mice treated with the carcinogen urethane to induce lung adenomas. Immunostaining for the TGF-beta ligands and receptors was detected in the epithelia of the bronchioles of untreated and treated A/J mice at similar levels. Immunostaining for the TGF-beta ligands and receptors was also detected in adenomas by 2 mo. While immunostaining for TGF-beta1, -beta2, and -beta3 and TGF-beta RI in adenomas was detected at levels comparable to those in bronchioles, immunostaining for TGF-beta RII was less intense in adenomas than in bronchioles. Decreased immunostaining for TGF-beta RII in adenomas persisted for at least 8 mo after exposure to urethane, whereas immunostaining for TGF-beta1, -beta2, and -beta3 and TGF-beta RI persisted at levels comparable to those in normal bronchioles. In situ hybridization studies conducted with TGF-beta receptor riboprobes showed a corresponding reduction in expression of TGF-beta RII mRNA but not of TGF-beta RI mRNA in adenomas compared with expression in bronchioles. Expression of TGF-beta RII mRNA was also examined in non-tumorigenic and tumorigenic mouse lung cells; expression of TGF-beta RII mRNA was lower in the tumorigenic cells derived from urethane-induced lung tumors. These data suggest that a decrease in expression of TGF-beta RII may contribute to autonomous cell growth and may play an important role in mouse lung carcinogenesis induced by urethane.

    Topics: Activin Receptors, Type I; Animals; Carcinogens; Cell Division; Cell Line; Cell Transformation, Neoplastic; Down-Regulation; Epithelial Cells; Female; Lung; Lung Neoplasms; Mice; Mice, Inbred A; Polymerase Chain Reaction; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Messenger; Transcription, Genetic; Transforming Growth Factor beta; Urethane

1998
Constitutive association of EGF receptor with the CrkII-23 mutant that inhibits transformation of NRK cells by EGF and TGF-beta.
    Cellular signalling, 1998, Volume: 10, Issue:4

    Crk belongs to the adapter proteins that participate in many signalling pathways from cell surface receptors. We have characterised the CrkII-23 mutant that inhibits the transformation of NRK cells induced by epidermal growth factor (EGF) and transforming growth factor (TGF)-beta. To study the biochemical difference, cDNAs of the wild-type CrkII and the CrkII-23 mutant were introduced stably into NIH 3T3 cells expressing EGF receptor (EGFR). Both CrkII and CrkII-23 were phosphorylated on tyrosine upon EGF simulation with similar time course and dose dependency. Whereas the wild-type CrkII bound to EGFR only after EGF stimulation, CrkII-23 bound to EGFR from before stimulation. Mutation in the Src homology (SH) 2 or amino-terminal SH3 domain did not abolish the binding of CrkII-23 to EGFR in the quiescent cells, suggesting that the binding is mediated by a novel mechanism. These CrkII-23-derived mutants, however, did not suppress transformation of NRK cells by EGF and TGF-beta. Hence, both the SH2 and amino-terminal SH3 domains are required to inhibit transformation of NRK cells. These results suggest that persistent signalling from CrkII-23 bound to EGFR suppresses transformation by EGF and TGF-beta in NRK23 cells.

    Topics: 3T3 Cells; Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Animals; Cell Differentiation; Cell Transformation, Neoplastic; COS Cells; DNA Mutational Analysis; Epidermal Growth Factor; ErbB Receptors; Eukaryotic Initiation Factor-2; Guanine Nucleotide Exchange Factors; Humans; Mice; Mutation; Neurons; PC12 Cells; Phosphorylation; Protein Binding; Protein Kinases; Protein Structure, Tertiary; Proteins; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-abl; Proto-Oncogene Proteins c-cbl; Proto-Oncogene Proteins c-crk; Rats; Shc Signaling Adaptor Proteins; Src Homology 2 Domain-Containing, Transforming Protein 1; src Homology Domains; Transforming Growth Factor beta; Tyrosine; Ubiquitin-Protein Ligases

1998
Suppression of decorin expression and partial induction of anchorage-independent growth by the v-src oncogene in human fibroblasts.
    European journal of biochemistry, 1998, Jun-01, Volume: 254, Issue:2

    To determine its role in cell transformations, the v-src oncogene was introduced into the human foetal diploid fibroblasts MRC-5 and into MRC-SV1, a simian virus 40 (SV40)-transformed cell line derived from them. Infected cells were found to contain stably integrated intact proviruses, as determined by Southern blot analysis. Although highly expressed, v-src did not change the morphology or growth patterns of MRC-5 cells and failed to induce foci or alter their saturation densities. However, overexpression of v-src reduced the plating efficiencies of MRC-5 and induced anchorage-independent growth in a low but significant number of cells. Northern blot analysis showed that v-src selectively abolished the expression of decorin, a small dermatan/chondroitin sulphate proteoglycan that interacts with extracellular-matrix components and modulates collagen-fibril formation and the activity of transforming growth factor (TGF) beta1. Addition of herbimycin A, a potent pp60src tyrosine-kinase inhibitor, resulted in the reexpression of decorin in MRC-5 carrying v-src. There were no changes in the expressions of fibronectin, procollagen type I, or tissue plasminogen activator, an activator of extracellular-matrix-degrading enzymes. Moreover, v-src did not alter the expressions of the epidermal-growth-factor receptor or TGFbeta1 or reduce the growth-factor requirements of MRC-5 fibroblasts. MRC-5 and MRC-SV1 expressing v-src remained non-tumourigenic when injected into nude mice. Constitutive expression of v-src did not alter the mRNA levels of c-jun and junB, suggesting that the effects of the oncogene are not mediated by AP-1. Decorin gene expression has been shown previously to be maximal in quiescent cells and virtually absent in transformed ones. Our data indicate that the ability to synthesise decorin can be suppressed in human fibroblasts without their becoming transformed, and that the relations between decorin synthesis and growth controls need further clarification.

    Topics: Animals; Cell Adhesion; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cell Transformation, Viral; Decorin; Defective Viruses; Extracellular Matrix Proteins; Fibroblasts; Gene Expression; Genes, src; Humans; Mice; Proteoglycans; Proto-Oncogenes; Retroviridae; RNA, Messenger; Simian virus 40; Transforming Growth Factor beta

1998
SV40 Tag transformation of the normal invasive trophoblast results in a premalignant phenotype. I. Mechanisms responsible for hyperinvasiveness and resistance to anti-invasive action of TGFbeta.
    International journal of cancer, 1998, Jul-29, Volume: 77, Issue:3

    Invasion of the uterus by first trimester human placental extravillous trophoblast (EVT) cells depends on mechanisms shared by malignant cells. However, unlike tumor invasion, trophoblast invasion of the uterus is stringently controlled in situ by local molecules such as transforming growth factor (TGF)beta. Since EVT cells possess active invasion-associated genes but are nontumorigenic, our objective was to induce premalignant and then malignant phenotype into a normal EVT cell line in order to identify the molecular basis of tumor progression. Simian virus 40 large T antigen (SV40 Tag) was introduced into a normal human first trimester invasive EVT cell line, HTR8, established in our laboratory. Since the HTR8 line has a limited in vitro lifespan of 12-15 passages, SV40 Tag-transformed cells were selected on the basis of extended lifespan. A long-lived line, RSVT-2, was produced and an immortalized subclone, RSVT2/C, was further derived under a forced crisis regimen. We examined transformation-induced alterations in proliferative and invasive abilities, responses to the invasion and proliferation-regulating growth factor TGFbeta and changes in gene expression for invasion-associated enzymes or enzyme inhibitors. RSVT-2 and RSVT2/C cell lines were hyperproliferative and hyperinvasive when compared with the parental HTR8 cell line. They were also variably resistant to the anti-proliferative and anti-invasive signals from TGFbeta. Since both cell lines remained non-tumorigenic in nude mice, these properties indicate that they attained a premalignant phenotype. Both cell lines showed reduced expression of tissue inhibitor of metalloproteases (TIMP)-1, while TIMP-2 and plasminogen activator inhibitor (PAI)-I expression was was also reduced in RSVT2/C cells, thus contributing to their hyperinvasiveness. Their resistance to the anti-invasive action of TGFbeta was explained by the failure of TGFbeta to upregulate TIMPs and PAI-I, in contrast to the TGFbeta-induced upregulation noted in parental HTR8 cells.

    Topics: Animals; Antigens, Viral, Tumor; Cell Division; Cell Line; Cell Transformation, Neoplastic; Choriocarcinoma; Clone Cells; Drug Resistance, Neoplasm; Female; Gene Expression Regulation, Neoplastic; Humans; Mice; Mice, Nude; Neoplasm Invasiveness; Phenotype; Plasminogen Activator Inhibitor 1; Precancerous Conditions; Pregnancy; Pregnancy Trimester, First; Simian virus 40; Tissue Inhibitor of Metalloproteinase-1; Tissue Inhibitor of Metalloproteinase-2; Transforming Growth Factor beta; Transplantation, Heterologous; Trophoblasts; Tumor Cells, Cultured

1998
Z-138: a new mature B-cell acute lymphoblastic leukemia cell line from a patient with transformed chronic lymphocytic leukemia.
    Leukemia research, 1998, Volume: 22, Issue:4

    We describe a new mature B-cell acute lymphoblastic leukemia (ALL) cell line designated Z-138 that was derived from a patient with chronic lymphocytic leukemia (CLL) whose disease underwent transformation to a rare, aggressive form of mature B-cell ALL. This cell line has an L3 morphology, ultrastructural characteristics of lymphoblasts, B-lineage surface markers and an immunoglobulin heavy-chain gene rearrangement identical to the rearrangement observed in the patient's blasts from whom the cell line was derived. Z-138 cells produce granulocyte-macrophage colony-stimulating factor (GM-CSF) and high levels of granulocyte-CSF (G-CSF), but they do not exhibit a proliferative response to either cytokine. Both the patient's lymphoblasts and Z-138 cells exhibited cytogenetic abnormalities including t(8;14), t(14;18) and a chromosome 11 abnormality similar to the t(11;14) of the parental cells, resulting in marked overexpression of cyclin D1 (BCL-1 (PRAD1)) mRNA in Z-138 cells. Since these karyotypic anomalies have been associated with low grade (t(14;18)), intermediate grade (t(11;14)) and high grade (t(8;14)) lymphomas, their development may be involved in the unusual aggressive transformation of this patient's CLL.

    Topics: Aged; Blotting, Southern; Bone Marrow Cells; Burkitt Lymphoma; Cell Transformation, Neoplastic; Cell Transformation, Viral; Chromosome Aberrations; Chromosomes, Human, Pair 9; Clone Cells; DNA; Fusion Proteins, bcr-abl; Gene Rearrangement, B-Lymphocyte, Heavy Chain; Granulocyte Colony-Stimulating Factor; Granulocyte-Macrophage Colony-Stimulating Factor; Herpesvirus 4, Human; Humans; Immunoglobulin J-Chains; Immunophenotyping; Interleukin-1; Interleukin-6; Karyotyping; Leukemia, Lymphocytic, Chronic, B-Cell; Lymphocyte Activation; Male; Microscopy, Electron; RNA; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

1998
A retinoblastoma-binding protein that affects cell-cycle control and confers transforming ability.
    Nature genetics, 1998, Volume: 19, Issue:4

    The retinoblastoma (RB) gene is one of the most extensively studied tumour-suppressor genes. Deletion or inactivation of both RB alleles is an essential, rate-limiting step in the formation of retinoblastoma and osteosarcoma that arise in families that carry mutant RB (ref. 2). RB inactivation is also found in other human tumours. Whereas loss of RB function is associated with the loss of cellular proliferative control, introduction of a wild-type RB can suppress cell growth and tumorigenicity. Thus, identification of factors that interfere with and/or control the function of the RB protein is critical for understanding both cell-cycle control and oncogenesis. Here we describe a new gene, Bog (for B5T over-expressed gene), which was identified and shown to be overexpressed in several transformed rat liver epithelial (RLE) cell lines resistant to the growth-inhibitory effect of TGF-beta1, as well as in primary human liver tumours. The Bog protein shares homology with other retinoblastoma-binding proteins and contains the Rb-binding motif LXCXE. Using the yeast two-hybrid system and co-immunoprecipitation, we demonstrated that Bog binds to Rb. In vivo, Bog/Rb complexes do not contain E2F-1, and Bog can displace E2F-1 from E2F-1/Rb complexes in vitro. Overexpression of Bog in normal RLE cells conferred resistance to the growth-inhibitory effect of TGF-beta1. Furthermore, normal RLE cells are rapidly transformed when Bog is continuously overexpressed and form hepatoblastoma-like tumours when transplanted into nude mice. These data suggest that Bog may be important in the transformation process, in part due to its capacity to confer resistance to the growth-inhibitory effects of TGF-beta1 through interaction with Rb and the subsequent displacement of E2F-1.

    Topics: Amino Acid Sequence; Animals; Carcinoma, Hepatocellular; Carrier Proteins; Cell Cycle; Cell Cycle Proteins; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cloning, Molecular; DNA-Binding Proteins; E2F Transcription Factors; E2F1 Transcription Factor; Epithelial Cells; Humans; Hydrolases; Intracellular Signaling Peptides and Proteins; Liver; Mice; Mice, Nude; Molecular Sequence Data; Neoplasm Proteins; Organ Specificity; Rats; Retinoblastoma Protein; Retinoblastoma-Binding Protein 1; RNA, Messenger; RNA, Neoplasm; Sequence Homology, Amino Acid; Serine Proteases; Transcription Factor DP1; Transcription Factors; Transforming Growth Factor beta; Tumor Cells, Cultured; Werner Syndrome Helicase; Xenopus Proteins

1998
Crystal structure of a Smad MH1 domain bound to DNA: insights on DNA binding in TGF-beta signaling.
    Cell, 1998, Sep-04, Volume: 94, Issue:5

    The Smad family of proteins, which are frequently targeted by tumorigenic mutations in cancer, mediate TGF-beta signaling from cell membrane to nucleus. The crystal structure of a Smad3 MH1 domain bound to an optimal DNA sequence determined at 2.8 A resolution reveals a novel DNA-binding motif. In the crystals, base-specific DNA recognition is provided exclusively by a conserved 11-residue beta hairpin that is embedded in the major groove of DNA. A surface loop region, to which tumorigenic mutations map, has been identified as a functional surface important for Smad activity. This structure establishes a framework for understanding how Smad proteins may act in concert with other transcription factors in the regulation of TGF-beta-responsive genes.

    Topics: Amino Acid Sequence; Binding Sites; Cell Transformation, Neoplastic; Crystallization; Crystallography, X-Ray; DNA; DNA-Binding Proteins; Humans; Models, Molecular; Molecular Sequence Data; Mutation; Peptide Fragments; Protein Structure, Secondary; Protein Structure, Tertiary; Signal Transduction; Smad3 Protein; Trans-Activators; Transforming Growth Factor beta

1998
Decreased fibronectin expression in Met/HGF-mediated tumorigenesis.
    Oncogene, 1998, Sep-03, Volume: 17, Issue:9

    The tyrosine kinase receptor Met and its ligand, hepatocyte growth factor (HGF)/scatter factor are involved in the etiology and progression of a number of human cancers. Coexpression of Met and HGF in mesenchymal cells increases the tumorigenic and metastatic potential of the cells. In the studies described here, we used differential display screening to identify changes in gene expression that are initiated by Met/HGF, and that may lead to these phenotypes. We learned that Met/HGF signaling resulted in greatly decreased fibronectin mRNA production in three different human and mouse tumor cell lines; these decreases in fibronectin mRNA were paralleled by decreases in fibronectin protein. We also found a progressive decrease in fibronectin in tumor explants and metastases derived from the Met/HGF transformed cells. The absence of fibronectin expression is a frequent cancer phenotype; our results indicate that decreases in fibronectin correlate with, but are not essential for, MetHGF/SF-mediated tumorigenesis.

    Topics: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Fibronectins; Gene Expression Regulation, Neoplastic; Hepatocyte Growth Factor; Humans; Mice; Mice, Nude; Neoplasms, Experimental; Proto-Oncogene Proteins c-met; RNA, Messenger; Transforming Growth Factor beta; Tumor Cells, Cultured

1998
Transforming growth factor beta 1 induces squamous carcinoma cell variants with increased metastatic abilities and a disorganized cytoskeleton.
    Experimental cell research, 1998, Nov-01, Volume: 244, Issue:2

    Previous studies indicated that mouse transformed keratinocytes undergo an epithelial-fibroblastic conversion when cultured in the presence of TGF-beta1. This conversion is associated in vivo with a squamous-spindle carcinoma transition. We derived epithelioid (A6, FPA6) and spindle (B5) clonal cell variants from a squamous carcinoma cell line (PDV) after treatment with TGF-beta1. FPA6 cells were isolated from the ascites fluid of an A6-tumor-bearing mouse. FPA6 and A6 cell lines produced in nude mice mixed carcinomas with a squamous and poorly differentiated component. Both cell lines coexpressed keratins and vimentin and synthesized E-cadherin protein, although FPA6 cells cultured at early passages (FPA6-ep) had reduced levels of E-cadherin mRNA and increased synthesis of keratin K8, a marker of malignant progression. Immunofluorescence analysis revealed that FPA6-ep cells exhibited a disorganized cytoskeleton with keratins forming focal juxtanuclear aggregates and loss of F-actin stress fibers and cortical bundles, and E-cadherin was localized in the cytoplasm out of cell-cell contact areas. Sporadic cells in A6 and PDV cultures also presented those anomalous keratin structures, suggesting that FPA6 cells originated from a subpopulation of A6 tumor cells that metastasized into the peritoneal cavity. The analysis of the spontaneous and experimental metastatic potentials of the cell lines showed that epithelioid and fibroblastic cell variants had acquired metastatic abilities compared to PDV which was nonmetastatic. The FPA6-ep cell line exhibited a highly aggressive behavior, killing the animals at about 17 days after intravenous injection of the cells into athymic mice. The phenotype of FPA6-ep cells was unstable and reverted at later passages in which the normal organization of keratin and F-actin in filaments and the localization of E-cadherin at cell-cell contacts were restored. This phenotypic reversion occurred concomitantly with a reduction of the experimental metastatic potential of FPA6 cells.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Cytoskeleton; Epithelial Cells; Fibroblasts; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Proteins; Phenotype; Transforming Growth Factor beta; Tumor Cells, Cultured

1998
p53-independent role of MDM2 in TGF-beta1 resistance.
    Science (New York, N.Y.), 1998, Dec-18, Volume: 282, Issue:5397

    Transforming growth factor-beta (TGF-beta) inhibits cell proliferation, and acquisition of TGF-beta resistance has been linked to tumorigenesis. A genetic screen was performed to identify complementary DNAs that abrogated TGF-beta sensitivity in mink lung epithelial cells. Ectopic expression of murine double minute 2 rescued TGF-beta-induced growth arrest in a p53-independent manner by interference with retinoblastoma susceptibility gene product (Rb)/E2F function. In human breast tumor cells, increased MDM2 expression levels correlated with TGF-beta resistance. Thus, MDM2 may confer TGF-beta resistance in a subset of tumors and may promote tumorigenesis by interference with two independent tumor suppressors, p53 and Rb.

    Topics: Animals; Breast Neoplasms; Carrier Proteins; Cell Cycle Proteins; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA-Binding Proteins; Drug Resistance, Neoplasm; E2F Transcription Factors; Gene Expression; Genes, p53; Genes, Retinoblastoma; Genetic Vectors; Humans; Mice; Mink; Nuclear Proteins; Phosphorylation; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-mdm2; Retinoblastoma Protein; Retinoblastoma-Binding Protein 1; Signal Transduction; Transcription Factor DP1; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Protein p53

1998
Reactive oxygen species as mediators of the transformed phenotype.
    International journal of cancer, 1997, Mar-04, Volume: 70, Issue:5

    Reactive oxygen species (ROS) are known to be involved in different pro- and anticarcinogenic mechanisms. However, their influence on the maintenance of the transformed phenotype has not been studied so far. Here we show that the anchorage-independent growth of transformed murine fibroblasts is inhibited by antioxidants and radical scavengers in a concentration-dependent and reversible manner. These agents also reduce TGF-beta-dependent stimulation of colony formation in soft agar, pointing to their specific interference with TGF-beta-triggered signal chains involved in the maintenance of the transformed state.

    Topics: 3T3 Cells; Animals; Cell Transformation, Neoplastic; Colony-Forming Units Assay; Dimethyl Sulfoxide; Mice; Phenotype; Reactive Oxygen Species; Transforming Growth Factor beta

1997
Fibroblasts transformed by chemical carcinogens are sensitive to intercellular induction of apoptosis: implications for the control of oncogenesis.
    Carcinogenesis, 1997, Volume: 18, Issue:2

    The ability of neighbouring normal cells to inhibit proliferation of transformed cells is regarded as the classical mode of intercellular control of potential tumour cells. This mechanism, however, only controls the pool size of transformed cells, but does not impair their survival. We have recently shown that cells transformed by biological agents are subject to a novel control system: transforming growth factor beta (TGF-beta) induces normal cells to release factors that mediate apoptosis specifically in transformed cells. Here we show that cells transformed by chemical carcinogens are also subject to this dominant control mechanism. The number of foci induced by methylcholanthrene, N-methyl-N'-nitro-N-nitrosoguanidine or quercetin was significantly reduced when the cultures were treated with TGF-beta. Established lines of chemically transformed cells proved to be sensitive to induction of apoptosis by neighbouring normal cells in the presence of TGF-beta. This finding demonstrates that sensitivity to induction of apoptosis is a general feature of transformed cells, irrespective of the transforming agent. It is particularly relevant for chemical carcinogenesis. As transformed cells were shown to trigger induction of their own apoptosis, the acquisition of resistance to this process may be a central regulatory step in carcinogenesis in vitro and possibly also in vivo. This study may help to elucidate mechanisms that protect transformed cells at an early stage of tumour progression that has until now not been the focus of investigation.

    Topics: Animals; Apoptosis; Carcinogens; Cell Communication; Cell Count; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; DNA Fragmentation; Fibroblasts; Methylcholanthrene; Methylnitronitrosoguanidine; Quercetin; Transforming Growth Factor beta; Tumor Stem Cell Assay

1997
Histomorphological and immunohistochemical characterization of colonic aberrant crypt foci in rats: relationship to growth factor expression.
    Carcinogenesis, 1997, Volume: 18, Issue:3

    Many studies have been conducted to assess the potential preneoplastic nature of colonic aberrant crypt foci (ACF), but still the biological significance of these foci and their relationship to colon neoplasia remains to be elucidated. In the present paper a battery of variables suggested to be indicative for colon cancer development has been studied in relation to ACF in rats. These include: (i) the degree of dysplasia; (ii) the type of mucus production; (iii) the cellular immunohistochemical expression and distribution of transforming growth factors alpha and beta and their respective receptors, epidermal growth factor receptor and transforming growth factor beta receptors I and II and phosphorylated cellular tyrosine. The parameters have been investigated in ACF selected from a previous study where the foci were induced under different circumstances, leading to disparities in the number as well as the crypt multiplicity obtained. The present study showed that for all parameters investigated, apart from sialomucin production, the different experimental conditions had no effect on the individual ACF, irrespective of the number and distribution of the different categories of ACF among the various diets. However, it was shown that the degree of dysplasia correlated strongly with crypt multiplicity and that all the investigated ACF lacked expression of transforming growth factor alpha and expressed a reduced amount of transforming growth factor beta compared with normal crypts. These observations may indicate that ACF are preneoplastic lesions and supports the suggestion that they may, at least in the rat, have the potential to gradually progress to tumors, but no single ACF showed particular characteristics indicating specific proneness to tumor development. The study could not confirm the presence of sialomucin-producing ACF as a valid marker for tumor development.

    Topics: Animals; Cell Transformation, Neoplastic; Colon; Colonic Neoplasms; Dietary Fats; Dietary Fiber; Dietary Sucrose; ErbB Receptors; Gene Expression Regulation; Growth Substances; Intestinal Mucosa; Male; Mucins; Precancerous Conditions; Rats; Receptors, Growth Factor; Receptors, Transforming Growth Factor beta; Sialomucins; Transforming Growth Factor alpha; Transforming Growth Factor beta

1997
Interference of bcl-2 with intercellular control of carcinogenesis.
    International journal of cancer, 1997, May-16, Volume: 71, Issue:4

    Induction of apoptosis in transformed fibroblasts by surrounding normal cells has been discussed as a potent early control step in carcinogenesis. According to this hypothesis, tumor progression should require resistance of transformed cells against this TGF-beta-triggered control mechanism. Here we show that Bcl-2, a protein involved in inhibition of apoptosis, can protect transformed cells from induction of apoptosis by surrounding cells. Rather than acting on the transformation process itself, Bcl-2 may thus represent an efficient modulator of carcinogenesis at an intercellular level.

    Topics: Animals; Apoptosis; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Disease Progression; Fibroblasts; Mice; Mice, Inbred C3H; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor beta; Ultraviolet Rays

1997
Induction of basal cell carcinoma features in transgenic human skin expressing Sonic Hedgehog.
    Nature medicine, 1997, Volume: 3, Issue:7

    Hedgehog (HH) signaling proteins mediate inductive events during animal development. Mutation of the only known HH receptor gene, Patched (PTC), has recently been implicated in inherited and sporadic forms of the most common human cancer, basal cell carcinoma (BCC). In Drosophila, HH acts by inactivating PTC function, raising the possibility that overexpression of Sonic Hedgehog (SHH) in human epidermis might have a tumorigenic effect equivalent to loss of PTC function. We used retroviral transduction of normal human keratinocytes to constitutively express SHH. SHH-expressing cells demonstrated increased expression of both the known HH target, BMP-2B, as well as bcl-2, a protein prominently expressed by keratinocytes in BCCs. These keratinocytes were then used to regenerate human skin transgenic for long terminal repeat-driven SHH (LTR-SHH) on immune-deficient mice. LTR-SHH human skin consistently displays the abnormal specific histologic features seen in BCCs, including downgrowth of epithelial buds into the dermis, basal cell palisading and separation of epidermis from the underlying dermis. In addition, LTR-SHH skin displays the gene expression abnormalities previously described for human BCCs, including decreased BP180/BPAG2 and laminin 5 adhesion proteins and expression of basal epidermal keratins. These data indicate that expression of SHH in human skin recapitulates features of human BCC in vivo, suggest that activation of this conserved signaling pathway contributes to the development of epithelial neoplasia and describe a new transgenic human tissue model of neoplasia.

    Topics: Animals; Autoantigens; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Carcinoma, Basal Cell; Cell Adhesion Molecules; Cell Transformation, Neoplastic; Cell Transplantation; Cells, Cultured; Collagen Type XVII; Disease Models, Animal; Genetic Vectors; Hedgehog Proteins; Humans; Kalinin; Keratinocytes; Mice; Mice, Nude; Mice, Transgenic; Non-Fibrillar Collagens; Protein Biosynthesis; Proteins; Proto-Oncogene Proteins c-bcl-2; Recombinant Fusion Proteins; Recombinant Proteins; Retroviridae; Skin Neoplasms; Trans-Activators; Transforming Growth Factor beta

1997
Restored expression of transforming growth factor beta type II receptor in k-ras-transformed thyroid cells, TGF beta-resistant, reverts their malignant phenotype.
    Journal of cellular physiology, 1997, Volume: 172, Issue:2

    Transforming growth factor beta 1 (TGF beta 1) inhibits the growth of normal rat epithelial thyroid cells (FRTL-5 strain) by counteracting thyrotropin (TSH)-stimulated DNA synthesis and by slowing the cells in the G1 phase of the cell cycle. Here, we have studied two clones of FRTL-5 thyroid cell line transformed by the wild type (wt) v-k-ras oncogene (K.M.A1, K.M.A2) and one clone (A6) transformed by a temperature-sensitive (ts) v-k-ras mutant. Anchorage-dependent as well as anchorage-independent growth of these k-ras-transformed cells was not inhibited by TGF beta 1. TGF beta 1 resistance appeared to be dependent by a functional p21 k-ras, because A6 cell growth was partially inhibited at the nonpermissive temperature (39 degrees C). To determine the basis for TGF beta 1 resistance in k-ras-transformed thyroid cells, we looked for possible defects in the expression of type I (T beta R-I/ALK5) and type II TGF beta receptors (T beta R-II). Lower levels of type II receptors were present in all of the k-ras-transformed clones, as revealed by both Northern blot and cross-linking experiments. A partial reversion of the malignant phenotype of the wt k-ras-transformed clone was obtained in two clones isolated after transfection of the malignant thyroid cells (K.M.A1) with a T beta R-II expression vector. These two clones also showed restored levels of exogenous T beta R-II mRNA and protein, and both clones showed a partially reacquired sensitivity to TGF beta 1. Similarly, the reversion of the malignant phenotype of the A6 clone grown at the nonpermissive temperature was accompanied by a restored expression of the T beta R-II receptors. These data indicate that active k-ras oncogene can induce TGF beta 1 resistance in rat thyroid cells and suggest that one of the possible mechanisms of escape from TGF beta 1 growth control in k-ras-induced thyroid carcinogenesis involves a reduced expression of T beta R-II receptors.

    Topics: Animals; Carrier Proteins; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Drug Resistance; Genes, ras; Phenotype; Rats; Receptors, Transforming Growth Factor beta; RNA, Messenger; Thyroid Gland; Transfection; Transforming Growth Factor beta

1997
[Inhibition of EGF and TGF-beta dependent transformation of NRK23 cells by Crk II-23 mutant].
    [Hokkaido igaku zasshi] The Hokkaido journal of medical science, 1997, Volume: 72, Issue:4

    Adaptor proteins participate in many signaling pathways from cell surface receptors. Crk protein was the first example of the adaptor protein. We have examined the function of Crk II mutant, Crk II-23. The Crk II-23 mutant contains two amino-acid substitutions in the carboxyl-terminal SH3 domain and is known to inhibit the transformation of NRK cells induced by epidermal growth factor (EGF) and transforming growth factor-beta (TGF-beta). There was no remarkable difference between Crk II and Crk II-23 in EGF-dependent binding to EGF receptor (EGFR). However, in contrast to the wild-type Crk II, the Crk II-23 mutant bound to EGFR in quiescent NIH 3T3 cells. To clarify the difference, both the Crk II and Crk II-23, proteins were expressed in E. coli and examined their binding capacity in vitro. They bound to EGFR from EGF-stimulated NIH 3T3 cells in vitro to a similar extent. Expression of Crk II-23 in NIH 3T3 cells did not affect the binding of bacterially expressed Crk II and Crk II-23 to EGFR. These results suggest that post-translational modification of Crk II-23, such as physical association to cellular proteins, induces binding of Crk II-23 to EGFR in quiescent cells. We also demonstrated that mutation of either the SH2 or the SH3 domain abolished the anti-oncogenic activity of Crk II-23, although both mutants bound to EGFR in the quiescent cells. From these results, it could be concluded that persistent signaling through Crk II-23 bound to EGFR is responsible for the suppression of transformation by EGF and TGF-beta.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Humans; Mutation; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-crk; Signal Transduction; Transforming Growth Factor beta

1997
Transforming growth factor beta1 increases the expressions of high molecular weight tropomyosin isoforms and vinculin and suppresses the transformed phenotypes in human lung carcinoma cells.
    Cancer letters, 1997, Dec-16, Volume: 121, Issue:1

    Exposure of the human lung carcinoma cell line, A549 cells, to transforming growth factor beta1 (TGFbeta1) resulted in an alteration in the morphology from epithelial-like to fibroblastic flat cells. Immunofluorescent study revealed that microfilament organization was partially restored in the TGFbeta1-treated cells. We also investigated the effects of TGFbeta1 on the transformed phenotypes including the anchorage-independent growth and invasive ability of A549 cells and found that the phenotypes were strikingly suppressed in TGFbeta1-treated A549 cells. Accompanying these changes, the levels of the expression of high molecular weight tropomyosin (TM) isoforms, especially TM1 and TM2, were significantly increased in the TGFbeta1-treated cells. The expression level of vinculin was also increased in these cells. These results suggest that the increased tropomyosin and vinculin syntheses may be involved in the suppression of the transformed phenotypes of A549 cells.

    Topics: Actins; Animals; Blotting, Western; Carcinoma; Cell Adhesion; Cell Size; Cell Transformation, Neoplastic; Chlorocebus aethiops; Dose-Response Relationship, Drug; Humans; Lung Neoplasms; Microscopy, Fluorescence; Neoplasm Invasiveness; Transforming Growth Factor beta; Tropomyosin; Tumor Cells, Cultured; Vero Cells; Vinculin

1997
[Growth hormones and oncogenes in mammary adenocarcinomas induced by medroxyprogesterone acetate in BALB/c mice].
    Medicina, 1997, Volume: 57 Suppl 2

    We have studied the involvement of growth factors (GF), their receptors (GF-R) and oncogenes in modulating tumor growth in the medroxyprogesterone acetate (MPA)-induced mammary tumor model in BALB/c mice. We demonstrated the presence of both ligands of the insulin-like growth factor family (IGF-I, IGF-II) and the two types of receptors (IGF-RI, IGF-RII). MPA upregulated IGF-II mRNA and protein levels in hormone-dependent lines (MPA-D). The progression to a hormone-independent phenotype was accompanied by a high constitutive expression of IGF-II and by a significant decrease in IGF-IIR number. An antisense strategy used to evaluate the role of IGF in the MPA-induced growth of epithelial MPA-D cells showed that IGF mediate progestin-induced mammary tumor growth by autocrine/intracrine pathways. We also studied the role of heregulins (HRG), the recently identified ligands for the c-erbB3 and c-erbB4 oncogenes. HRG mRNA expression was restricted to tumors of ductal origin. MPA induced an in vivo up-regulation of HRG expression. Finally, we also found that MPA may be exerting its proliferative effect on MPA-D lines by inhibiting the expression of transforming growth factor beta 1, (TGF-beta 1) and the lack of expression of TGF-beta 1 in hormone-independent tumors may be related to the acquisition of autonomous growth.

    Topics: Adenocarcinoma; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; Growth Substances; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Mammary Neoplasms, Experimental; Medroxyprogesterone Acetate; Mice; Mice, Inbred BALB C; Oncogenes; Receptors, Growth Factor; Transforming Growth Factor beta

1997
Transforming growth factor beta expression and transformation of rat lung epithelial cells by crystalline silica (quartz).
    International journal of cancer, 1996, Mar-01, Volume: 65, Issue:5

    Crystalline silica (quartz) induces silicosis and associated peripheral lung carcinomas in rats. The role and pattern of expression of transforming growth factor (TGF)-beta1/beta2 mRNA transcripts were investigated in the fetal rat lung epithelial cell line FRLE, its neoplastic transformants and derived tumors in athymic nude mice. FRLE cells, treated with 100 microgram/cm2 of quartz in serum-free medium, gave rise to phenotypically altered, tumorigenic cells. Quartz-treated, transformed and tumorigenic cells, subcultured directly (QTT-C1) or after growth in soft agar (QTT-C2), formed tumors in athymic nude mice (QTT-T1). Cells subcultured from the tumors (QTT-T1C) were also tumorigenic in nude mice (QTT-T2). QTT-T1 and QTT-T2 tumors were poorly differentiated carcinomas with variable amounts of extracellular matrix-associated TGF-beta1 and desmoplasia. For comparison, a tumorigenic cell line derived from FRLE cells transformed with a mutated K-ras plasmid (RT-C1) and cells subcultured from a corresponding nude mouse tumor (RT-T1) and designated RT-T1C were used. Whereas TGF-beta1 and TGF-beta2 inhibited the growth of QTT-T1C and FRLE cells in a dose-dependent fashion, RT-T1C cells, containing an activated ras gene, were relatively unaffected. TGF-beta1 and TGF-beta2 mRNAs were expressed at higher levels in QTT-T1C cells than in FRLE and TR-T1C cells, and there was an increase in TGF-beta type II receptor (TGR-betaR) mRNA expression in QTT-T1C and RT-T1C cells compared to FRLE cells. Carcinomas in nude mice derived from QTT and RT cells and silicosis-associated lung carcinomas induced in rats by intra-tracheal quartz did not express either active or latent forms of TGF-beta1 protein on immunohistochemistry. The disparity between TGF-beta1 mRNA and TGF-beta1 protein expression in QTT tumors may be due to post-transcriptional regulation of TGF-beta1.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Female; Gene Expression Regulation, Neoplastic; Male; Mice; Mice, Nude; Microscopy, Electron; Neoplasms, Experimental; Proteolipids; Pulmonary Alveoli; Pulmonary Surfactants; Rats; RNA, Messenger; Silicon Dioxide; Transforming Growth Factor beta

1996
Type II TGF(beta) receptor expression in intestinal cell lines and in the intestinal tract.
    Carcinogenesis, 1996, Volume: 17, Issue:5

    The recent identification and cloning of mammalian transforming growth factor beta (TGFbeta) receptors permits further analysis of the importance of the TGFbeta family in intestinal biology. Expression of the type II TGFbeta receptor was examined in gastrointestinal cell lines and tissues. The 5.5 kb type II mRNA species was detected in poly-(A) mRNA isolated from the rat small bowel and colon. Northern blot analysis of RNA isolated from epithelial and non-epithelial small intestinal cell fractions showed the majority of receptor mRNA localized in the non-epithelial compartment. Immunohistochemical localization in the small intestine and colon supported the RNA findings; that is, expression was greatest in the lamina propria and muscularis. Staining was also detectable in the epithelium, where it was most prominent in the villus tip cells and absent in crypt cells. These findings mirror expression of TGFbeta in the epithelial compartment. The IEC-6, IPEC and RIE-1 cell lines, all of which are non-transformed, were growth inhibited by TGFbeta and expressed type II receptor mRNA and protein. By contrast, the ras-transfected RIE-1, HT-29, Caco-2 and SW-620 transformed lines were not growth inhibited by TGFbeta and all demonstrated a marked reduction in type II TGFbeta receptor mRNA expression and protein abundance by cross-linking. In conclusion, (i) colocalization of both ligand and receptor establishes the existence of potential autocrine and/or paracrine pathways for TGFbeta in the normal intestine and (ii) down-regulation of the type II TGFbeta receptor occurs in association with cellular transformation and may contribute to intestinal carcinogenesis.

    Topics: Animals; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Genes, ras; Intestinal Neoplasms; Intestines; Rats; Receptors, Transforming Growth Factor beta; RNA, Messenger; Transforming Growth Factor beta

1996
TGF-beta1 and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells.
    Genes & development, 1996, Oct-01, Volume: 10, Issue:19

    Metastasis of epithelial tumor cells can be associated with the acquisition of fibroblastoid features and the ability to invade stroma and blood vessels. Using matched in vivo and in vitro culture systems employing fully polarized, mammary epithelial cells, we report here that TGF-beta1 brings about these changes in Ras-transformed cells but not in normal cells. When grown in collagen gels in the absence of TGF-beta, both normal and Ras-transformed mammary epithelial cells form organ-like structures in which the cells maintain their epithelial characteristics. Under these conditions, treatment of normal cells with TGF-beta results in growth arrest. The same treatment renders Ras-transformed epithelial cells fibroblastoid, invasive, and resistant to growth inhibition by TGF-beta. After this epithelial-fibroblastoid conversion, the Ras-transformed cells start to secrete TGF-beta themselves, leading to autocrine maintenance of the invasive phenotype and recruitment of additional cells to become fibroblastoid and invasive. More important, this cooperation of activated Ha-Ras with TGF-beta1 is operative during in vivo tumorigenesis and, as in wound healing processes, is dependent on epithelial-stromal interactions.

    Topics: Animals; Cell Line, Transformed; Cell Polarity; Cell Transformation, Neoplastic; Chick Embryo; Collagen; Epithelial Cells; Fibroblasts; Gels; Genes, ras; Growth Substances; Heart; Mammary Glands, Animal; Mammary Neoplasms, Experimental; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Invasiveness; Neoplasms, Glandular and Epithelial; Oncogene Protein p21(ras); Receptors, Transforming Growth Factor beta; RNA, Messenger; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation

1996
The absence of p21Cip1/WAF1 alters keratinocyte growth and differentiation and promotes ras-tumor progression.
    Genes & development, 1996, Dec-01, Volume: 10, Issue:23

    p21Cip1/WAF1 was the first cyclin-dependent kinase (CDK) inhibitor to be identified, as a mediator of p53 in DNA damage-induced growth arrest, cell senescence, and direct CDK regulation. p21 may also play an important role in differentiation-associated growth arrest, as its expression is augmented in many terminally differentiating cells. A general involvement of p21 in growth/differentiation control and tumor suppression has been questioned, as mice lacking p21 undergo a normal development, harbor no gross alterations in any of their organs, and exhibit no increase in spontaneous tumor development. However, a significant imbalance between growth and differentiation could be unmasked under conditions where normal homeostatic mechanisms are impaired. We report here that primary keratinocytes derived from p21 knockout mice, transformed with a ras oncogene, and injected subcutaneously into nude mice exhibit a very aggressive tumorigenic behavior, which is not observed with wild-type control keratinocytes nor with keratinocytes with a disruption of the closely related p27 gene. p21 knockout keratinocytes tested under well-defined in vitro conditions show a significantly increased proliferative potential, which is also observed but to a lesser extent with p27 knockout cells. More profound differences were found in the differentiation behavior of p21 versus p27 knockout keratinocytes, with p21 (but not p27) deficiency causing a drastic down-modulation of differentiation markers linked with the late stages of the keratinocyte terminal differentiation program. Thus, our results reveal a so far undetected role of p21 in tumor suppression, demonstrate that this function is specific as it cannot be attributed to the closely related p27 molecule, and point to an essential involvement of p21 in terminal differentiation control, which may account for its role in tumor suppression.

    Topics: Animals; Calcium; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Genes, ras; Keratinocytes; Mice; Mice, Knockout; Neoplasms, Experimental; Transforming Growth Factor beta

1996
Effect of transforming growth factor beta on rat hepatocyte cell lines depends upon the state of tumorigenic progression.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1996, Volume: 7, Issue:4

    The effect of transforming growth factor beta (TGF-beta) on morphology, actin cytoskeleton organization, anchorage-dependent proliferation, anchorage-independent proliferation, and alpha-fetoprotein secretion in a series of related hepatocyte-derived cell lines was measured. We reported previously that TGF-beta 1 partially suppressed the transformed phenotype of a ras- transformed SV40-immortalized hepatocyte cell line designated NR4. In the present study, the CWSV14 cell line, which was derived by transfecting rat hepatocytes with SV40 DNA, was analyzed at low (14LP), mid (14MP), and high (14HP) passage. CWSV14 cells are weakly tumorigenic at low passage and spontaneously transform with in vitro passaging. Tumor cell lines (14T1 and 14T2) derived from hepatocellular carcinomas produced by CWSV14 cells were also analyzed. TGF-beta 1 partially suppressed the transformed phenotype of 14HP, 14T1 and 14T2 cells but had no effect on cell proliferation in these cells. In contrast, TGF-beta 1 induced apoptosis in 14LP and 14MP cells. Studies using both the NR4 and 14T1 cells showed that suppression of the transformed phenotype also could be induced by TGF-beta 2 or TGF-beta 3. We conclude that TGF-beta-induced suppression of the transformed phenotype can be observed in ras-and spontaneously transformed hepatocyte cell lines. It is also apparent that the effect of TGF-beta 1 on hepatocyte cell lines differs depending upon the state of progression toward malignancy; that is, while TGF-beta 1 suppressed the transformed phenotype in highly transformed and tumor cell lines representing late stages in progression, it induced apoptosis in weakly or moderately transformed cell lines representing early stages in progression.

    Topics: Actins; alpha-Fetoproteins; Animals; Apoptosis; Cell Division; Cell Transformation, Neoplastic; DNA; Fluorescence; Liver; Phalloidine; Phenotype; Rats; Time Factors; Transforming Growth Factor beta

1996
[rhTGF-beta 1 induced differentiation of human promonocytic leukemia THP-1 cells].
    Shi yan sheng wu xue bao, 1996, Volume: 29, Issue:4

    With promonocytic leukemia cell line THP-1 cells as an experimental material, the present paper described the proliferation, differentiation and maturation of these cells into m phi-like cells when they were treated with rhTGF-beta 1. Both cell number count and 3H-TdR uptake experiments indicated that rhTGF-beta 1 obviously inhibited the proliferation of THP-1 cells, and the inhibiting effect was related to its concentration. At the same time, the changes in the mode of cell growth and morphology occurred. The cells changed gradually from suspensive into adherent state and formed two groups of cell populations. The number of adherent cells formed was dependent on the concentration and duration of the treatment of rhTGF-beta 1. Therefore, based on the degree of inhibition of cell proliferation and the number of adherent cells with different rhTGF-beta 1 concentrations in a trial experiment, 1.25 ng/ml rhTGF-beta 1 was chosen as the dose in other experiments. From scanning electronmicroscopic observation, it was found that the external morphology of rhTGF-beta 1 treated THP-1 cells gradually transformed into typical macrophage-like cells. Concomitantly, their subcellular organelles also became progressively matured, with primary lysosomes typical for early M phi in 72 h and secondary lysosomes and phagosomes for mature M phi in 120 h of induction, as observed with transmission electron microscope. The ANAE activity, NBT reduction and phagocytosis of differentiated adherent cells were higher than those of control cells and suspensive cells. Specific anti-human TGF-beta-neutralizing mAb could completely block the differentiation of THP-1 cells into M phi-like cells. To sum up, from the results of the studies on cell morphology, growth mode, ultrastructures, phagocytosis, enzyme activation and TGF-beta 1 mAb blocking of induction and differentiation, it is clear that rhTGF-beta 1 can induce THP-1 cells to differentiate and mature into M phi-like cells, with the parallel development of cytoplasmic organoids, phenotype variation and the gaining of phagocytosis activity etc. Concordantly, rhTGF-beta 1 made the M phi-like cells to an activated state as they became matured during the induced differentiation.

    Topics: Cell Transformation, Neoplastic; Humans; Leukemia, Myeloid; Recombinant Proteins; Transforming Growth Factor beta; Tumor Cells, Cultured

1996
TGF-beta in mammary development and neoplasia.
    Journal of mammary gland biology and neoplasia, 1996, Volume: 1, Issue:4

    Topics: Animals; Breast; Breast Neoplasms; Cell Transformation, Neoplastic; Female; Humans; Mammary Glands, Animal; Mammary Neoplasms, Experimental; Transforming Growth Factor beta

1996
Dominant-negative p53 can restore tumorigenicity of L-929 cells in immunocompetent mice.
    International journal of cancer, 1995, Nov-15, Volume: 63, Issue:4

    To study the effect of a transforming allele of the tumor suppressor p53 upon the anti-tumor immune response, antigenic L-929 cells were transfected with the dominant-negative valine135 mutant of murine p53. Several p53val135-expressing transfectants formed non-regressing tumors in immunocompetent hosts. The growth rates of tumorigenic and non-tumorigenic clones were equivalent in vitro in sublethally irradiated C3H/HeN mice and in nude mice. Tumorigenic and non-tumorigenic p53val135-expressing L-929 clones expressed equivalent levels of cell surface class I major histocompatibility complex (MHC) glycoproteins. Immunization with a tumorigenic Lp53val135 clone protected mice from subsequent challenge and primed MHC class I-restricted cytotoxic T-lymphocytic precursors. Secretion of an immunosuppressive cytokine, transforming growth factor beta-1 and sensitivity to tumor necrosis factor-alpha were equivalent from tumorigenic and non-tumorigenic cell lines. These data suggest that expression of a transforming allele of p53 can allow L-929 cells to escape the host immune system.

    Topics: Alleles; Animals; Cell Transformation, Neoplastic; Genes, Dominant; Genes, p53; Histocompatibility Antigens Class I; Immunocompetence; Male; Mice; Mice, Inbred BALB C; Mice, Inbred C3H; Mice, Nude; Skin Neoplasms; T-Lymphocytes, Cytotoxic; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha; Tumor Suppressor Protein p53

1995
Thrombospondin 1 expression in transformed endothelial cells restores a normal phenotype and suppresses their tumorigenesis.
    Proceedings of the National Academy of Sciences of the United States of America, 1995, Jul-18, Volume: 92, Issue:15

    Murine endothelial cells are readily transformed in a single step by the polyomavirus oncogene encoding middle-sized tumor antigen. These cells (bEND.3) form tumors (hemangiomas) in mice which are lethal in newborn animals. The bEND.3 cells rapidly proliferate in culture and express little or no thrombospondin 1 (TS1). To determine the role of TS1 in regulation of endothelial cell phenotype, we stably transfected bEND.3 cells with a human TS1 expression vector. The cells expressing human TS1 were readily identified by their altered morphology and exhibited a slower growth rate and lower saturation density than the parental bEND.3 cells. The TS1-expressing cells also formed aligned cords of cells instead of clumps or cysts in Matrigel. Moreover, while the bEND.3 cells formed large tumors in nude mice within 48 hr, the TS1-expressing cells failed to form tumors even after 1 month. The TS1-transfected cells expressed transforming growth factor beta mRNA and bioactivity at levels similar to those of the parental or vector-transfected bEND.3 cells, indicating that the effects of TS1 expression are not due to the activation of transforming growth factor beta by TS1. TS1 expression resulted in a > 100-fold decrease in net fibrinolytic (urokinase-type plasminogen activator, uPA) activity due to more plasminogen-activator inhibitor 1 and less uPA secretion. TS1 thus appears to be an important regulator of endothelial cell phenotype required for maintaining the quiescent, differentiated state.

    Topics: Animals; Cell Differentiation; Cell Transformation, Neoplastic; Endothelium, Vascular; Gene Expression Regulation, Neoplastic; Hemangioma; Humans; Membrane Glycoproteins; Mice; Neoplasms, Experimental; Phenotype; Plasminogen Activator Inhibitor 1; Recombinant Proteins; Suppression, Genetic; Thrombospondins; Transfection; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator

1995
Progressive loss of sensitivity to growth control by retinoic acid and transforming growth factor-beta at late stages of human papillomavirus type 16-initiated transformation of human keratinocytes.
    Advances in experimental medicine and biology, 1995, Volume: 375

    Retinoids (vitamin A and its natural and synthetic derivatives) have shown potential as chemopreventive agents, and diets poor in vitamin A and/or its precursor beta-carotene have been linked to an increased risk of cancer at several sites including the cervix. Human papillomavirus (HPV) plays an important role in the etiology of cervical cancer. We have developed an in vitro model of cancer progression using human keratinocytes (HKc) immortalized by HPV16 DNA (HKc/HPV16). Although immortal, early passage HKc/HPV16, like normal HKc, require epidermal growth factor (EGF) and bovine pituitary extract (BPE) for proliferation and undergo terminal differentiation in response to serum and calcium. However, following prolonged culture, growth factor independent HKc/HPV16 lines that no longer require EGF and BPE can be selected (HKc/GFI). Further selection of HKc/GFI produces lines that are resistant to serum- and calcium- induced terminal differentiation (HKc/DR). HKc/DR, but not early passage HKc/HPV16, are susceptible to malignant conversion following transfection with viral Harvey ras or Herpes simplex virus type II DNA. We have investigated the sensitivity of low to high passage HKc/HPV16 and HKc/GFI to growth control by all-trans-retinoic acid (RA, an active metabolite of vitamin A). Early passage HKc/HPV16 are very sensitive to growth inhibition by RA, and in these cells RA decreases the expression of the HPV16 oncogenes E6 and E7. However, as the cells progress in culture they lose their sensitivity to RA. Growth inhibition by RA may be mediated through the cytokine transforming growth factor-beta (TGF-beta), a potent inhibitor of epithelial cell proliferation. RA treatment of HKc/HPV16 and HKc/GFI results in a dose-and time-dependent induction (maximal of 3-fold) in secreted levels of TGF-beta. Also, Northern blot analysis of mRNA isolated from HKc/HPV16 demonstrated that RA treatment induced TGF-beta 1 and TGF-beta 2 expression about 3- and 50-fold, respectively. We next studied the effect of TGF-beta 1 and TGF-beta 2 on the proliferation of early to late passage HKc/HPVa6, HKc/GFI and HKc/DR. While early passage HKc/HPV16 were as sensitive as normal HKc to growth inhibition by TGF-beta 1 and TGF-beta 2, the cells became increasingly resistant to TGF-beta during in vitro progression, with the proliferation of HKc/DR being virtually unaffected by TGF-beta 1 or TGF-beta 2 treatment. Overall, loss of growth inhibition by RA parallels loss of TGF-beta sensi

    Topics: Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cell Transformation, Viral; Female; Humans; Keratinocytes; Male; Models, Biological; Papillomaviridae; RNA, Messenger; Transforming Growth Factor beta; Tretinoin; Uterine Cervical Neoplasms

1995
Transformation-restoring factor: a low molecular weight secreted factor required for anchorage-independent growth of oncogene-resistant mutant cell lines.
    Oncogene, 1995, Apr-06, Volume: 10, Issue:7

    We have previously described two independent mutant rat fibroblast cell lines that fail to form colonies in soft agar when infected with a v-H-ras-expressing retrovirus, yet still undergo transformation-related morphological alterations in response to this oncogene. We report here that conditioned medium (CM) from non-transformed rat fibroblasts contains an activity that specifically corrects this defect in the mutant cell lines, rendering them capable of anchorage-independent growth in response to ras. The major activity in CM, designated transformation-restoring factor (TRF), is approximately 1300 molecular weight, lipid insoluble, and heat, protease, acid and base stable. Latent activity, distinct from TRF, is also present in CM; several lines of evidence indicate that transforming growth factor (TGF) beta is responsible for this activity. TRF, however, cannot substitute for TGF beta in the phenotypic transformation of NRK cells. TRF activity is decreased in CM of control cells transformed by ras and this response to ras is retained by the mutant cell lines. We propose that whereas wild-type cells transformed by ras may constitutively activate a TRF-regulated pathway, thus becoming independent of TRF for growth in soft agar, these mutants have acquired dependence on an exogenous supply of TRF for this aspect of the transformed phenotype. Cellular activities regulated, directly or indirectly, by TRF may be effectors of the anchorage-independent growth property that is a hallmark of transformed rodent fibroblasts.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Gene Expression; Genes, ras; Growth Substances; In Vitro Techniques; Molecular Weight; Oncogenes; Rats; RNA, Messenger; Transforming Growth Factor beta

1995
Dissociation of mitogenesis and transforming activity by C-terminal truncation of the insulin-like growth factor-I receptor.
    Experimental cell research, 1995, Volume: 218, Issue:1

    We have investigated the mitogenic and transforming ability of an IGF-I receptor with a 108-amino-acid C-terminal truncation in R- cells, which are 3T3-like cells derived from mouse embryos in which the IGF-I receptor genes have been disrupted by targeted homologous recombination. R- cells stably transfected with expression plasmids encoding either a wild-type or a truncated human IGF-I receptor were capable of growing in serum-free medium supplemented solely with IGF-I. This response was observed over a wide range of receptor levels. R- cells overexpressing the wild-type IGF-I receptor also formed colonies in soft agar, and colony formation was augmented by coexpression of the SV40 large T antigen. However, all the examined clones of R- cells expressing the truncated IGF-I receptor exhibited a dramatically impaired ability to grow in soft agar, even in the presence of the T antigen. The inability to form colonies in soft agar was not due to a quantitative impairment of signal transduction, because: (1) SV40-transformed cells with a physiological level of the wild-type IGF-I receptor did not respond to IGF-I with cell proliferation, but grew in soft agar; (2) R- cells stably transfected with both a truncated receptor and T antigen, on the contrary, responded with mitogenesis to IGF-I but could not form colonies in soft agar; (3) some clones with the truncated receptor expressed levels of receptor roughly 100-fold the level of wild-type cells; and (4) several parameters of IGF-I receptor signal transduction were not impaired in cells stably transfected with a truncated receptor. Furthermore, overexpression of an activated ras in cells with the truncated IGF-IR did not restore their ability to proliferate under anchorage-independent conditions. We conclude that the 108 amino acids of the IGF-I receptor are not essential for a mitogenic response to IGF-I, but are required for transformation (as assessed by the ability to grow in soft agar), indicating that these two functions can be dissociated at an intramolecular level. Moreover, although ras (activated) certainly plays a role in transformation, the transforming activity of the IGF-IR also requires signaling elements that are ras-dependent.

    Topics: 3T3 Cells; Animals; Antigens, Polyomavirus Transforming; Cell Division; Cell Transformation, Neoplastic; Clone Cells; Culture Media, Serum-Free; Humans; Insulin Receptor Substrate Proteins; Mice; Phosphatidylinositol 3-Kinases; Phosphoproteins; Phosphotransferases (Alcohol Group Acceptor); ras Proteins; Receptor, IGF Type 1; Recombinant Proteins; Sequence Deletion; Simian virus 40; Transfection; Transforming Growth Factor beta

1995
Malignant transformation by H-ras results in aberrant regulation of ribonucleotide reductase gene expression by transforming growth factor-beta 1.
    Journal of cellular biochemistry, 1995, Volume: 57, Issue:3

    Ribonucleotide reductase is a key rate-limiting and regulatory step in DNA synthesis and plays a crucial role in the coordination of DNA synthesis, DNA repair, and cell proliferation. The present study demonstrates a link between alterations in TGF-beta 1 regulation during malignant conversion and the expression of ribonucleotide reductase. H-ras-transformed mouse 10T1/2 cell lines exhibiting malignant potential were examined for possible TGF-beta 1-mediated alterations in ribonucleotide reductase expression. Selective induction of ribonucleotide reductase gene expression occurred, since only H-ras-transformed highly metastatic cells exhibited marked elevations in ribonucleotide reductase expression, whereas nontransformed normal 10T1/2 cells were unaffected by TGF-beta 1 treatment. These changes occurred without any detectable modifications in DNA synthesis rates, suggesting that these changes were regulated by a novel mechanism independent of the S-phase of the cell cycle. Furthermore, this TGF-beta 1-mediated regulation of ribonucleotide reductase expression was shown to occur through an autocrine mechanism. TGF-beta 1-modulated regulation of ribonucleotide reductase expression requires de novo protein synthesis and involves, at least in part, transcriptional and post-transcriptional events. Furthermore, evidence is presented to suggest a possible role for protein kinase C-mediated events, protein phosphatases, and G-protein-coupled events in the TGF-beta 1-mediated regulation of ribonucleotide reductase expression in H-ras-transformed malignant cells. TGF-beta 1 regulation of ribonucleotide reductase in highly malignant cells appears to be complex and multifaceted and constitutes an integral part of an altered growth regulatory program.

    Topics: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Fibrosarcoma; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Genes, ras; Mice; Ribonucleotide Reductases; S Phase; Transfection; Transforming Growth Factor beta

1995
Catechol interferes with TGF-beta-induced elimination of transformed cells by normal cells: implications for the survival of transformed cells during carcinogenesis.
    International journal of cancer, 1995, Feb-08, Volume: 60, Issue:4

    We have recently shown that TGF-beta-treated normal fibroblasts are able to induce apoptosis of transformed fibroblasts, leading to their elimination. Here we describe a test system that allows the quantitative analysis of the elimination of G418-resistant transformed cells by TGF-beta-treated normal cells. This assay system was used to screen for substances that interfere with the elimination of transformed cells. Catechol and hydroquinone, but not resorcinol, were found to represent potent antagonists of TGF-beta-induced elimination of transformed cells by normal cells. Protection of transformed cells from negative effects derived from their cellular environment defines a hitherto unrecognized crucial mechanism for the survival of transformed cells. The protective effect of catechol as seen in this experimental system may act in concert with its co-carcinogenic and promoting activities during carcinogenesis.

    Topics: Animals; Antioxidants; Apoptosis; Catechols; Cell Line, Transformed; Cell Transformation, Neoplastic; Cocarcinogenesis; Contact Inhibition; Fibroblasts; Hydroquinones; Mice; Mice, Inbred C3H; Resorcinols; Transforming Growth Factor beta; Tumor Cells, Cultured

1995
Interaction between p53 and TGF beta 1 in control of epithelial cell proliferation.
    Oncogene, 1995, Jan-19, Volume: 10, Issue:2

    Although loss of sensitivity to transforming growth factor beta (TGF beta) may be a key step in the escape of epithelial tumours from normal growth control, the intracellular signals determining responsiveness remain controversial, particularly the role of p53. We have investigated this question using thyroid epithelial lines as a model. We analysed (i) human thyroid cancer cell lines having either wild-type (wt) or mutant p53; (ii) rat thyroid lines derived by spontaneous immortalisation following introduction of mutant H-ras, which exhibit high levels of wt p53 but loss of p53-mediated cell-cycle control. Loss of response to TGF beta 1 was found in all human lines bearing mutant p53, and in the majority of the functionally equivalent rat lines, consistent with a role of wt p53 in mediating response. However, introduction of a dominant negative p53 mutant into TGF beta 1 responsive human lines containing wt p53 did not reduce responsiveness, demonstrating that p53 function is not necessary for TGF beta 1 response. On the other hand, expression of a temperature-sensitive (ts) p53 gene in a partially-responsive rat line demonstrated a highly significant modulation of TGF beta response, which fell from 65% inhibition of 3H-thymidine labelling index at 32.5 degrees C (wt p53 conformation) to only 14% at 37.5 degrees C (mutant conformation). The results suggest that p53 and TGF beta generate separate but interacting inhibitory signals, i.e. that p53 modulates but does not mediate TGF beta response. This conclusion explains previous conflicting data and is consistent with current models of cell cycle control by multiple inhibitors of cyclin-dependent kinases.

    Topics: Animals; Base Sequence; Cell Division; Cell Transformation, Neoplastic; Genes, ras; Humans; Models, Biological; Molecular Sequence Data; Mutation; Rats; Temperature; Thyroid Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Protein p53

1995
Regulation of transforming growth factor beta receptors in H-ras oncogene-transformed rat intestinal epithelial cells.
    Cancer research, 1995, Dec-15, Volume: 55, Issue:24

    We have reported that over expression of the H-ras oncogene causes resistance to growth inhibition by transforming growth factor beta 1 (TGF-beta 1) and a time-dependant switch of type II to type I TGF-beta receptor expression in the rat intestinal epithelial cell line IEC-18 (J. Filmus, J. Zhao, and R. N. Buick, Oncogene, 7: 521-526, 1992). Here, we investigate the possible mechanisms involved in H-ras-mediated regulation of TGF-beta receptors in an IEC-18 cell clone expressing H-ras, conditional on the activity of a dexamethasone-sensitive promoter. The switch from type II to type I receptor expression in response to H-ras expression has a requirement for de novo RNA synthesis. In addition, accumulation of TGF-beta receptor type II mRNA is approximately 5-fold lower in ras-expressing cells compared to control cells. Nuclear run-on experiments suggest that the down-regulation of type II receptor mRNA by H-ras oncogene is based, at least in part, on reduced transcription. We have also analyzed the consequences of H-ras expression on the properties of the TGF-beta receptors. Type I and II in IEC-18 cells and type I receptors in ras-transformed cells have similar characteristics in terms of binding affinities for TGF-beta 1 (or TGF-beta 2) turnover rates and glycosylation states. Notably, the type I receptors in ras-transformed cells are not capable of ligand-induced internalization. Although H-ras expression in IEC-18 cells causes resistance to TGF-beta-mediated growth inhibition, the cells remain responsive to TGF-beta 1 stimulation of fibronectin expression. These results are discussed in the context of the knowledge of TGF-beta receptor complexity and signal transduction, and with reference to the potential role for loss of TGF-beta-mediated negative growth regulation in malignant transformation.

    Topics: Animals; Cell Division; Cell Nucleus; Cell Transformation, Neoplastic; Down-Regulation; Fibronectins; Genes, ras; Glycosylation; Intestinal Mucosa; Ligands; Protein Binding; Proto-Oncogene Proteins p21(ras); Rats; Receptors, Transforming Growth Factor beta; RNA, Neoplasm; Transforming Growth Factor beta; Tumor Cells, Cultured

1995
Effects of IL-11 on the growth of intestinal epithelial cells in vitro.
    Cell proliferation, 1995, Volume: 28, Issue:11

    The network of interacting factors that control proliferation in the intestinal epithelium is largely unknown. Recently, IL-11 was found to protect animals from lethal doses of cytotoxic agents. Part of this protective action was ascribed to a reduced level of damage in the intestinal epithelium. Whether this was due to a direct effect on epithelial cell cycle progression was unclear. We have addressed this question in vitro and found that IL-11 reversibly inhibited proliferation in untransformed small intestinal IEC18 cells. However, IL-11 did not inhibit transformed SW620 or HT29 colonic cell lines. IL-6 behaved in a similar manner to IL-11. Thus, these results suggest that IL-11 may be an ideal therapy adjuvant, protecting normal cells and further, these results suggest that IL-11 may be involved in the normal growth controls in the intestinal epithelium. The inhibitory response evoked by IL-11 is lost during carcinogenic transformation.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Colon; Dose-Response Relationship, Drug; Epithelial Cells; Epithelium; Interleukin-11; Interleukin-6; Intestines; Rats; Transforming Growth Factor beta

1995
Transforming growth factor-beta isoform autocrine production by cultured ras-transfected and tumour-derived human keratinocytes.
    Biochemical Society transactions, 1995, Volume: 23, Issue:3

    Topics: Cell Differentiation; Cell Line; Cell Transformation, Neoplastic; Genes, ras; Humans; Keratinocytes; Mutation; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

1995
Regulation of immunoreactive insulin-like growth factor binding protein-6 in normal and transformed human fibroblasts.
    The Journal of biological chemistry, 1994, Apr-15, Volume: 269, Issue:15

    Insulin-like growth factor-binding proteins (IGFBPs) have been shown to both potentiate and inhibit IGF bioactivity in vitro; thus, changes to the type or amount of IGFBPs present in the cellular environment will ultimately affect insulin-like growth factor action. In this study, we have investigated the production of immunoreactive IGFBP-6 by normal human fibroblasts (NHF) and an SV-40-transformed human fibroblast line (AG2804). When analyzed by SDS-polyacrylamide gel electrophoresis and immunoblotting, IGFBP-6 appeared as a doublet of 32-34-kDa in conditioned medium of both cell lines, with the lower molecular mass band predominating in the NHF cell line. Measured by a specific radioimmunoassay, serum-free NHF, and AG2804 cultures secreted IGFBP-6 at 1.44 +/- 0.09 and 1.23 +/- 0.08 ng/10(4) cells (mean +/- S.E.), respectively. Despite a relatively weak IGFBP-6 signal by ligand blot compared with IGFBP-3, the two proteins were secreted in similar molar concentrations by NHF. Retinoic acid increased IGFBP-6 by 3-fold in NHF and AG2804-conditioned media, maximal at approximately 100 nM retinoic acid. In contrast, IGFBP-6 production was inhibited by transforming growth factor-beta 1 and agents that increase intracellular cAMP concentrations, including dibutyryl cAMP, forskolin, isobutylmethylxanthine, and cholera toxin. This study indicates that IGFBP-6 has a pattern of regulation unique among the IGFBPs, supporting the concept of specific roles for each binding protein in regulating cell growth and metabolism.

    Topics: 1-Methyl-3-isobutylxanthine; Bucladesine; Carrier Proteins; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Cholera Toxin; Colforsin; Culture Media, Conditioned; Cycloheximide; Electrophoresis, Polyacrylamide Gel; Fibroblasts; Humans; Insulin-Like Growth Factor Binding Protein 6; Insulin-Like Growth Factor II; Kinetics; Male; Molecular Weight; Recombinant Proteins; Simian virus 40; Skin; Transforming Growth Factor beta; Tretinoin; Virulence Factors, Bordetella

1994
[The effects of recombinant human transforming growth factor beta on inducing differentiation of fresh leukemia cells in acute monocytic leukemia M5].
    Zhonghua nei ke za zhi, 1994, Volume: 33, Issue:5

    By using fresh leukemia cells from 5 cases of acute monocytic leukemia M5 as in vitro model, we investigated the effects of recombinant human transforming growth factor beta 1 (rhTGF-beta 1) on differentiation induction of fresh leukemia cells. The results indicated that after 6 days of induction with TGF-beta 1 in a concentration of 10 ng/ml, leukemia cells in 5 AML-M5 patients differentiated obviously to maturation. The proportion of monoblasts and premonocytes was reduced, while that of mature mononuclear cells elevated. Following administration of TGF-beta 1, alpha-nonspecific esterase (alpha-NSE), whose expression could be inhibited by sodium fluoride, remained positive and peroxidase (POX) was shown to be weakly positive. These results demonstrated that TGF-beta 1 may induce in vitro differentiation of fresh leukaemia cells, but the reactions to TGF-beta 1 may vary in different cases.

    Topics: Adult; Aged; Cell Transformation, Neoplastic; Female; Humans; Leukemia, Monocytic, Acute; Male; Middle Aged; Recombinant Proteins; Transforming Growth Factor beta

1994
Characterization of the TGF beta 1-inducible hic-5 gene that encodes a putative novel zinc finger protein and its possible involvement in cellular senescence.
    The Journal of biological chemistry, 1994, Oct-28, Volume: 269, Issue:43

    Transforming growth factor (TGF) beta 1 is a potent cytokine that inhibits the growth of several types of cells. Our earlier study suggested that the mouse osteoblastic cell line, MC3T3-E1, was sensitive to growth inhibition by TGF beta 1 and that this effect was partly mediated by H2O2. To identify the molecules that participate in the negative regulation of growth by these stimuli, we carried out differential screening of cDNA libraries and isolated a set of genes induced by TGF beta 1. Among the clones isolated, one originally named tsc-5 was found to be induced by H2O2 as well as TGF beta 1. Analysis of this cDNA renamed hic (hydrogen peroxide-inducible clone)-5 suggested that Hic-5 protein has four LIM motifs, each of which contained two (or one) putative zinc fingers. The expression of hic-5 mRNA was repressed in Ki-ras-transformed mouse fibroblasts and in several cell lines established from human tumor. On the other hand, its expression was augmented in the in vitro senescent process of human diploid fibroblasts. Among the mouse organs examined, hic-5 was highly expressed in the lung and spleen. Finally, a colony formation assay using an hic-5 expression vector driven by the cytomegalovirus promoter suggested that hic-5 overexpression had a cytostatic effect on cellular growth, depending upon the cell type. Although the relationship between hic-5 function and the signal transduction pathway of TGF beta 1 remains unresolved, these results implied that hic-5 has some role in the growth-inhibitory pathway associated with in vitro senescence, and that down-regulation of hic-5 contributes to tumorigenesis.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Base Sequence; Cell Transformation, Neoplastic; Cellular Senescence; Cloning, Molecular; Cytoskeletal Proteins; DNA-Binding Proteins; DNA, Complementary; Fibroblasts; Gene Expression Regulation; Genes, ras; Humans; Hydrogen Peroxide; Intracellular Signaling Peptides and Proteins; LIM Domain Proteins; Mice; Molecular Sequence Data; Osteoblasts; Protein Biosynthesis; Selection, Genetic; Sequence Homology, Amino Acid; Signal Transduction; Tissue Distribution; Transforming Growth Factor beta; Tumor Cells, Cultured; Zinc Fingers

1994
Transforming growth factor beta 1 promotes spontaneous transformation of cultured rat liver epithelial cells.
    Cancer research, 1994, Dec-01, Volume: 54, Issue:23

    The neoplastic transformation of cultured rat liver epithelial cells by various means has consistently been associated with the development of resistance to the mito-inhibitory effect of transforming growth factor beta (TGF-beta), suggesting that such phenotype plays a mechanistic role during the transformation of these cells. We have studied the induction of the "TGF-beta-resistant" phenotype in a clonal strain of early passage WB-F344 normal cultured rat liver epithelial cells, the proliferation of which was markedly inhibited by TGF-beta. The control WB cells in continuous culture slowly developed TGF-beta resistance. However, when the same cells were exposed to step-wise increases of TGF-beta concentration in their culture medium, the development of TGF-beta resistance was accelerated. Cells which had been grown in medium containing 1 ng/ml TGF-beta developed colony-forming capacity in soft agar containing epidermal growth factor. Cells which were grown in media containing 5 and 10 ng/ml TGF-beta demonstrated a low level of colony-forming efficiency in soft agar medium without added epidermal growth factor and tumorigenicity in isogeneic rats. These TGF-beta-resistant cells also exhibited progressively increasing levels of expression of the c-fos and and myc mRNA, and increased resistance to the cytotoxicity of Adriamycin and melphalan. The latter phenomenon was accompanied by an increase in the mdr-1 mRNA expression, cellular glutathione level, and glutathione S-transferase activity. The results suggest that chronic exposure to high concentration of TGF-beta promotes the spontaneous neoplastic transformation of cultured rat liver epithelial cells, and that this process may represent one of the mechanisms of cellular adaptation for induction of the multidrug-resistant phenotype during the carcinogenesis of epithelial cells.

    Topics: Animals; ATP Binding Cassette Transporter, Subfamily B, Member 1; Cell Transformation, Neoplastic; Cells, Cultured; Drug Resistance, Multiple; Epithelium; Liver; Phenotype; Rats; Rats, Inbred F344; RNA, Messenger; Transforming Growth Factor beta

1994
Transforming growth factor-beta 1 induces growth inhibition of a human medullary thyroid carcinoma cell line despite an increase in steady state c-myc messenger ribonucleic acid levels.
    Endocrinology, 1994, Volume: 135, Issue:5

    Medullary thyroid cancer (MTC) is an endocrine tumor of the thyroid C-cells which provides an important experimental model for studies of tumor differentiation and progression. We investigated the effects of transforming growth factor-beta 1 (TGF beta 1) on the growth and functional characteristics of a human medullary thyroid carcinoma cell line (TT). Because the c-myc protooncogene may play an important role in the growth inhibition induced by TGF beta 1, we also assessed steady state c-myc messenger RNA (mRNA) levels in these cells. A 6-day exposure of TT cells to TGF beta 1 resulted in a dose-dependent inhibition of cell proliferation. In addition, TGF beta 1 exposure led to a 3-fold increase in nonadherent floating TT cells in the culture supernatants. The floating cells exhibited ultrastructural features of dying or apoptotic cells, including chromatin condensation, cytoplasmic and nuclear vesicularization, and DNA degradation with evidence of internucleosomal DNA "laddering." Despite inhibition of cell proliferation, steady state c-myc mRNA levels were 3.6 +/- 0.6-fold higher in cells exposed to TGF beta 1 compared to those in control cells (P < 0.001). Exposure of cells to a 15-base antisense c-myc oligonucleotide (10 microM) resulted in an attenuation of the TGF beta 1-induced growth inhibition and induction of cell death. TGF beta 1 also resulted in an approximately 3-fold decrease in steady state calcitonin and calcitonin gene-related peptide mRNA levels. Finally, using a sensitive bioassay for TGF beta, TT cells were shown to produce and activate significant amounts of TGF beta, particularly under conditions of serum deprivation. Our data thus indicate that TGF beta 1 has multiple effects on TT cell growth and function. It induces growth inhibition in the presence of an increase in steady state mRNA levels of the c-myc protooncogene, which is usually associated with cell proliferation. In addition, TGF beta 1 accelerates apoptosis in TT cells.

    Topics: Base Sequence; Blotting, Northern; Carcinoma, Medullary; Cell Division; Cell Transformation, Neoplastic; Dose-Response Relationship, Drug; Humans; Microscopy, Electron; Molecular Sequence Data; Proto-Oncogene Proteins c-myc; RNA, Messenger; Thyroid Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured

1994
Growth suppression of transformed BALB/c 3T3 cells by transforming growth factor beta 1 occurs only in the presence of their normal counterparts.
    Carcinogenesis, 1994, Volume: 15, Issue:6

    Transforming growth factor beta 1 (TGF-beta 1) enhances the yield of transformed foci of BALB/c 3T3 cells, but the continuous presence of TGF-beta 1 after foci formation inhibits the growth of transformed foci. The focus-forming ability of Ha-ras-, v-src- and PyMT-transformed cells growing on a monolayer of non-transformed cells was completely suppressed by TGF-beta 1, whereas growth of the transformed cells was little inhibited by TGF-beta 1 in the absence of their normal counterparts. The inhibition by TGF-beta 1 of focus formation by transformed BALB/c 3T3 cells on a normal cell monolayer remained when TGF-beta 1 was removed from the culture medium after 2 weeks. However, the transformed cells were not killed, since they grew in culture conditions under which only transformed cells are able to grow (soft agar). These results suggest that TGF-beta 1 suppresses growth of transformed cells in the presence of normal cells. Furthermore, when non-transformed cells were treated with TGF-beta 1 before co-culture with Ha-ras-transformed cells, formation of transformed foci was inhibited. When normal and transformed cells were cultured in the same dish but separated physically, focus formation was still inhibited. On the other hand, TGF-beta 1 enhanced the growth and changed the morphology of non-transformed cells only in the presence of transformed counterparts. The growth inhibitory effect of TGF-beta 1 on transformed cells and its growth stimulatory effect on non-transformed cells in co-culture conditions suggest the induction of reciprocal paracrine growth regulatory factors. As TGF-beta 1 inhibits the growth of transformed BALB/c 3T3 cells only in the presence of their normal counterparts, a paracrine negative growth control mechanism appears to be operating.

    Topics: 3T3 Cells; Animals; Cell Communication; Cell Death; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Genes, ras; Mice; Mice, Inbred BALB C; Transforming Growth Factor beta

1994
Growth factor responsiveness and alterations in growth factor homeostasis in Syrian hamster embryo cells during in vitro transformation.
    Carcinogenesis, 1994, Volume: 15, Issue:6

    Syrian hamster embryo (SHE) cells were investigated for their growth factor responsiveness as well as changes in growth factor homeostasis, including alterations in autocrine growth factor production and growth factor responsiveness, during in vitro transformation. For wild-type SHE cells, fetal bovine serum (FBS), epidermal growth factor (EGF) family members, platelet derived growth factor (PDGF) family members, fibroblast growth factor family members, interleukin-4, interleukin-9, oncostatin M, hepatocyte growth factor, erythropoietin and pituitary extract were found to be mitogenic. SHE cell mitogenesis was inhibited in response to transforming growth factor beta (TGF-beta) family members, interleukin-1 alpha, interleukin-1 beta and nerve growth factor. Additional experiments were conducted to study alterations in growth factor responsiveness to three SHE cell mitogens (FBS, EGF and PDGF) and one inhibitor of mitogenesis (TGF-beta) during SHE cell in vitro transformation. Alterations in either EGF, PDGF or TGF-beta responsiveness were observed in 7/8 SHE transformed lineages during the stepwise transformation process. Finally, 6/8 lineages underwent alterations which resulted in the production of autocrine growth factors during the transformation process. These results indicate that multiple alterations in growth factor homeostasis occur during the in vitro transformation process.

    Topics: Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Cricetinae; Epidermal Growth Factor; Growth Substances; Homeostasis; Mesocricetus; Platelet-Derived Growth Factor; Transforming Growth Factor alpha; Transforming Growth Factor beta

1994
beta ig-h3: a transforming growth factor-beta-responsive gene encoding a secreted protein that inhibits cell attachment in vitro and suppresses the growth of CHO cells in nude mice.
    DNA and cell biology, 1994, Volume: 13, Issue:6

    beta ig-h3 is a novel gene first discovered by differential screening of a cDNA library made from A549 human lung adenocarcinoma cells treated with transforming growth factor-beta 1 (TGF-beta 1). It encodes a 683-amino-acid protein containing a secretory signal sequence and four homologous internal domains. Here we show that treatment of several types of cells, including human melanoma cells, human mammary epithelial cells, human keratinocytes, and human fibroblasts, with TGF-beta resulted in a significant increase in beta ig-h3 RNA. A portion of the beta ig-h3 coding sequence was expressed in bacteria, and antisera against the bacterially produced protein was raised in rabbits. This antisera was used to demonstrate that several cell lines secreted a 68-kD beta IG-H3 protein after treatment with TGF-beta. Transfection of beta IG-H3 expression plasmids into Chinese hamster ovary (CHO) cells led to a marked decrease in the ability of these cells to form tumors in nude mice. The beta IG-H3 protein was purified from media conditioned by recombinant CHO cells, characterized by immunoblotting and protein sequencing and shown to function in an anti-adhesion assay in that it inhibited the attachment of A549, HeLa, and WI-38 cells to plastic in serum-free media. Sequencing of cDNA clones encoding murine beta ig-H3 indicated 90.6% conservation at the amino acid level between the murine and human proteins. Finally, the beta ig-h3 gene was localized to human chromosome 5q31, a region frequently deleted in preleukemic myelodysplasia and leukemia. The corresponding mouse beta ig-h3 gene was mapped to mouse chromosome 13 region B to C1, which confirms a region of conservation on human chromosome 5 and mouse chromosome 13. We suggest that this protein be named p68 beta ig-h3.

    Topics: Amino Acid Sequence; Animals; Base Sequence; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; CHO Cells; Chromosome Mapping; Chromosomes, Human, Pair 5; Cloning, Molecular; Cricetinae; DNA; Extracellular Matrix Proteins; Humans; Mice; Mice, Nude; Molecular Sequence Data; Neoplasm Proteins; Sequence Homology, Amino Acid; Transforming Growth Factor beta; Tumor Cells, Cultured

1994
Transforming growth factor beta 1 suppresses transformation in hepatocytes by regulating alpha 1 beta 1 integrin expression.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1994, Volume: 5, Issue:5

    We previously reported that (a) treatment of the ras-transformed hepatocyte cell line NR4 with transforming growth factor (TGF) beta 1 suppresses many characteristics associated with the transformed phenotype including altered morphology, actin cytoskeleton reorganization, and anchorage-independent growth such that the cells more closely resemble the immortalized CWSV1 parent cell line; (b) transformed NR4 cells expressed significantly less alpha 1 integrin RNA than the immortalized CWSV1 cells; and (c) TGF-beta 1 treatment of NR4 cells stimulated the expression of alpha 1 and beta 1 integrin RNAs. In this report, the role of the alpha 1 beta 1 integrin in TGF-beta 1-mediated suppression of the ras-transformed phenotype was investigated. We determined that (a) the cell surface integrin that increased in response to TGF-beta 1 treatment of NR4 cells was alpha 1 integrin; (b) TGF-beta 1 altered the ability of NR4 cells to attach to collagen and laminin, the extracellular matrix components that interact with the alpha 1 beta 1 integrin receptor; (c) TGF-beta 1 treatment resulted in relocalization of the alpha 1 integrin on the NR4 cell surface; and (d) TGF-beta 1-mediated inhibition of anchorage-independent growth was blocked by the presence of alpha 1 integrin antibody. A cell line that overexpresses alpha 1 integrin was derived from NR4 cells; characterization of these cells indicated that they continued to express H-ras RNA but were less transformed than the parent NR4 cells. Specifically, they had an altered morphology, an organized actin cytoskeleton, and reduced ability to demonstrate anchorage-independent growth.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Animals; Cell Adhesion; Cell Line, Transformed; Cell Membrane; Cell Transformation, Neoplastic; Collagen; Cytoskeleton; Gene Expression; Genes, ras; Integrin alpha1beta1; Integrins; Laminin; Liver; Phenotype; Rats; RNA, Messenger; Transforming Growth Factor beta

1994
Regulation of transformation frequency by exogenous and endogenous growth factors in rat tracheal epithelial cells.
    Carcinogenesis, 1994, Volume: 15, Issue:3

    The purpose of our studies was to re-evaluate the rat tracheal epithelial (RTE) transformation system and to identify critical variables that affect the development of enhanced growth variants (EGV). The enhanced growth variant colony, which is a preneoplastic cell variant, is the quantifiable transformation endpoint in RTE cultures. Using a standard protocol the frequency of EGV colony formation was shown to be inversely related to the number of clonogenic cells (CFU) seeded per dish in control cultures as well as in cultures treated with the transforming agent 6-nitrochrysene (6-NC). Experiments showed that the major mechanisms that underlie the CFU density-dependent inhibition of EGV colony formation are depletion of growth factors from and accumulation of autocrine TGF-beta in the media. Thus the cells themselves are creating the selection environment, which allows only the EGVs to survive. The effects of agents such as 6-NC, which increase the frequency of EGV colony formation, are to induce a cellular phenotype that is less susceptible to the selection environment. We showed that TGF-beta-neutralizing antibodies added to the selection media significantly increased EGV colony formation in control cultures but not in 6-NC-exposed cultures. In addition we demonstrated that the development of EGV colonies is much less susceptible to inhibition by (exogenous) TGF-beta in 6-NC-exposed than in control cultures. Thus spontaneous and 6-NC EGV colony formation are distinguishable based on TGF-beta sensitivity. To conduct quantitative cell transformation experiments with RTE cells it is essential that the number of surviving CFU per dish is the same in control and treated cultures. Under the conditions used in the studies described here, 350-500 CFU per culture was found to be the optimum CFU density. Besides 6-NC, agents that have been shown to increase EGV colony frequency under conditions similar to those described here are nitrosamines, NNK, nickel compounds and X-rays.

    Topics: Animals; Cell Count; Cell Transformation, Neoplastic; Chrysenes; Culture Media, Serum-Free; Growth Substances; Male; Precancerous Conditions; Rats; Rats, Inbred F344; Tracheal Neoplasms; Transforming Growth Factor beta; Tumor Stem Cell Assay

1994
Comparison of normal and tumorigenic endothelial cells: differences in thrombospondin production and responses to transforming growth factor-beta.
    Journal of cell science, 1994, Volume: 107 ( Pt 1)

    Cultured endothelial cells constitutively synthesize significant levels of thrombospondin, an extracellular matrix-associated protein with reported anti-anti-angiogenic properties. However, two murine endothelial cell lines, bEND.3 and Py-4-1, which have been immortalized with polyoma T oncogenes and which generate vascular malformations in vivo, produce little or no thrombospondin though bEND.3 (but not Py-4-1) growth is inhibited by the addition of exogenous thrombospondin. In addition, Py-4-1 cells are not growth-inhibited by transforming growth factor-beta, a potent endothelial inhibitor. These results indicate that these two cell lines may be useful tools in understanding the role and mechanism of action of thrombospondin and transforming growth factor-beta in endothelial cell biology. A role for thrombospondin in vascular development is further suggested by the observation of significant differences in the levels of thrombospondin mRNA and protein between capillary and aortic endothelial cells. Transforming growth factor-beta-1 treatment of normal endothelial cells increases steady-state levels of thrombospondin mRNA and protein and results in extensive deposition of thrombospondin into the extracellular matrix. In contrast, transforming growth factor-beta-1 has little effect on thrombospondin levels in the tumorigenic endothelial cell lines. In view of our earlier finding that contact between endothelial cells and mural cells generates activated transforming growth factor-beta-1, and the fact that thrombospondin is present in a fibrillar network around vascular structures in vitro, we speculate that modulation of thrombospondin production and distribution by transforming growth factor-beta may be a physiological process to enjoin stabilization of vessels and cessation of vessel growth.

    Topics: Animals; Antigens, Polyomavirus Transforming; Aorta; Blotting, Northern; Brain; Capillaries; Cattle; Cell Adhesion Molecules; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Endothelium, Vascular; Humans; Immunohistochemistry; Membrane Glycoproteins; Mice; Oncogenes; Rats; Recombinant Proteins; RNA, Messenger; Thrombospondins; Transforming Growth Factor beta

1994
Ras transformation alters the composition of extracellular matrix proteoglycans in rat fibroblasts.
    Biochemical and biophysical research communications, 1994, Apr-29, Volume: 200, Issue:2

    ts371 KiMuSV-transformed rat kidney fibroblasts growing at the permissive temperature to allow the expression of a transforming p21 ras protein secrete a different pattern of chondroitin/dermatan sulfate proteoglycans than when growing at the non-permissive temperature. Transformed cells produce less versican and larger amounts of biglycan and decorin than their normal counterpart, and they do not secrete 35S-labeled PG-100. Western blot experiments showed the presence of PG-100 epitope, suggesting that the apparent effect of the oncoprotein is due to a reduced sulfation of the molecule. Transformed and non-transformed cells are similarly sensitive to the proteoglycan-induction effect of TGF-beta.

    Topics: Animals; Biglycan; Cell Transformation, Neoplastic; Cells, Cultured; Chondroitin Sulfate Proteoglycans; Decorin; Extracellular Matrix Proteins; Fibroblasts; Genes, ras; Lectins, C-Type; Proteoglycans; Rats; Temperature; Transforming Growth Factor beta; Versicans

1994
Regulated expression of Wnt family members during proliferation of C57mg mammary cells.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1994, Volume: 5, Issue:2

    At least six members of the Wnt gene family are expressed in the murine mammary gland during growth and differentiation, whereas several other Wnt family members participate in malignant transformation of this tissue. We have used the C57mg mammary cell line, which naturally expresses the Wnt-4 and Wnt-5a genes, to examine Wnt gene expression during proliferation. The data show that the growth factors basic fibroblast growth factor, transforming growth factor beta 1, and epidermal growth factor are mitogenic for C57mg cells, and partial transformation by Wnt-1 can substitute for the proliferative signal provided by these factors. Several different mitogenic stimuli selectively down-modulate the levels of endogenous Wnt-4 and Wnt-5a RNA in C57mg cells. Partial transformation by either Wnt-1 or Wnt-2 is accompanied by a dramatic decrease in Wnt-4 RNA and a small decrease in Wnt-5a RNA. Mitogenic stimulation by basic fibroblast growth factor or partial transformation by Int-2, a fibroblast growth factor family member, also leads to a selective decrease in the levels of endogenous Wnt RNA. No expression of the Wnt-4 and Wnt-5a genes is detectable in C57mg cells that are fully transformed by the activated tyrosine kinase oncogene Neu. In contrast, overexpression of Wnt-5a in C57mg cells does not lead to a transformed phenotype and is not accompanied by a decrease in endogenous Wnt-4 RNA levels. Overexpression of Wnt-5a does lead to a small decrease in endogenous Wnt-5a levels, perhaps through autoregulation. These data indicate that Wnt-4 and Wnt-5a expression in mammary cells is responsive to growth regulatory signals, and the down-modulation of expression of either or both genes correlates with cell proliferation. The inverse correlation between expression of the endogenous Wnt genes and cell proliferation suggests that Wnt-4 and Wnt-5a may participate in restricting the proliferation of C57mg cells.

    Topics: Animals; Cell Division; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; DNA; DNA Replication; Epidermal Growth Factor; Female; Fibroblast Growth Factor 2; Gene Expression Regulation; Kinetics; Mammary Glands, Animal; Mice; Multigene Family; Phenotype; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Thymidine; Transforming Growth Factor beta; Wnt Proteins; Wnt-5a Protein; Wnt1 Protein; Wnt4 Protein; Zebrafish Proteins

1994
Overexpression of c-K-ras, c-N-ras and transforming growth factor beta co-segregate with tumorigenicity in morphologically transformed C3H 10T1/2 cell lines.
    Carcinogenesis, 1994, Volume: 15, Issue:5

    Morphologic transformation and tumorigenicity are separate cellular phenotypes in transformed 10T1/2 cells. We have investigated the levels of expression of genes for c-myc, c-H-ras, c-K-ras, c-N-ras, TGF beta and Rb in 42 morphologically transformed 10T1/2 cell lines, in an attempt to define the molecular mechanisms governing morphologic transformation and tumorigenicity in the 10T1/2 cell system. The 10T1/2 cell lines investigated generally overexpressed mRNAs for c-myc, c-H-ras, and TGF beta relative to the levels expressed by wild-type 10T1/2 cells (levels of expression > 1.5-fold that of wild-type 10T1/2 cells). In contrast, only half of these cell lines overexpressed mRNAs for c-N-ras and/or Rb relative to wild-type 10T1/2 cells, and only 25% overexpressed c-K-ras mRNA. The mean levels of mRNA expression for each of c-K-ras, c-N-ras and TGF beta genes in tumorigenic cell lines were significantly greater than the mean levels of expression in non-tumorigenic cell lines, suggesting an association between tumorigenicity and the levels of expression of these specific genes. In contrast, levels of expression for c-myc, c-H-ras and Rb genes were not correlated with tumorigenicity. Cell lines that coexpressed high levels of c-K-ras, c-N-ras and TGF beta genes were likely to be tumorigenic (11/12 cell lines were tumorigenic), whereas cell lines that coexpressed low levels of these genes were unlikely to be tumorigenic (1/10 cell lines were tumorigenic). High expression of TGF beta was sufficient for tumorigenicity in the absence of high levels of expression of c-K-ras and c-N-ras (5/5 cell lines were tumorigenic). Elevated expression of either c-K-ras or c-N-ras alone was insufficient for tumorigenicity, however, coordinate overexpression of both c-K-ras and c-N-ras was associated with tumorigenicity irrespective of the expression status for TGF beta (13/15 cell lines were tumorigenic). These results suggest that overexpression of c-myc, c-H-ras and TGF beta are commonly associated with, and possibly mechanistically related to, the process of morphologic transformation in 10T1/2 cells. In addition, these results suggest that progression from morphologic transformation to tumorigenicity in 10T1/2 cell lines is frequently accompanied by overexpression of c-K-ras and c-N-ras, and by enhancement of the level of overexpression of TGF beta.

    Topics: Animals; Cell Line, Transformed; Cell Separation; Cell Transformation, Neoplastic; Gene Expression; Genes, ras; Genes, Retinoblastoma; Mice; Mice, Inbred C3H; Poly A; RNA, Messenger; Transforming Growth Factor beta

1994
Regulation of differentiation and protein kinase C expression in 3T3 T proadipocytes: effects of TGF-beta and transformation.
    Cell proliferation, 1994, Volume: 27, Issue:3

    We are studying the mechanisms that regulate proliferation and differentiation of normal 3T3 T proadipocytes and neoplastically transformed clones which have lost the ability to differentiate. The phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) and transforming growth factor beta (TGF-beta) are known inhibitors of the same step of the differentiation process in 3T3 T cells. Here, we examined the expression of the phorbol ester receptor/protein kinase C (PKC) during adipocytic differentiation of 3T3 T cells and its modulation by the differentiation inhibitor TGF-beta. PKC receptor assays were performed using a tritiated analogue of TPA and it was found that PKC receptor levels decreased approximately threefold during differentiation. Northern blot analyses revealed an even greater decrease of PKC transcripts during differentiation. TGF-beta inhibited not only differentiation, but the differentiation-dependent decrease in PKC levels as well. Transformed 3T3 T cells which have lost the ability to differentiate were found to express aberrant levels of PKC. The data suggest that TGF-beta may inhibit differentiation via a PKC-dependent pathway and that disruption of normal PKC levels or its regulation may be involved in the loss of differentiation control in transformed 3T3 T cells.

    Topics: 3T3 Cells; Adipocytes; Animals; Cell Differentiation; Cell Transformation, Neoplastic; Clone Cells; Mice; Protein Kinase C; Receptors for Activated C Kinase; Receptors, Cell Surface; Stem Cells; Transforming Growth Factor beta

1994
Growth-regulatory effects of sensory neuropeptides, epidermal growth factor, insulin, and somatostatin on the non-transformed intestinal epithelial cell line IEC-6 and the colon cancer cell line HT 29.
    Scandinavian journal of gastroenterology, 1993, Volume: 28, Issue:10

    A non-transformed small-intestinal cell line from the rat (IEC-6) and a human colon cancer cell line (HT 29) were examined for their trophic response to sensory neuropeptides. Substance P, neurokinin A (NKA), calcitonin gene-related peptide (CGRP), vasoactive intestinal peptide (VIP), and peptide YY (PYY) were tested. Epidermal growth factor (EGF), insulin, and somatostatin-14 were also used. Interaction studies were performed on IEC-6 cells by combining EGF or insulin with somatostatin-14. The sensory neuropeptides had no effect either on IEC-6 cell growth and DNA synthesis or on HT29 cell growth. EGF and insulin stimulated cell growth and DNA synthesis in IEC-6 cells and cell growth in HT 29 cells in a dose-dependent fashion. Somatostatin-14 had no effect either alone or in combination with EGF or insulin on IEC-6 cell growth and DNA synthesis. HT 29 cell growth was inhibited by somatostatin-14 only in the presence of serum with a maximal and significant response at 10(-7) M. Our observations suggest that the sensory neuropeptides do not exert a direct growth-regulatory effect either on IEC-6 cells or on HT 29 cells. Somatostatin, however, inhibits serum-induced HT 29 cell growth but does not interfere directly with the proliferative effect of serum, EGF, or insulin on IEC-6 cells in this model.

    Topics: Animals; Calcitonin Gene-Related Peptide; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Colonic Neoplasms; DNA, Neoplasm; Dose-Response Relationship, Drug; Epidermal Growth Factor; Epithelium; Gastrointestinal Hormones; Humans; Insulin; Intestinal Mucosa; Intestines; Neurokinin A; Neurons, Afferent; Neuropeptides; Peptide YY; Peptides; Rats; Somatostatin; Substance P; Transforming Growth Factor beta; Vasoactive Intestinal Peptide

1993
Loss of expression of transforming growth factor beta in skin and skin tumors is associated with hyperproliferation and a high risk for malignant conversion.
    Proceedings of the National Academy of Sciences of the United States of America, 1993, Jul-01, Volume: 90, Issue:13

    Mouse skin carcinomas arise from a small subpopulation of benign papillomas with an increased risk of malignant conversion. These papillomas arise with limited stimulation by tumor promoters, appear rapidly, and do not regress, suggesting that they differ in growth properties from the majority of benign tumors. The transforming growth factor beta (TGF-beta) proteins are expressed in the epidermis and are growth inhibitors for mouse keratinocytes in vitro; altered TGF-beta expression could influence the growth properties of high-risk papillomas. Normal epidermis, tumor promoter-treated epidermis, and skin papillomas at low risk for malignant conversion express TGF-beta 1 in the basal cell compartment and TGF-beta 2 in the suprabasal strata. In low-risk tumors, 90% of the proliferating cells are confined to the basal compartment. In contrast, the majority of high-risk papillomas are devoid of both TGF-beta 1 and TGF-beta 2 as soon as they arise; these tumors have up to 40% of the proliferating cells in the suprabasal layers. Squamous cell carcinomas are also devoid of TGF-beta, suggesting that they arise from the TGF-beta-deficient high-risk papillomas. In some high-risk papillomas, TGF-beta 1 loss can occur first and correlates with basal cell hyperproliferation, while TGF-beta 2 loss correlates with suprabasal hyperproliferation. Similarly, TGF-beta 1-null transgenic mice, which express wild-type levels of TGF-beta 2 in epidermis but no TGF-beta 1 in the basal layer, have a hyperproliferative basal cell layer without suprabasal proliferation. In tumors, loss of TGF-beta is controlled at the posttranscriptional level and is associated with expression of keratin 13, a documented marker of malignant progression. These results show that TGF-beta expression and function are compartmentalized in epidermis and epidermal tumors and that loss of TGF-beta is an early, biologically relevant risk factor for malignant progression.

    Topics: Animals; Base Sequence; Carcinoma, Squamous Cell; Cell Division; Cell Transformation, Neoplastic; Epidermal Cells; Epidermis; Female; Keratins; Mice; Mice, Transgenic; Molecular Sequence Data; Papilloma; Risk; Skin Neoplasms; Transforming Growth Factor beta

1993
Potentiation of metastatic capacity by transforming growth factor-beta 1 gene transfection.
    Japanese journal of cancer research : Gann, 1993, Volume: 84, Issue:6

    This study was designed to assess whether the excessive secretion of transforming growth factor-beta 1 (TGF-beta 1) by Chinese hamster ovary (CHO) cells transfected with TGF-beta 1 gene may be linked to the development of a metastatic phenotype. We observed large numbers of metastatic colonies in the lungs of nude mice inoculated with the transfected CHO cells. The tumors derived from these transfected cells demonstrated marked angiogenesis. We postulate that the overproduction of TGF-beta 1 by these tumors may participate in the metastatic progression following establishment of angiogenesis at the primary tumor site.

    Topics: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; CHO Cells; Cricetinae; Female; Lung Neoplasms; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Metastasis; Neoplasm Transplantation; Neovascularization, Pathologic; Phenotype; Transfection; Transforming Growth Factor beta

1993
Loss of thyrotropin regulation and transforming growth factor beta-induced growth arrest in erbB-2 overexpressing rat thyroid cells.
    Cancer research, 1993, Nov-15, Volume: 53, Issue:22

    Amplification of erbB-2 gene and overexpression of gp185erbB-2 gene product is found in approximately one-third of primary human breast and ovarian cancer. Overexpression of gp185erbB-2 was recently found in human papillary thyroid carcinomas, but not in thyroid follicular carcinomas or adenomas. The erbB-2 gene encodes a cell surface growth factor receptor with intrinsic tyrosine kinase activity. Wild type human erbB-2 has been shown to act as a potent oncogene when overexpressed in mouse fibroblasts. To test whether overexpression of normal human erbB-2 gene can transform epithelial differentiated rat thyroid cells, these cells were infected with a recombinant retroviral expression vector containing the erbB-2 protooncogene. Rat thyroid cells expressing high levels of gp185erbB-2 do not display a fully transformed and tumorigenic phenotype. However, the isolated cell clones that overexpress gp185erbB-2, show changes in their growth properties if compared to normal thyroid cells, since they can grow in absence of thyrotropin, the main growth factor controlling thyroid cell proliferation in vitro, and do not respond to the growth inhibitory effect of transforming growth factor beta.

    Topics: Animals; Blotting, Western; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Cinnamates; Drug Resistance; Hygromycin B; Mice; Mice, Nude; Neoplasm Transplantation; Oncogene Proteins, Viral; Proto-Oncogenes; Rats; Rats, Inbred F344; Receptor, ErbB-2; Receptors, Thyrotropin; RNA, Messenger; Thyroid Gland; Transforming Growth Factor beta

1993
Sequential changes in MHC antigen expression induced by the v-Ki-ras oncogene.
    Cancer immunology, immunotherapy : CII, 1993, Volume: 37, Issue:6

    A series of early-passage cell lines were transformed with the v-Ki-ras oncogene with the aim of examining the effect of an activated ras gene on the ability of these cells to express major histocompatibility complex (MHC) antigens. These cell lines were found to undergo multiple phenotypic changes upon transformation and subsequent proliferation. At early passage, the predominant effect of ras was an increased ability to express class II antigens when induced with interferon gamma (IFN gamma). For class I antigens, maximum levels of expression induced with IFN gamma were largely unaffected, however, decreased sensitivity to induction with this lymphokine was noted. With subsequent in vitro or in vivo passage, both class I and class II antigen inducibility was attenuated. The latter phenotypic change was found to be transferable by coculture, implicating a soluble IFN gamma antagonist. Conditioned media from ras-transformed cells treated to activate their latent transforming growth factor beta (TGF beta) content mediated similar changes in MHC antigen inducibility, suggesting that TGF beta may be involved in modulating MHC antigen expression in ras-transformed cells.

    Topics: Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Culture Media, Conditioned; Flow Cytometry; Gene Expression Regulation, Neoplastic; Genes, ras; Histocompatibility Antigens Class I; Histocompatibility Antigens Class II; Interferon-gamma; Mice; Mice, Inbred BALB C; Mice, Inbred C3H; Proto-Oncogene Proteins p21(ras); Transforming Growth Factor beta

1993
Negative regulation of mitogen-stimulated, anchorage-independent cell growth by a tumor-suppressor gene function.
    Molecular carcinogenesis, 1993, Volume: 7, Issue:4

    Immortal, nontumorigenic cell lines of Syrian hamster embryo (SHE) cells with different tumor-suppressing activity were isolated. Subclones from the parental cells were isolated that either had retained (supB+) or lost (supB-) the ability to suppress tumorigenicity after hybridization with tumor cells. The growth properties of these cells were studied to determine how this tumor-suppressor gene function influences cell growth. When the cells were grown on plastic, their growth properties were similar, and neither cell type grew in soft agar containing 10% serum, which supported the growth of tumorigenic cells. However, in agar supplemented with growth factors and 10% serum, supB- cells formed colonies whereas supB+ cells did not. Efficient growth (colony-forming efficiencies greater than 20%) of supB- cells was obtained in agar supplemented with serum and a combination of epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and insulin (EPI) or with serum and basic fibroblast growth factor (bFGF). The effect of EPI and bFGF together was additive. supB+ cells failed to grow under any of these conditions, suggesting that the suppressor gene function blocked the growth response of the cells to multiple growth factors when the cells were suspended in agar. In SupB- cells, transforming growth factor-beta 1 and retinoic acid inhibited anchorage-independent growth response to EPI but not the growth response to bFGF. These observations are consistent with the hypothesis that bFGF stimulates the growth of supB- cells by a signal transduction pathway that differs from the pathway stimulated by EGF or PDGF. Thus, this suppressor gene function may regulate anchorage-independent growth at some common point in signal transduction for multiple mitogens.

    Topics: Animals; Cell Adhesion; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cricetinae; Drug Interactions; Epidermal Growth Factor; ErbB Receptors; Fetus; Fibroblast Growth Factor 2; Genes, Tumor Suppressor; Growth Substances; Insulin; Interferon-gamma; Mesocricetus; Platelet-Derived Growth Factor; Receptor, Insulin; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

1993
Relationship of levels and kinetics of H-ras expression to transformed phenotype and loss of TGF-beta 1-mediated growth regulation in intestinal epithelial cells.
    Experimental cell research, 1993, Volume: 204, Issue:1

    Immortalized, nontumorigenic rat intestinal epithelial cells (IEC-18) can be transformed to tumorigenicity by expression of an activated human H-ras gene. Here we describe the characteristics of an IEC-18 cell line in which the activated human H-ras gene has been introduced under the control of the steroid-sensitive promoter of the mouse mammary tumor virus long-terminal repeat. The clonal cell line (IEC-18 C125) is phenotypically normal in the absence of the transcription inducer, dexamethasone, and transformed when treated with high levels of inducer. Transformed morphology and growth characteristics are dependent on levels of H-ras expression. IEC-18 C125 cells have been used to demonstrate a general relationship (dose and kinetic) between H-ras expression and loss of TGF-beta 1-mediated growth regulation. This effect occurs concomitantly with a ras-dependent change in the profile of TGF-beta 1 binding proteins.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Clone Cells; Dexamethasone; Epithelium; Gene Expression; Genes, ras; Intestines; Kinetics; Rats; Transforming Growth Factor beta

1993
Orthovanadate both mimics and antagonizes the transforming growth factor beta action on normal rat kidney cells.
    Journal of cellular physiology, 1993, Volume: 154, Issue:2

    Normal rat kidney [NRK] cells grown in the presence of epidermal growth factor (EGF) or platelet-derived growth factor (PDGF) have a normal phenotype and undergo density-dependent growth inhibition, whereas in the presence of multiple growth factors, density arrest is lost and the cells become phenotypically transformed. We studied the influence of the protein tyrosine phosphatase (PTPase) inhibitor sodium orthovanadate on the mitogenic stimulation of NRK cells by growth factors and on transformation-linked properties as loss of density-dependent growth inhibition and anchorage-independent growth. The fraction of cells in serum-deprived monolayer cultures that is induced to proliferate upon mitogenic stimulation by EGF or PDGF is only slightly enhanced upon addition of low concentrations (25-50 microM) of vanadate. Addition of vanadate per se induces proliferation of only a very limited amount of cells, but results in a shift of the dose-response curves for other growth factors to lower concentrations. Vanadate added in combination with EGF or PDGF is able to mimic the effect of transforming growth factor beta (TGF beta) in inducing phenotypic transformation. In monolayer cultures density-dependent growth inhibition is lost and anchorage-independent proliferation is observed on dishes coated with poly(2-hydroxy-ethyl methacrylate) (polyHEMA). The extent of these changes is similar to that induced by TGF beta. However, the morphology of the obtained colonies in polyHEMA-coated dishes is quite different. Cells transformed by TGF beta in the presence of EGF form rather amorphous colonies, whereas in the presence of orthovanadate colonies are formed that tend to fall apart in loose cells. The effect of vanadate on cell transformation is dependent on the growth factor conditions in a bimodal way. When a suboptimal dose of growth factor(s) is used, 25 microM vanadate is very effective in preventing density-induced growth inhibition and stimulating anchorage-independent proliferation. However, the same concentration of vanadate is inhibitory when cells are maximally stimulated and antagonizes the transforming effect of TGF beta added in combination with other growth factors. It is hypothesized that vanadate acts on a set of different protein tyrosine phosphatases. Some of these are positive and others negative regulators of growth.

    Topics: Animals; Cell Adhesion; Cell Transformation, Neoplastic; Cells, Cultured; Drug Interactions; Epidermal Growth Factor; Kidney; Platelet-Derived Growth Factor; Rats; Transforming Growth Factor beta; Vanadates

1993
The 12S adenoviral E1A protein immortalizes avian cells and interacts with the avian RB product.
    Oncogene, 1993, Volume: 8, Issue:3

    Quail cells were immortalized for the first time by using retroviruses expressing the 12S adenoviral E1A gene. In these cells, interaction between the 12S E1A product and the quail RB protein was shown, suggesting that the 12S adenoviral E1A product works in avian cells through similar biochemical pathways as in mammalian cells by interacting and inactivating host cellular proteins, including the RB product. These results confirm that the RB product exhibits a universal function among higher vertebrates in controlling cellular growth and tumor progression.

    Topics: Adenoviridae; Adenovirus E1A Proteins; Animals; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Coturnix; Gene Expression; Retinoblastoma Protein; Transforming Growth Factor beta

1993
Loss of differentiation control in transformed 3T3 T proadipocytes.
    Cancer research, 1993, Apr-15, Volume: 53, Issue:8

    Nontransformed 3T3 T mesenchymal/proadipocyte stem cells can be readily induced to differentiate, yet previous work has shown that 3T3 T cells that are spontaneously or virally transformed not only lose their normal growth control mechanisms but also lose the ability to differentiate. Loss of growth control can be due to autocrine mechanisms in some transformed cells, but the mechanisms involved in disrupting differentiation control are poorly understood. Our goal is to further define the growth and differentiation defects that arise in neoplastically transformed cells and the mechanisms underlying those defects. For example, exogenous transforming growth factor beta and tumor necrosis factor, both of which are secreted aberrantly by some tumor cells, are known inhibitors of different steps of the normal 3T3 T adipocyte differentiation process, suggesting a potential role as autocrine factors in disrupting differentiation of transformed 3T3 T cells. In the current study we transformed 3T3 T cells in vitro with chemical or UV irradiation treatment in order to determine if the acquisition of the transformed phenotype after these treatments is also associated with loss of differentiation control as it is with spontaneously or virally transformed cells. Four chemically and two UV-treated 3T3 T cell lines were isolated from type III foci and all have been found to be tumorigenic in syngeneic animals and to have lost the ability to differentiate. Relative to the parental cell line the differentiation abilities of the transformed clones ranged from 0 to less than 5%. In this regard, we also analyzed the normal and aberrant expression of three growth factors and differentiation inhibitors in transformed cells. Both transforming growth factor alpha and beta were found to be expressed in non-transformed 3T3 T cells as determined by Northern blot analyses. In addition, both were found to be down-regulated during differentiation of 3T3 T cells. Transformed/differentiation-defective 3T3 T cells expressed varied levels of transforming growth factor alpha and beta. Three of the new transformed clones expressed particularly high levels of transforming growth factor alpha. Very low levels of tumor necrosis factor expression were found in the normal cells and the transformed cells appeared to express tumor necrosis factor at similar levels. In contrast, none of the transformed cells expressed any of the differentiation-specific genes tested (lipoprotein lipase, glycerol-3-

    Topics: 3T3 Cells; Adipose Tissue; Animals; Cell Differentiation; Cell Line, Transformed; Cell Transformation, Neoplastic; Gene Expression; Male; Mice; Mice, Inbred BALB C; Stem Cells; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

1993
TGF-beta induces an inhibitory effect of normal cells directed against transformed cells.
    International journal of cancer, 1993, Apr-22, Volume: 54, Issue:1

    Co-cultivation of normal and transformed fibroblasts in the presence of exogenously added transforming growth factor type beta I (TGF-beta I) leads to a drastic reduction of colony-forming transformed cells. This inhibitory effect is dependent both on the dose of TGF-beta applied and the density of normal cells. It does not reflect a direct action of TGF-beta on transformed cells, but is rather induced by TGF-beta in normal cells. In the absence of exogenously added TGF-beta, normal cells exhibit a basal inhibitory activity, which can be abrogated by anti-TGF-beta. TGF-beta-mediated inhibition of transformed cells by normal cells may be considered as a novel potent controlling step in carcinogenesis.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; In Vitro Techniques; Mice; Rats; Transforming Growth Factor beta

1993
Autocrine production of TGF-alpha and TGF-beta during tumour progression of rat oral keratinocytes.
    Carcinogenesis, 1993, Volume: 14, Issue:5

    This study describes a new technique to separate transforming growth factor-alpha (TGF-alpha) and transforming growth factor-beta (TGF-beta) from culture supernatants using ion exchange chromatography; assays of competitive inhibition of ligand binding were used to quantify the amount of growth factor. The method was simple, inexpensive and did not require large volumes of culture medium. The autocrine production of TGF-alpha and TGF-beta was examined in oral keratinocyte cell lines derived from the palatal and lingual mucosa of rats painted with the carcinogen 4-nitroquinoline N-oxide (4NQO). Escape from cellular senescence (immortality) was associated with a marked increase in TGF-alpha production (cell line R2P) but tumour progression, as reflected by the development of anchorage independence in agarose gels and tumorigenicity in athymic mice, did not result in a consistent increase or decrease of TGF-alpha production compared to normals. Four cell lines (R8AP, R1T, R3T, R1P), with different functional cellular phenotypes, produced two to three times more TGF-alpha than normals. TGF-alpha production was inversely correlated to epidermal growth factor cell surface receptor expression. The autocrine production of TGF-beta was variable with the majority of cell lines producing markedly little TGF-beta; three cell lines (R4T, R8BP, R9T) produced more TGF-beta than normals. The production of TGF-beta was unrelated to tumour progression, the expression of TGF-beta cell surface receptors or TGF-alpha production. The results indicate that the autocrine production of TGF-alpha and TGF-beta are not accurate markers of tumour progression in the rat 4NQO model of oral carcinogenesis.

    Topics: 4-Nitroquinoline-1-oxide; Animals; Blotting, Western; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Keratinocytes; Mice; Mice, Nude; Mouth Mucosa; Neoplasm Transplantation; Rats; Thymidine; Transforming Growth Factor alpha; Transforming Growth Factor beta

1993
Transformed rat tracheal epithelial cells exhibit alterations in transforming growth factor-beta secretion and responsiveness.
    Molecular carcinogenesis, 1992, Volume: 5, Issue:1

    The purpose of our studies was to define abnormalities in the transforming growth factor-beta (TGF-beta) system of transformed rat tracheal epithelial (RTE) cells that might cause their abnormal growth behavior. We found that many, but not all, of the transformed cell lines were hyporesponsive or unresponsive to the growth inhibitory effects of TGF-beta 1. Scatchard and receptor cross-linking analyses indicated that loss of TGF-beta 1 responsiveness of transformed cells was probably not due to changes in receptor number or affinity, or to changes in expression of the three TGF-beta-binding protein subtypes. Transformed cells were found to secrete far less TGF-beta-like activity (less than 1/10) than primary cells. Cultured normal and transformed RTE cells expressed three TGF-beta 1 transcripts of 2.5, 1.9, and 1.4 kb. In contrast, rat kidney tissue, a rat embryo fibroblast cell line, and a rat liver cell line expressed only the typical 2.5-kb mRNA transcript commonly reported in the literature. In spite of the marked differences in TGF-beta secretion between normal and transformed cells, their levels of TGF-beta 1 mRNA expression were similar. This suggests a change in the posttranscriptional regulation of TGF-beta 1 expression. TGF-beta 2 message was not detected in either normal or transformed RTE cells in culture. These findings are consistent with the hypothesis that the abnormal growth behavior of transformed RTE cells is at least in part due to disturbances of the TGF-beta system.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Cross-Linking Reagents; Epithelium; Gene Expression; In Vitro Techniques; Rats; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; RNA, Messenger; Trachea; Transforming Growth Factor beta

1992
Overexpression of H-ras oncogene induces resistance to the growth-inhibitory action of transforming growth factor beta-1 (TGF-beta 1) and alters the number and type of TGF-beta 1 receptors in rat intestinal epithelial cell clones.
    Oncogene, 1992, Volume: 7, Issue:3

    In this report, we utilize rat intestinal cell (IEC-18) clones expressing an activated human H-ras gene to investigate the relationship between malignant transformation and growth control by transforming growth factor beta (TGF-beta). We demonstrate that clones expressing high levels of H-ras oncogene lose sensitivity to the growth inhibitory action of TGF-beta. The loss of sensitivity is related to the degree of H-ras expression and is shown to be a direct consequence of H-ras expression through the use of a clonal cell line with inducible expression of activated H-ras. Co-incident with the loss of growth inhibition, ras-expressing clones display an altered expression of TGF-beta-binding proteins as detectable by [125I]TGF-beta cross-linking. While IEC-18 cells express type II (92 kDa) binding protein predominantly, H-ras expression induces a shift to predominantly type I (69 kDa) binding protein expression.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Clone Cells; Gene Expression; Genes, ras; Growth Inhibitors; In Vitro Techniques; Intestinal Mucosa; Proto-Oncogene Proteins p21(ras); Rats; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

1992
Regulation of growth and gene expression in human papillomavirus-transformed keratinocytes by transforming growth factor-beta: implications for the control of papillomavirus infection.
    Molecular carcinogenesis, 1992, Volume: 6, Issue:2

    Cervical carcinogenesis is a multistep process that appears to be initiated by infection of squamous epithelial cells in the cervix with one of a limited number of human papillomavirus (HPV) types. However, the mechanisms involved in the evolution of benign, HPV-induced lesions to malignancy have not yet been fully elucidated. Transforming growth factor-beta (TGF-beta), a multifunctional growth factor produced by cells in the skin, inhibits the proliferation of foreskin and cervical keratinocytes in vitro. We examined the effects of TGF-beta on growth and virus early-gene expression in cell lines immortalized by two HPV types associated with cervical carcinogenesis as well as the expression of TGF-beta 1 mRNA transcripts in normal and HPV-positive cells in vivo and in vitro. We found that normal and HPV-positive cells expressed similar levels of TGF-beta 1 mRNAs and exhibited similar patterns of responsiveness to three isoforms of TGF-beta in both monolayer and modified organotypic cultures. Of particular interest is our finding that the expression of the E6 and E7 early viral transforming regions of both HPV16 and HPV18 was reversibly and rapidly inhibited by TGF-beta. In one HPV16-positive cell line examined in detail, inhibition of HPV expression required protein synthesis and occurred at the level of transcription. HPV-immortalized cells selected for resistance to in vitro differentiation signals remained sensitive to TGF-beta-mediated growth inhibition. These results, showing that both growth and virus gene expression in HPV-transformed cells were responsive to TGF-beta, suggest that endogenous growth factors produced by different cell types in squamous epithelium may play a role in the progression of cervical neoplasia.

    Topics: Blotting, Northern; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Down-Regulation; Ethionine; Female; Gene Expression Regulation, Neoplastic; Humans; Keratinocytes; Male; Papillomaviridae; RNA, Messenger; Transcription, Genetic; Transfection; Transforming Growth Factor beta; Tumor Virus Infections; Uterine Cervical Neoplasms; Viral Proteins

1992
Progressive abrogation of TGF-beta 1 and EGF growth control is associated with tumour progression in ras-transfected human keratinocytes.
    International journal of cancer, 1992, Sep-30, Volume: 52, Issue:3

    This study examined the response of human keratinocytes in different stages of transformation to exogenous TGF-beta 1 and EGF as well as their receptor and growth-factor expression. Cells of the spontaneously immortalized HaCaT cell line and c-Ha-ras transfected clones (I-6, I-7, II-3, II-4) exhibited different tumorigenic potentials when transplanted to athymic mice. HaCaT- and I-6 cells were non-tumorigenic, I-7 cells formed persisting epidermal cysts (benign tumours) and II-3 and II-4 cells developed into invasive squamous-cell carcinomas. TGF-beta 1 inhibited thymidine uptake in a dose-dependent manner, a progressive decrease in response being associated with an increasing malignant potential (HaCaT greater than I-6 greater than I-7 = II-4). HaCaT-cells and ras-clones expressed TGF-beta 1 mRNA at similar levels, but cells of increasing malignant potential secreted markedly less receptor-binding TGF-beta (HaCaT greater than I-6 = I-7 greater than II-3 greater than II-4) into the culture medium. Whilst ras-transfected cells expressed fewer TGF-beta receptors than HaCaT cells, there was little difference between TGF-beta receptor number or affinity between the 4 transfected cell clones. The same was true for the TGF-beta receptor types, but Type-II receptors were expressed at lower levels by the malignant clones II-3 and II-4. When HaCaT and ras-transfected cells were investigated for their response to exogenous EGF, cells were refractory (I-7, II-4), partially stimulated (I-6) or fully stimulated (HaCaT). Cells with increasing malignant potential produced increasing amounts of endogenous TGF-alpha (II-4 = II-3 greater than I-7 = I-6 greater than HaCaT). All tumorigenic ras clones expressed higher mRNA levels than HaCaT-cells. Ras-transfected clones expressed fewer high- and low-affinity EGF receptors than HaCaT cells with a tendency toward increased numbers of high-affinity EGF receptors associated with increasing malignant potential (II-4 = II-3 greater than I-7 greater than I-6) but these changes were associated with a progressive decrease in receptor affinity. The results indicate that tumour progression in human epidermal keratinocytes transfected with c-Ha-ras is associated with a progressive abrogation of TGF-beta 1 and EGF growth control. They suggest that the increased autonomous growth potential associated with advanced stages of epithelial tumour progression can be defined more closely using a cellular profile of TGF-beta and EGF.

    Topics: Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Genes, ras; Humans; Keratinocytes; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transfection; Transforming Growth Factor alpha; Transforming Growth Factor beta

1992
Transforming growth factor beta stimulates mitogenically mouse NIH3T3 fibroblasts and those cells transformed by the EJ-H ras oncogene.
    Growth factors (Chur, Switzerland), 1992, Volume: 6, Issue:4

    TGF-beta 1 stimulates thymidine incorporation and the growth rate of mouse NIH3T3 fibroblasts and of those cells transformed by the EJ-H-ras oncogene (TR15 cells), in the presence and the absence of serum. Thymidine incorporation, in serum-deprived cells, is stimulated to a higher degree by 0.1-1 ng/ml of TGF-beta in NIH3T3 than in TR15 cells, which have a 10-fold higher basal level of incorporation. In both cell types TGF-beta 1 is as active, or more active than other mitogens (TGF-alpha, PDGF-AB, bFGF) at the same concentration. The growth rate of NIH3T3 cells, in low serum or serum-free (S-) medium, is stimulated by only 10 picograms/ml of TGF-beta 1, and that of TR15 cells, in S- medium, by only 1 picogram/ml. In contrast, TGF-beta 1 inhibits mitogenically unestablished mouse embryo fibroblasts and these fibroblasts immortalized spontaneously and able to grow in S- medium. It also inhibits the anchorage-independent growth of TR15 cells. NIH3T3 and TR15 cells respond, similarly, to TGF-beta activated by acification of their culture medium. The kinetics of thymidine incorporation and of activation of the c-myc proto-oncogene, observed already after 1 hr, in treated NIH3T3 and TR15 cells, suggests a direct mitogenic stimulation. The level of activated c-myc RNA is 2-fold higher at 2 hr, and subsequently decreases relatively less in the TR15 cells.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Culture Media, Conditioned; DNA; Fibroblasts; Genes, myc; Genes, ras; Kinetics; Mice; Thymidine; Transcription, Genetic; Transforming Growth Factor alpha; Transforming Growth Factor beta

1992
Stepwise transformation of primary thyroid epithelial cells by a mutant Ha-ras oncogene: an in vitro model of tumor progression.
    Molecular carcinogenesis, 1992, Volume: 6, Issue:2

    Activating mutations of the ras oncogene family occur at high frequency in all stages of thyroid tumorigenesis, both human and experimental. To test the causal nature of this association, and to investigate the biological role of ras mutation, we introduced a mutant c-Ha-ras gene into normal rat thyroid follicular cells using an ecotropic retroviral vector. The major immediate effect was to greatly extend the proliferative lifespan of these cells in culture from less than 3 to more than 15 doublings, without any observable loss of growth-factor dependence or differentiated functions. This in vitro phenotype strongly supports an initiating role for ras mutation in the genesis of benign thyroid tumors (adenomas) in vivo. Spontaneous transformation was observed at low frequency on continuous culture of mutant ras-expressing cells, giving rise to fully immortalized, growth factor-independent, highly tumorigenic lines. Transformation was associated with (i) loss of responsiveness to the growth inhibitor TGF-beta 1, and (ii) greatly increased nuclear levels of p53 protein, which unexpectedly was not due to point mutation in the conserved regions of the p53-coding sequence. We postulate that these two phenomena are causally related to each other and to the transformed phenotype.

    Topics: Adenoma; Animals; Base Sequence; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; DNA Probes; Dose-Response Relationship, Drug; Genes, ras; In Vitro Techniques; Male; Molecular Sequence Data; Mutation; Proto-Oncogene Proteins p21(ras); Rats; Rats, Inbred Strains; Serum Albumin, Bovine; Thyroglobulin; Thyroid Neoplasms; Thyrotropin; Transduction, Genetic; Transforming Growth Factor beta; Tumor Suppressor Protein p53

1992
Raf-1 protein kinase is an integral component of the oncogenic signal cascade shared by epidermal growth factor and platelet-derived growth factor.
    Molecular and cellular biology, 1992, Volume: 12, Issue:11

    Our recent studies with cell mutants indicate that a cascade shared by the epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) signals exists in NRK cells and mediates oncogenic signals induced by many oncogenes (A. Masuda, S. Kizaka-Kondoh, H. Miwatani, Y. Terada, H. Nojima, and H. Okayama, New Biol. 4:489-503, 1992). We have employed the antisense RNA technique to investigate possible involvement of Raf-1 kinase in this signal transduction cascade. NRK cell clones highly reduced in the Raf-1 production are generated by the expression of a c-raf-1 antisense RNA. They have no apparent growth defects and retain proper mitotic responses to growth factors but are refractory to transformation by EGF or PDGF plus transforming growth factor beta, v-erbB, v-fms, v-K-ras, v-mos, v-fos, v-src, simian virus 40 large T, and polyomavirus middle T but not by v-raf or adenovirus E1A. These results not only support our model for the oncogenic signal cascade but also lead to the conclusion that Raf-1 protein kinase is a downstream component of this oncogenic signal cascade shared by EGF and PDGF.

    Topics: Blotting, Western; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cloning, Molecular; Epidermal Growth Factor; Oncogenes; Platelet-Derived Growth Factor; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-raf; Signal Transduction; Transforming Growth Factor beta

1992
Malignant transformation of ovarian epithelium.
    Journal of the National Cancer Institute, 1992, Apr-15, Volume: 84, Issue:8

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Chromosome Aberrations; Epithelial Cells; Female; Genes, fms; Genes, p53; Humans; Mutation; Ovarian Neoplasms; Ploidies; Transforming Growth Factor beta

1992
Reactivity to TGF-beta is one step towards transformation in a murine fibroblast cell line.
    International journal of cancer, 1992, Apr-22, Volume: 51, Issue:1

    High doses of TGF-beta induce reactivity to TGF-beta in a distinct number of C3H10T1/2 fibroblasts suspended in soft agar. Reactive cells can be isolated and stably maintain this property. In the absence of exogenously added TGF-beta, these cells do not form colonies, but show distinct changes in their morphology and cytoskeleton. In contrast to normal C3H10T1/2 cells, they are able to form colonies when low doses of TGF-beta are added. TGF-beta-reactive cells are more easily transformed by UV light than normal C3H10T1/2 cells, and so maintain their transformed phenotype in the absence of added TGF-beta. Reactivity for TGF-beta, therefore, represents one step towards transformation.

    Topics: Animals; Cell Transformation, Neoplastic; Colony-Forming Units Assay; Fibroblasts; Mice; Transforming Growth Factor beta

1992
Loss of transforming growth factor beta 1 (TGF-beta 1)-induced growth arrest and p34cdc2 regulation in ras-transfected epithelial cells.
    Oncogene, 1992, Volume: 7, Issue:8

    Transforming growth factor beta 1 (TGF-beta 1) is a potent inhibitor of mink lung epithelial (CCL64) cell growth in culture. The fact that many transformed epithelial cells have escaped from negative growth control by TGF-beta 1 suggests that transfected epithelial cells may be an appropriate model for investigating the growth-inhibitory mechanism of TGF-beta 1. We transfected CCL64 cells with a mouse c-myc oncogene (pSVc-myc1), a mutated Harvey-ras (Ha-ras) oncogene, or a combination of both. The results indicate that cells transfected with c-myc alone exhibit normal morphology and maintain sensitivity to TGF-beta 1 growth arrest, but are unable to form colonies in soft agar in the presence or absence of TGF-beta 1. Cells transfected with Ha-ras, or co-transfected with c-myc, display a transformed morphology, grow spontaneously under anchorage-independent conditions and acquire a complete resistance to growth inhibition by TGF-beta 1. Affinity cross-linking of [125I]TGF-beta 1 to cell-surface receptors from these transfectants revealed that all three TGF-beta receptor types were present and no significant differences in [125I]TGF-beta 1 labeling of these receptors was observed. Since we have previously demonstrated that modulation of p34cdc2 kinase is a marker for TGF-beta 1 growth inhibition, we investigated p34cdc2 activity in the CCL64 transfected clones. The results show that in the control CCL64 cells and in the myc-transfected clones TGF-beta 1 regulation of p34cdc2 activity is maintained. In the ras- and ras + myc-transfected cells p34cdc2 phosphorylation and histone H1 kinase activity is significantly increased and regulation by TGF-beta 1 is lost.

    Topics: Animals; Blotting, Northern; CDC2 Protein Kinase; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epithelium; Gene Expression Regulation, Neoplastic; Genes, myc; Genes, ras; Mink; Phosphorylation; Transfection; Transforming Growth Factor beta

1992
Altered metabolic and adhesive properties and increased tumorigenesis associated with increased expression of transforming growth factor beta 1.
    The Journal of cell biology, 1992, Volume: 118, Issue:3

    Transforming growth factor-beta (TGF-beta) is a potent mediator of cell proliferation and extracellular matrix formation, depending on the cell type and the physiological conditions. TGF-beta is usually secreted in a "latent" complex that needs activation before it can exert its effects. Several observations correlate increased expression of TGF-beta 1 with tumorigenesis. To evaluate the physiological relevance of increased TGF-beta 1 synthesis in tumor cells we established cell clones overexpressing TGF-beta 1 and observed the resulting physiological changes in TGF-beta overproducing cells in vitro and in vivo. As a model system we used the human E1A-transformed 293 tumor cells, which are insensitive to the direct growth modulatory effects of TGF-beta. The selection of this cell line allows an assessment of physiological alterations independent of TGF-beta induced proliferative changes. The use of two TGF-beta 1 expression vectors containing either the natural or a modified TGF-beta 1 precursor cDNA permitted the establishment of separate 293 cell lines overexpressing latent or active TGF-beta. Comparison of the resulting changes in glycolytic rate, adhesiveness and integrin and plasminogen activator expression established that, in vitro, both types of clones behaved similarly, indicating that expression of latent TGF-beta induces autocrine changes in the tumor cells and thus suggesting that some level of cell-associated activation occurs. TGF-beta overexpression resulted in an increased metabolic rate due to enhanced glycolysis, a property long associated with tumor cells. This increased glycolysis was not associated with altered proliferation. Cells overexpressing TGF-beta also displayed enhanced fibronectin mRNA and plasminogen activator synthesis and increased adhesiveness in vitro. They showed enhanced survival when plated sparsely on plastic in the absence of serum, and attached more readily to laminin. In addition, synthesis of several beta 1 integrins, in particular the alpha 1/beta 1, alpha 2/beta 1, and alpha 3/beta 1, all of which recognize laminin, were enhanced. Finally, cells overexpressing active TGF-beta, but not latent TGF-beta, also showed increased tumorigenicity in nude mice. Thus, an increase in endogenous TGF-beta synthesis confers several proliferation-independent phenotypic changes which may be of significance for the survival of the tumor cell inoculum or its subsequent growth, and for tumor formation and development. In the case

    Topics: Animals; Base Sequence; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; DNA; Endopeptidases; Fibronectins; Gene Expression; Humans; Integrins; Mice; Mice, Nude; Molecular Sequence Data; Neoplasm Invasiveness; Neoplasms, Experimental; Transforming Growth Factor beta; Tumor Cells, Cultured

1992
Eosinophils, tissue eosinophilia, and eosinophil-derived transforming growth factor alpha in hamster oral carcinogenesis.
    Cancer research, 1992, Jan-15, Volume: 52, Issue:2

    Eosinophilia in tissues and/or circulating blood is known to be associated with a wide variety of malignancies but the role of the eosinophil in neoplastic conditions is not known. Using the cheek pouch of the Syrian hamster as an experimental model for oral carcinogenesis, it has recently been shown that eosinophils at sites of developing oral cancer express the multifunctional cytokine, transforming growth factor alpha (TGF-alpha). This study investigated the time course of eosinophil infiltration, tissue eosinophilia associated with malignant epithelium, and eosinophil-derived TGF-alpha mRNA during the 16-week 7,12-dimethylbenz[a]anthracene (DMBA)-induced oral cancer development process. The results reveal that the occasional eosinophil is normally present in the lamina propria of hamster oral mucosa. With progressive DMBA treatments, there is an increase of eosinophils infiltrating into the lamina propria. By weeks 12-16, the number of eosinophils is significantly higher in DMBA-treated pouches than in control pouches treated with the vehicle mineral oil alone. Analysis of the infiltrating eosinophils into fully developed hamster oral carcinomas reveals that tissue eosinophilia is associated with 78% of the stromal areas associated with malignant epithelium, while only 7% of sites associated with non-tumor oral epithelium (normal, hyperplastic-dysplastic) exhibited eosinophilia. Furthermore, the majority of the eosinophils associated with malignant epithelium were found to contain TGF-alpha mRNA. The number of TGF-alpha mRNAs containing eosinophils associated with malignant oral epithelium is significantly higher than that associated with nonmalignant oral epithelium. Together, these results suggest that eosinophils are recruited to tumor-developing sites, that they predominantly associate with malignant epithelium, and that most tumor-associated eosinophils express the cytokine TGF-alpha.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Cell Transformation, Neoplastic; Cricetinae; Eosinophils; Gene Expression; Male; Mesocricetus; Mouth Neoplasms; Precancerous Conditions; RNA, Messenger; Transforming Growth Factor beta

1992
The effects of continuous exposure to epidermal growth factor on the spontaneous transformation of cultured rat liver epithelial cells.
    The American journal of pathology, 1992, Volume: 140, Issue:1

    A long-term continuous exposure to epidermal growth factor (EGF) enhanced the tumorigenicity of spontaneously transformed cells arising in a clonal population of normal cultured rat liver epithelial cells propagated in a selective growth condition. Lengthy EGF exposure also induced the expression of several phenotypes that differed from the phenotypes of rat liver epithelial cells transformed spontaneously in the absence of EGF. Epidermal growth factor treatment caused consistently an enhancement of the constitutive mRNA expression of transforming growth factor-alpha (TGF-alpha), but not of the EGF receptor and transforming growth factor-beta. The overexpression of TGF-alpha persisted in cell lines derived from tumors formed by the EGF-treated transformed cells. These tumors also exhibited high metastatic incidence and ductal cell differentiation. In contrast, untreated spontaneously transformed cells formed non-metastatic tumors with hepatocellular differentiation. These results suggest that long-term, continuous exposure to EGF/TGF-alpha may modulate the phenotypic expressions of neoplastic transformation.

    Topics: Animals; Blotting, Northern; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; Epithelium; ErbB Receptors; Gene Expression Regulation, Neoplastic; Liver; Liver Neoplasms; Neoplasm Metastasis; Phenotype; Proto-Oncogene Proteins c-myc; Proto-Oncogene Proteins p21(ras); Rats; Rats, Inbred F344; RNA, Messenger; Transforming Growth Factor alpha; Transforming Growth Factor beta

1992
Effect of transforming growth factor-beta 1 and -beta 2 on the proliferation of Burkitt lymphoma and lymphoblastoid cell lines.
    Growth factors (Chur, Switzerland), 1991, Volume: 4, Issue:2

    We tested the effect of transforming growth factor (TGF)-beta 1 and TGF-beta 2 on the proliferation of human B cell lines. The panel was selected to give information whether (1) their origin, (2) their phenotype, (3) their Epstein-Barr virus (EBV) carrier state, influence their responsiveness. The growth of lymphoblastoid cell lines (LCL) was not inhibited by TGF-beta 1. The EBV-carrying Burkitt lymphoma (BL) lines, Daudi, Jijoye, Rael but not Raji were inhibited. Three EBV-negative BL lines and the majority of their converted sublines were sensitive. The cell lines tested expressed TGF-beta receptors and TGF-beta 1 transcripts. The proliferation of EBV-infected B cells was inhibited by TGF-beta, their sensitivity decreased, however, after 3 days. The results suggest that the activation state of the B cells is decisive for TGF-beta sensitivity and EBV influences it indirectly by changing the cell phenotype.

    Topics: B-Lymphocytes; Burkitt Lymphoma; Cell Line; Cell Transformation, Neoplastic; Herpesvirus 4, Human; Humans; Lymphocyte Activation; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

1991
Expression of a new type high molecular weight receptor (type V receptor) of transforming growth factor beta in normal and transformed cells.
    Biochemical and biophysical research communications, 1991, Aug-30, Volume: 179, Issue:1

    A new type high molecular weight receptor (type V receptor) of transforming growth factor beta (TGF-beta) was recently purified from bovine liver plasma membranes and appears not to be related to receptors previously described for TGF-beta (Pauline O'Grady, Ming-Der Kuo, Joseph J. Baldassare, Shuan Shian Huang and Jung San Huang [1991] J. Biol. Chem. 266:8583-8589). This type V receptor may be important in the regulation of cell growth by TGF-beta. We examined its distribution in a wide range of normal and transformed cells. The type V receptor was found to be expressed in many normal cells including cells of epithelial, endothelial, fibroblastic and chondrocytic origins. However, a number of human epithelial tumor cells (5 out of 6 examined) did not express detectable levels of the type V TGF-beta receptor. These results suggest that loss of the type V receptor may potentially contribute to the transformed state of certain epithelial tumor cells.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Humans; Molecular Weight; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Reference Values; Transforming Growth Factor beta

1991
Induction of transforming growth factor beta 1 resistance by the E1A oncogene requires binding to a specific set of cellular proteins.
    Proceedings of the National Academy of Sciences of the United States of America, 1991, Apr-15, Volume: 88, Issue:8

    Transforming growth factors beta (TGF-beta s) are potent inhibitors of epithelial cell growth in culture and might play a similar role in vivo. Several studies have suggested that acquisition of TGF-beta resistance is an important step in epithelial tumor development. Here, we show that resistance to TGF-beta 1 growth inhibition can be induced by transformation of keratinocytes with the E1A, but not the ras, oncogene. Mutational analysis revealed that these effects closely correlate with the ability of E1A proteins to bind to the retinoblastoma gene product (p105) as well as to three other cellular proteins (p60, p107, and p300). Only partial resistance to TGF-beta 1 growth inhibition was elicited by E1A mutants that bind to a subset of proteins, whereas complete resistance was induced by E1A mutants that bind to all four proteins together. Total protection against TGF-beta growth inhibition was also induced by concomitant introduction into cells of an E1A mutant binding to the p60/p105/p107 proteins and one binding to p300. In parallel with these effects, epidermal transglutaminase, a marker of keratinocyte differentiation, was induced by TGF-beta in control but not in E1A-transformed cells. TGF-beta 1 receptor levels were only partially down-modulated by an intact E1A gene and not significantly affected by the various truncated mutants. Thus, the ability of E1A to induce TGF-beta resistance depends on its ability to bind, and presumably inactivate, several cellular proteins that may be involved in transmission of the TGF-beta signal and seem to act downstream from its receptor(s).

    Topics: Adenovirus Early Proteins; Animals; Cell Differentiation; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA Mutational Analysis; Down-Regulation; Enzyme Induction; Keratinocytes; Mice; Oncogene Proteins, Viral; Protein Binding; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Retinoblastoma Protein; Transforming Growth Factor beta; Transglutaminases

1991
Expression of protein disulfide isomerase in cultured rat liver epithelial cells is unrelated to the growth inhibitory effect of TGF-beta 1.
    Biochemical and biophysical research communications, 1991, Jan-31, Volume: 174, Issue:2

    The effect of TGF-beta 1 treatment on the level of protein disulfide isomerase (PDI) mRNA in normal and chemically or spontaneously transformed rat liver epithelial cell lines was investigated. TGF-beta 1 at 1 or 10 ng/ml concentrations did not significantly decrease the mRNA level of PDI at 4 or 24 hours after exposure to TGF-beta 1, irrespective whether the cell line was sensitive or resistant to the growth-inhibitory effect of TGF-beta 1 at these concentrations. The results indicate that in normal or neoplastic rat liver epithelial cells, the expression of PDI is unrelated to the growth inhibitory effect of TGF-beta 1.

    Topics: Animals; Biological Transport; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA Probes; DNA Replication; Epithelium; Isomerases; Liver; Nucleic Acid Hybridization; Protein Disulfide-Isomerases; Rats; Rats, Inbred F344; RNA; Thymidine; Transforming Growth Factor beta

1991
TGF-beta 1 and skin carcinogenesis: antiproliferative effect in vitro and TGF-beta 1 mRNA expression during epidermal hyperproliferation and multistage tumorigenesis.
    Molecular carcinogenesis, 1991, Volume: 4, Issue:2

    The role of transforming growth factor-beta 1 (TGF-beta 1) in multisage carcinogenesis in mouse skin was assessed by studying its growth inhibitory effects on nontumorigenic and tumorigenic keratinocytes and by examining its mRNA expression in vitro and during epidermal hyperproliferation and multistage carcinogenesis. While growth of primary basal keratinocytes was inhibited by TGF-beta 1 in doses as low as 0.1 ng/mL, the immortal keratinocyte line MCA/3D ("putatively initiated" cells) responded to TGF-beta 1 with slightly reduced sensitivity, and the papilloma-producing keratinocyte line 308 was considerably less sensitive. In contrast, the squamous carcinoma cell line Carc B was completely nonresponsive, and two other tumorigenic cell lines (PDV and PDVC57) were sensitive to growth inhibition by TGF-beta 1. Steady-state levels of TGF-beta 1 mRNA were high in all the malignant cell lines and in line 308 papilloma cells, but low in primary basal cells and in the nontumorigenic keratinocyte lines V2, Reb1, and MCA/3D. Our in vivo studies showed that tumor promoters, but not mitogenic or weak hyperplasiogenic agents, were able to induce transient expression of TGF-beta 1 mRNA in mouse epidermis. A constitutive overexpression of TGF-beta 1 mRNA was observed in malignant carcinomas but not in the benign premalignant lesions, indicating that overexpression may be associated with malignant progression.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Animals, Newborn; Blotting, Northern; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; DNA; DNA Replication; DNA, Neoplasm; Female; Gene Expression; Keratinocytes; Mice; Mice, Inbred Strains; Phorbol Esters; RNA; RNA, Messenger; RNA, Neoplasm; Skin; Skin Neoplasms; Skin Physiological Phenomena; Tetradecanoylphorbol Acetate; 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
Development of resistance to the growth inhibitory effects of transforming growth factor beta 1 during the spontaneous transformation of rat liver epithelial cells.
    Cancer research, 1991, Nov-01, Volume: 51, Issue:21

    The temporary maintenance of a rat liver epithelial cell population at confluence before passaging followed by periods of rapid proliferation resulted in the generation of spontaneous transformants after about 108 population doublings. The appearance of morphologically aberrant transformants correlated directly with an increased resistance of the population to the growth inhibitory effects of transforming growth factor beta 1 (TGF-beta 1). Clonal cell lines derived from the transformants were resistant to TGF-beta 1 dependent inhibition of DNA synthesis. These cell lines were also highly tumorigenic and aneuploid, with characteristic gross chromosomal abnormalities, and they expressed a number of phenotypic markers common to rat liver epithelial cells transformed by oncogenes or chemicals. In contrast, apparently normal looking cell lines cloned from the same population were nontumorigenic and near diploid, with few chromosomal abnormalities, and they were as sensitive to TGF-beta 1 as early passage normal rat liver epithelial cells. Morphologically normal late passage rat liver epithelial cells were sensitive to transformation by the DNA hypomethylating agent 5-aza-2-deoxycytidine, in contrast to earlier passage cells, and this transformation was accompanied by the development of resistance to the growth inhibitory effects of TGF-beta 1. These findings suggest that acquisition of resistance to the effects of growth inhibitors such as TGF-beta 1 is an important and possibly essential stage in the spontaneous transformation of rat liver epithelial cells.

    Topics: Animals; Blotting, Northern; Cell Division; Cell Line; Cell Transformation, Neoplastic; Clone Cells; DNA Probes; DNA Replication; Drug Resistance; Epithelial Cells; Epithelium; Karyotyping; Liver; Oncogenes; Poly A; Rats; Restriction Mapping; RNA; RNA, Messenger; Transforming Growth Factor beta

1991
Experimental oncogene induced prostate cancer.
    Cancer surveys, 1991, Volume: 11

    The mouse prostate reconstitution model exploits the ability of the fetal urogenital sinus to differentiate into a mature prostate when grafted under the renal capsule of an adult isogenic male host. By use of a recombinant retroviral vector, the ras and myc oncogenes are introduced singly or in combination into the fetal urogenital sinus--resulting in distinct phenotypes of prostatic pathology: dysplasia (caused by ras), hyperplasia (caused by myc) and frank carcinomas (caused by a combination of ras+myc). This unique experimental model creates in vivo conditions that mimic the natural initiation and progression of cancer. An expanded MPR protocol allows restricted retrovirus infection of the mesenchyme or epithelial compartments to evaluate paracrine activities. It enables almost unparalleled flexibility in addressing fundamental questions in prostate cancer. We have identified genetic variance in the susceptibility to tumour induction between two different strains of mice (mimicking the observation of racial variability in the predisposition to clinical prostate cancer). The MPR model supports data from other tumour models and implicates TGF-beta 1 and TGF-beta 3 as being strongly associated with tumour progression. Finally, with this model, we have established clonal prostate adenocarcinomas to study directly the affects of castration on gene expression. Not only are TGF-beta 1 and TGF-beta 3 mRNA levels increased in association with malignancy but they are also further enhanced by castration treatment. Based on these experimental studies, we believe that TGF-beta 1 and TGF-beta 3 expression strongly influences the progression of prostate cancer. This information will hopefully impact on the development of more effective therapy for this important malignancy.

    Topics: Adenocarcinoma; Androgens; Animals; Blotting, Northern; Castration; Cell Transformation, Neoplastic; Disease Models, Animal; Genes, myc; Genes, ras; Male; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Neoplasms, Experimental; Prostatic Neoplasms; RNA; Transforming Growth Factor beta

1991
Early induction of ribonucleotide reductase gene expression by transforming growth factor beta 1 in malignant H-ras transformed cell lines.
    The Journal of biological chemistry, 1991, Dec-15, Volume: 266, Issue:35

    Previous investigations have indicated that the suppression of proliferation by transforming growth factor (TGF) beta 1 is often lost upon cellular transformation, and that proliferation of some tumors is stimulated by TGF-beta. The present study provides the first observation of a link between TGF-beta 1 regulation of this process and alterations in the expression of ribonucleotide reductase, a highly controlled rate-limiting step in DNA synthesis. A series of radiation and T24-H-ras-transformed mouse 10T1/2 cell lines exhibiting increasing malignant potential was evaluated for TGF-beta 1 induced alterations in ribonucleotide reductase M1 and M2 gene expression. Early increases in M1 and/or M2 message and protein levels were observed only in malignant cell lines. The TGF-beta 1 induced changes in M1 and/or M2 gene expression occurred prior to any detectable changes in the rates of DNA synthesis, supporting the novel concept that ribonucleotide reductase gene expression can be elevated by TGF-beta 1 without altering the proportion of cells in S phase. T24-H-ras-transformed 10T1/2 cells were transfected with a plasmid containing the coding region of TGF-beta 1 under the control of a zinc-sensitive metallothionein promoter. When these cells were cultured in the presence of zinc, a large induction of TGF-beta 1 message was observed within 1 h. Both M1 and M2 genes were also induced, with increased mRNA levels appearing 2 h after zinc treatment, or 1 h after TGF-beta 1 message levels were clearly elevated. In total, the data suggests a mechanism of autocrine stimulation of malignant cells by TGF-beta 1, in which early alterations in the regulation of ribonucleotide reductase may play an important role.

    Topics: Animals; Blotting, Northern; Blotting, Western; Cell Line; Cell Transformation, Neoplastic; DNA Replication; Gene Expression Regulation, Enzymologic; Genes, ras; Glyceraldehyde-3-Phosphate Dehydrogenases; Mice; Ribonucleotide Reductases; RNA, Messenger; Transforming Growth Factor beta

1991
Recessive (mediator-) revertants from c-H-ras oncogene-transformed NIH 3T3 cells: tumorigenicity in nude mice and transient anchorage and serum independence of the recovered tumor cells in culture.
    Journal of cellular physiology, 1991, Volume: 149, Issue:2

    We have reported earlier the isolation of two recessive, serum- and anchorage-dependent revertants (R116 and R260) from a c-H-ras oncogene-transformed NIH 3T3 line. In both revertants, the oncogene was fully expressed and fusion of either revertant with (untransformed) NIH 3T3 cells, or of the two revertants with one another, resulted in transformed progeny. These, and other data, indicated that the transforming activity of the oncogene was impaired in the two revertants in consequence of defects in distinct genes needed to mediate this activity. We report here that neither revertant could be re-transformed by the K-ras or N-ras oncogene (though they could be re-transformed by several other oncogenes). The two revertants turned out to be tumorigenic in nude mice (though less so than the parental transformed cells). The tumor cells, as recovered, formed foci and had a transformed morphology and a greatly diminished serum and anchorage dependence. Growth of the cells in culture (for 20 passages) resulted in their regaining the characteristics (i.e., anchorage and serum dependence) of cultured R116 and R260 cells. Proliferation of the cells in nude mice was not accompanied by a change in the level of ras oncogene expression or in gene amplification, at least as manifested in the lack of appearance of double-minute chromosomes. The addition of the growth factors TGF alpha and beta to the medium of either revertant did not support anchorage-independent growth.

    Topics: 3T3 Cells; Animals; Blood; Cell Adhesion; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Culture Media; Gene Amplification; Genes, ras; Genes, Recessive; Mice; Mice, Nude; Neoplasm Transplantation; Transfection; Transforming Growth Factor alpha; Transforming Growth Factor beta

1991
Telomeric associations and consistent growth factor overexpression detected in giant cell tumor of bone.
    Cancer genetics and cytogenetics, 1991, Oct-15, Volume: 56, Issue:2

    Tumor specimens from 15 patients with giant cell tumor (GCT) of bone were cytogenetically analyzed. A subset of five individuals had tumor cells harvested and polyadenylated RNA isolated. Multiple Northern blots were performed utilizing radiolabeled probes for the growth factors TGF beta 1, TGF beta 2, TGF beta 3, and TGF alpha (TGF, transforming growth factor). RNAs from other types of neoplasms and nonneoplastic cells were examined as controls. The most consistent cytogenetic abnormality detected involved multiple telomeric associations (TAs), most frequently involving the terminus of the long arm of chromosome 19 (19q). Northern blot analysis revealed a consistent expression of TGF beta 1 and TGF beta 2 with an inconsistent mRNA expression for the other TGFs. There was a relative overexpression of mRNA for TGF beta 2. The gene location for TGF beta 1 is near the 19q terminus and thus it is speculated that TGF beta may play a role in the neoplastic transformation of GCT.

    Topics: Adult; Blotting, Northern; Bone Neoplasms; Cell Transformation, Neoplastic; Child; Chromosomes, Human, Pair 19; Female; Gene Expression; Giant Cell Tumors; Humans; Male; Middle Aged; Radioligand Assay; Telomere; Transforming Growth Factor alpha; Transforming Growth Factor beta

1991
Altered response to growth factors or retinoic acid in phenotypic transformation of normal rat kidney cells expressing human c-fos gene.
    Biochemical and biophysical research communications, 1991, Dec-31, Volume: 181, Issue:3

    Anchorage-independent growth of normal rat kidney (NRK) fibroblast in soft agar depends on both transforming growth factor beta (TGF beta) and epidermal growth factor (EGF). To examine whether c-fos protein is involved in phenotypic transformation of NRK cells, we have transfected and isolated several NRK cell lines that carry the human c-fos gene fused to the metallothionein IIA promoter. A transfectant, Nf-1, had constitutive levels of the human c-fos expression. Anchorage-independent growth of Nf-1 was already stimulated by EGF alone, and the colony sizes of Nf-1 were comparable to those of the parental NRK in the presence of both EGF and TGF beta. Anchorage-independent growth of NRK could be observed in the presence of TGF beta or retinoic acid or platelet derived growth factor (PDGF) and EGF. No growth of NRK in soft agar appeared when basic fibroblast growth factor (bFGF) and EGF were present. By contrast, anchorage-independent growth of Nf-1 was surprisingly enhanced by EGF and TGF beta or retinoic acid or PDGF or bFGF. Expression of the human c-fos gene may compensate the signal to phenotypic transformation induced by TGF beta as well as retinoic acid or PDGF or bFGF.

    Topics: Animals; Blotting, Northern; Blotting, Southern; Blotting, Western; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblast Growth Factor 2; Genes, fos; Growth Substances; Humans; Kidney; Nuclear Proteins; Phenotype; Plasmids; Platelet-Derived Growth Factor; Rats; Recombinant Proteins; Transfection; Transforming Growth Factor beta; Tretinoin

1991
Immediate early gene responses of NIH 3T3 fibroblasts and NMuMG epithelial cells to TGF beta-1.
    Growth factors (Chur, Switzerland), 1991, Volume: 5, Issue:4

    Transforming growth factor beta has a wide range of physiological effects on cell growth and metabolism. We have previously reported on the rapid induction of jun transcription factors in TGF beta-treated cells. Here we show that the early genomic response to TGF beta-1 includes activation of a broad spectrum of serum-inducible genes both in NIH 3T3 fibroblasts and in NMuMG epithelial cells, which are growth-stimulated and growth-inhibited by TGF beta, respectively. Of particular interest is the presence of a putative nuclear DNA-binding receptor (N10) and zinc finger transcription factors (Krox 20 and Krox 24) among the TGF beta-induced genes. In addition to the stimulatory effects of TGF beta, expression of a few genes including c-myc is decreased in both types of cells. In cells transformed by neu or ras oncogenes the immediate early mRNA responses to TGF beta are deregulated. Our results suggest that certain transcription factors are required for both positive and negative regulation of cell proliferation by TGF beta, and that their relative concentrations may determine the subsequent cellular responses.

    Topics: 3T3 Cells; Animals; Blotting, Northern; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; Epithelium; Gene Expression Regulation; Mice; Oncogenes; RNA, Messenger; Signal Transduction; Transforming Growth Factor beta

1991
Correlated abnormalities of transforming growth factor-beta 1 response and p53 expression in thyroid epithelial cell transformation.
    Molecular and cellular endocrinology, 1991, Volume: 76, Issue:1-3

    Using the thyroid follicular cell as a model for multi-stage carcinogenesis, we have investigated the role of two potential negative growth regulators ('anti-oncogenes') in epithelial tumour progression--transforming growth factor-beta 1 (TGF beta 1) and p53. Normal follicular cells, as expected, showed marked growth inhibition in response to TGF beta 1. Adenoma cells were equally inhibited. In contrast, spontaneously and SV40-immortalised follicular cell lines showing features of malignant transformation (notably loss of growth factor dependence) had lost all responsiveness to TGF beta 1, accompanied by a partial loss of its receptors. p53 protein was below detectable limits in normal and in adenoma cells but in contrast very high levels were observed in all three transformed lines. In the SV40-immortalised cells, this was expected in view of the known stabilising effect of the viral large T protein. In the spontaneous line we found strong evidence for point mutation of p53, which is known to have the same effect. Both mechanisms result in loss of p53 tumour suppressor function despite increased protein content. We conclude that loss of inhibition by TGF beta and inactivation of p53 are important steps in in vitro immortalisation and/or in vivo tumour progression in human thyroid follicular cells, and speculate that p53 may mediate or be required for the inhibitory signal normally induced by TGF beta 1.

    Topics: Adenoma; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Epithelium; Humans; Immunohistochemistry; Precipitin Tests; Thyroid Gland; Thyroid Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Protein p53

1991
Synergistic inhibition of phorbol ester-induced transformation of JB6 cells by transforming growth factor-beta and retinoic acid.
    Cancer research, 1991, Feb-15, Volume: 51, Issue:4

    Transforming growth factor-beta (TGF-beta) plays a complex role as a regulator of proliferation and differentiation of many cell types, including cells of epithelial origin. In this study, we examined whether TGF-beta, alone or in combination with retinoic acid, was able to inhibit the transformation of the murine epidermal cell line JB6. When treated with phorbol myristate acetate (PMA) and other tumor promoters, the nontumorigenic and anchorage-dependent JB6 cells acquired a tumor phenotype, as shown by the acquisition of tumorigenicity and anchorage independence. We found that TGB-beta inhibited the PMA-induced transformation of a subclone of JB6 cells. The effect of TGF-beta was due to an anti-transformation promoting activity, rather than to generalized growth inhibition, since TGF-beta neither inhibited the growth of monolayer cultures of JB6 cells, nor affected the colony-forming efficiency in agar of the JB6-derived permanently transformed RT101 cell line. TGF-beta was synergistic with retinoic acid, a known anti-tumor promoter, in inhibiting the PMA-induced transformation of JB6 cells. Examination of TGF-beta receptor expression on JB6 cells, by both binding and affinity labeling, showed that treatment with PMA significantly decreased TGF-beta receptor expression while retinoic acid counteracted this effect of PMA, thus suggesting that the synergy between retinoic acid and TGF-beta may be due, at least in part, to modulation of TGF-beta receptor expression. TGF-beta, therefore, appears to function as an incomplete antipromoter whose action can be permitted and/or complemented by retinoic acid. Our data demonstrating that TGF-beta has anti-transformation promoting activity suggest that TGF-beta plays a role in maintaining homeostasis of epithelial cells, not only by regulating cell proliferation and differentiation, but also by counteracting events that lead to malignant transformation.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Colony-Forming Units Assay; Dose-Response Relationship, Drug; Drug Synergism; Mice; Mice, Inbred BALB C; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Tetradecanoylphorbol Acetate; Transforming Growth Factor beta; Tretinoin

1991
Tumor-promoting activity of Epstein-Barr-virus-inducing factor transforming growth factor type beta (EIF/TGF-beta) is due to the induction of irreversible transformation.
    International journal of cancer, 1991, Apr-01, Volume: 47, Issue:6

    EBV-inducing factor/transforming growth factor type beta (EIF/TGF-beta) exhibits tumor-promoting activity for C3H-10T1/2 mouse fibroblasts in vitro. Treatment of C3H 10 T 1/2 fibroblasts seeded at low density with initiating doses of UV light, followed by culture in the presence of EIF/TGF-beta leads to the appearance of foci of stably transformed cells that have the potential to grow in soft agar. The promoting effect depends both on the dose of EIF/TGF-beta applied and its continuous presence for 2 to 3 weeks. In addition to its procarcinogenic effect in tumor promotion, EIF/TGF-beta exhibits a strong negative effect on transformed cells surrounded by normal cells, indicating a dual role of EIF/TGF-beta in carcinogenesis. The lack of completely transformed individual cells in the initiated cell population and the negative effect of EIF/TGF-beta on transformed cells in contact with normal cells exclude any possible explanation of the tumor-promoting effect of EIF/TGF-beta as being the result of a selection process due to the establishment of growth advantages for cells transformed by the initiator. The data, in fact, indicate that tumor promotion by EIF/TGF-beta implies the stable acquisition of distinct qualitative changes by the cells.

    Topics: Animals; Blood Platelets; Cell Line; Cell Transformation, Neoplastic; Dose-Response Relationship, Drug; Herpesvirus 4, Human; Humans; Mice; Tetradecanoylphorbol Acetate; Transforming Growth Factor beta

1991
Differential expression of transforming growth factor-beta 1 (TGF-beta 1) receptors in murine myeloid cell lines transformed with oncogenes. Correlation with differential growth inhibition by TGF-beta 1.
    The Journal of biological chemistry, 1991, May-25, Volume: 266, Issue:15

    Transforming growth factor-beta 1 (TGF-beta 1) is a pleiotropic polypeptide hormone known to play an important role as a modulator of hematopoietic processes in human and murine cells. One of the characteristics of TGF-beta 1 is the ability to inhibit the growth of several cell types, including cells of the myeloid lineage. To study the mechanism by which TGF-beta 1 inhibits the growth of myeloid cells, we have used three murine myeloid cell lines, the parental interleukin-3-dependent 32D-123 cell line and two retrovirally infected interleukin-3-independent cell lines (32D-abl, 32D-src), all of which are growth inhibited by TGF-beta 1. Each of these oncogene-transfected cells expresses a greater number of TGF-beta 1 receptors than the parental cell line and responds to TGF-beta 1 with increased sensitivity; 32D and 32D-src cells are 2- and 58-fold more sensitive to TGF-beta 1 inhibition than the parental cell line (ED50 = 35 pM). Both 32D-abl- and 32D-src-transformed cell lines expressed higher levels of the 65- and 85-kDa TGF-beta 1 receptor species than did the parental cells. We observed a correlation between the greater sensitivity of 32D-src cells to TGF-beta 1 and the more rapid down-modulation and reappearance of cell surface TGF-beta 1 receptors on 32D-src cells. Thus, the level of TGF-beta 1 receptor expression and rate of reexpression both have a crucial regulatory effect on the functional activity of the TGF-beta 1 ligand.

    Topics: Animals; Autoradiography; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cross-Linking Reagents; Electrophoresis, Polyacrylamide Gel; Interleukin-3; Mice; Oncogenes; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta

1991
Natural killer target molecules associated with the transformation of the oncogene-transfected fibroblast.
    Cancer research, 1991, Aug-15, Volume: 51, Issue:16

    Cell surface antigens, the expression of which is highly enhanced along with the transformation of cells, were analyzed. W14 and W31, EJ-ras oncogene-induced transformants of a WKA rat fetus-derived fibroblast WFB, strongly expressed several transformation-associated antigens as defined by monoclonal antibodies 109, 061, and 081. These monoclonal antibodies recognized Mr 86,000, 62,000, and 101,000 molecules, each composed of a single polypeptide chain. The expression of these transformation-associated antigens was negligible on parental WFB cells. Transforming growth factor-beta could enhance the expression of all of these transformation-associated antigens, but platelet-derived growth factor could only enhance the Mr 86,000 kd molecule expression. In the cytotoxicity assays, poly-I:C-induced rat splenic NK cells were cytotoxic to W14 and W31, but not to WFB. The data also showed that the cytotoxicity by these NK cells against NK-sensitive YAC-1 cells was absorbed with the addition of W14, W31, platelet-derived growth factor, or transforming growth factor-beta-stimulated WFB cells. This indicates that NK cells may recognize common target antigens that are expressed among these target cells. It was also indicated that Mr 86,000 and 62,000 molecules were strongly involved in this cytotoxicity, possibly as the target antigens, since F(ab')2 fragments of monoclonal antibodies 109 and 061 strongly inhibited the cytotoxicity. The addition of monoclonal antibody 109, but not 061, inhibited the cytotoxicity even at 60 min after mixing with the effector and target cells, suggesting that the Mr 86,000 molecule may participate in the lethal hit phase of cytotoxicity by NK cells. These data may indicate that some, but not all, transformation-associated antigens are virtually important in the antitumor surveillance mechanisms by the host effector cells, such as NK cells.

    Topics: Animals; Antibodies, Monoclonal; Antigens, Surface; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cytotoxicity, Immunologic; Fibroblasts; Genes, ras; Killer Cells, Natural; Kinetics; Platelet-Derived Growth Factor; Poly I-C; Rats; Transforming Growth Factor beta

1991
[Growth inhibitors and feedback regulation of cell proliferation].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 1991, Volume: 36, Issue:7

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Feedback; Humans; Transforming Growth Factor beta

1991
Effects of type-beta 1 transforming growth factor on the proliferation and differentiation of mouse myelomonocytic leukemia cells (M1).
    Experimental cell research, 1991, Volume: 196, Issue:1

    Murine myelomonocytic leukemia M1 cells have been used to examine the effects of type-beta 1 transforming growth factor (TGF-beta 1) on cellular proliferation and differentiation in monocyte-macrophage lineage. TGF-beta 1 inhibited immature M1 cell growth due to a general slowdown of the cell cycle, without arrest at any specific point. Ten nanograms per milliliter TGF-beta 1 completely suppressed phagocytic activity and adhesion to the dish surface and partially inhibited the expression of Fc receptors and vimentin during the differentiation of M1 cells induced by IL-6. IL-6-induced declines in the expression of c-myc mRNA and in the accumulation of G0/G1 cells were also partially blocked by TGF-beta 1. When treated concurrently with IL-6 and TGF-beta 1, approximately 50% of M1 cells were morphologically converted to promonocyte or monocyte-like cells, which did not exhibit the characteristics of mature macrophages. Although pretreatment with TGF-beta 1 also inhibited the IL-6-induced phagocytic activity, this inhibition was reversible. Once TGF-beta 1 was removed from the culture medium after 72 h of incubation with IL-6, the kinetics of differentiation induced by IL-6 were faster in pretreated cells than in nonpretreated cells. TGF-beta 1 appears to inhibit the IL-6 induced conversion of M1 cells at the intermediate stage of monocytic differentiation.

    Topics: Animals; Blotting, Northern; Cell Cycle; Cell Division; Cell Transformation, Neoplastic; Cytokines; Interleukin-6; Leukemia, Experimental; Leukemia, Myelomonocytic, Acute; Macrophages; Mice; Monocytes; Phagocytosis; Proto-Oncogene Proteins c-myc; Receptors, Fc; RNA, Messenger; Transforming Growth Factor beta; Tumor Cells, Cultured; Vimentin

1991
Differential sensitivity of human colonic adenoma and carcinoma cells to transforming growth factor beta (TGF-beta): conversion of an adenoma cell line to a tumorigenic phenotype is accompanied by a reduced response to the inhibitory effects of TGF-beta.
    Oncogene, 1991, Volume: 6, Issue:8

    The growth of three non-tumorigenic human colonic adenoma cell lines, designated AA/C1, RG/C2 and RR/C1, was inhibited by low concentrations of transforming growth factor beta (TGF-beta) (0.05-0.5 ng ml-1). However, the growth of five human colon cancer cell lines under identical conditions was resistant to high concentrations of TGF-beta (2-10 ng ml-1). This is the first report of well-characterized premalignant human colonic cells showing sensitivity to TGF-beta. The TGF-beta-sensitive adenoma cell line AA/C1 was derived from a relatively large adenoma with a K-ras gene mutation and represents a relatively late-stage adenoma, indicating that loss of response to TGF-beta occurs at a relatively late stage in colorectal carcinogenesis and that the presence of a ras gene mutation does not necessarily confer resistance to TGF-beta. Of further interest, the RG/CZ cell line has a p53 mutation showing that p53 mutations do not necessarily lead to TGF-B insensitivity. Furthermore, in this paper we show that the conversion of the AA/C1 adenoma cell line to a tumorigenic phenotype [Williams et al., (1990) Cancer Res., 50, 4724] is accompanied by a reduced response to the growth-inhibitory effects of TGF-beta up to 10 ng ml-1. Reduced responsiveness to the inhibitory effects of TGF-beta may be an important event in the loss of growth control in colorectal carcinogenesis.

    Topics: Adenoma; Carcinoma; Cell Line, Transformed; Cell Transformation, Neoplastic; Colorectal Neoplasms; Dose-Response Relationship, Drug; Drug Resistance; Genes, ras; Humans; Mutation; Phenotype; Precancerous Conditions; Transforming Growth Factor beta; Tumor Cells, Cultured

1991
Loss of growth control by TGF-beta occurs at a late stage of mouse skin carcinogenesis and is independent of ras gene activation.
    Oncogene, 1991, Volume: 6, Issue:8

    The relationship between the expression of a mutant ras gene in epithelial cells and loss of responsiveness to the negative effects of transforming growth factor beta (TGF-beta) is presently unclear. We have investigated this question using a series of cell lines derived from benign and malignant mouse skin tumours which express mutant forms of the H-ras gene. Immortalised, non-tumorigenic mouse epidermal cells respond to TGF-beta by cessation of growth, whereas in a series of malignant carcinoma lines the response was substantially reduced. Introduction of a mutant H-ras gene into the immortalised cells did not lead to any appreciable change in TGF-beta responsiveness, suggesting that initiation of carcinogenesis by ras mutation does not directly alter growth control by this pathway. Of two non-tumorigenic papilloma lines tested which had mutant H-ras genes, one retained complete sensitivity to TGF-beta, whereas the other showed a similar response to carcinomas. We conclude that growth control by TGF-beta is lost at a relatively late stage of carcinogenesis in this system, and is independent of ras gene activation.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; DNA, Neoplasm; Epithelium; Gene Expression Regulation, Neoplastic; Genes, ras; Mice; Mutation; Skin Neoplasms; Thymidine; Transcriptional Activation; Transforming Growth Factor beta; Tritium; Tumor Cells, Cultured

1991
Elevated transforming growth factor-beta 1 and beta 3 mRNA levels are associated with ras + myc-induced carcinomas in reconstituted mouse prostate: evidence for a paracrine role during progression.
    Molecular endocrinology (Baltimore, Md.), 1991, Volume: 5, Issue:4

    Mouse prostate reconstitution is a useful model for studying the progression of ras + myc-induced carcinomas. When these oncogenes were introduced into both the epithelial and the mesenchymal compartments, poorly differentiated adenocarcinomas resulted. Restricted introduction of both oncogenes into the epithelium produced epithelial hyperplasia. Malignancies were produced in two out of 17 cases of selectively transformed epithelium, suggesting that the hyperplastic condition represents a premalignant phenotype. Restricted introduction of both oncogenes into the mesenchyme produced only mesenchymal dysplasia. Transforming growth factor-beta 1 (TGF-beta 1) and beta 3 (TGF-beta 3) mRNA levels were elevated in the ras + myc-induced carcinomas when compared to the normal controls or to the epithelial hyperplasias. In contrast, TGF-beta 2 mRNA levels were similar in all control and ras + myc-induced carcinomas. Elevated TGF-beta 1 mRNA levels were also found in mesenchymal dysplasia pointing to a potential paracrine activity by the ras + myc transformed mesenchyme. We conclude that elevated TGF-beta 1 and beta 3 are correlated with progression to malignancy and that mesenchyme derived TGF-beta 1 may play an important role in the promotion of ras + myc-induced carcinomas in this model system.

    Topics: Animals; Blotting, Northern; Cell Transformation, Neoplastic; Epithelium; Genes, myc; Genes, ras; Male; Mesoderm; Mice; Prostatic Neoplasms; RNA, Messenger; Transforming Growth Factor beta

1991
A comparison of the growth promoting properties of ascitic fluids, cyst fluids and peritoneal fluids from patients with ovarian tumours.
    British journal of cancer, 1991, Volume: 63, Issue:1

    The growth promoting properties of ascitic fluids, cyst fluids and peritoneal fluids from patients with ovarian malignancy, benign ovarian tumours and non-tumour related gynaecological conditions have been investigated using an ovarian carcinoma cell line (OAW 42), mesothelial cells (58MC) and rat kidney cells (NRK-49F). Colony stimulating activity (CSA) for tumour cells and transforming activity (TA) for mesothelial cells were weakly correlated, but whereas elevated TA was tumour-associated, CSA was not. However, TA was not cancer-associated and, although the difference between the mean TA values of benign and malignant cyst fluids was of borderline significance, some benign cyst fluids from cystadenomas showed high TA values. Higher levels of TA in the cystadenomas showed a significant correlation with the menopausal status of the patient and higher levels of TA in the malignant cyst fluid/peritoneal fluid groups were associated with more advanced disease. Results indicated that some fluids contained TGF-beta-like activity, but there was no direct evidence for the presence of TGF-alpha/EGF-like activity in the fluids. Heparin inhibited clonogenic growth of tumour cells but not mesothelial cells. The reduced CSA which was observed after treatment of fluids with both heparin and thrombin implicated coagulation factors in the manifestation of CSA. It was concluded that CSA in the fluids was due, at least partly, to fibrin coagulation, and TA was due to unknown growth factor(s) which may include TGF-beta-like activity. The results are discussed in the context of the aetiology of ovarian carcinoma, and the possible clinical significance of TA.

    Topics: Animals; Ascitic Fluid; Blood Coagulation Factors; Body Fluids; Cell Division; Cell Transformation, Neoplastic; Colony-Stimulating Factors; Cysts; Epidermal Growth Factor; Female; Growth Substances; Heparin; Humans; Mice; Ovarian Neoplasms; Thrombin; Transforming Growth Factor beta; Tumor Cells, Cultured

1991
Mechanisms controlling growth of hepatocytes in primary culture.
    Digestive diseases and sciences, 1991, Volume: 36, Issue:4

    Mature hepatocytes in primary culture express most of the functions and hormonal responsiveness seen in normal liver studied in vivo. The growth of hepatocytes in culture is regulated by various growth factors. We have identified a hepatocyte growth factor that is isolated from rat platelets. We found that rat platelets also contain a growth inhibitor, transforming growth factor-beta which is secreted as a latent molecule. Its latency is due to its binding with a masking protein. Growth of hepatocytes is also suppressed by interleukin-1 (IL-1) and IL-6. Moreover, the growth and functions of liver cells in culture are regulated reciprocally by cell density: at higher cell density liver-specific functions are expressed and growth is suppressed, whereas the opposite situation is observed at lower cell density. In contrast, neonatal hepatocytes in culture grow autonomously without a requirement for added hormones. This autonomous growth is due to an autocrine mechanism in which the cells secrete one or more growth factors into the culture medium. However, this autonomous growth ceases one week after birth at a time when the cells begin to express differentiated characteristics. Based upon these data, the mechanisms of liver regeneration, differentiation, and hepatocarcinogenesis are discussed.

    Topics: Animals; Blood Platelets; Cell Transformation, Neoplastic; Cells, Cultured; Extracellular Matrix; Growth Substances; Interleukin-1; Interleukin-6; Liver Regeneration; Rats; Transforming Growth Factor beta

1991
Polyomavirus middle tumor antigen increases responsiveness to growth factors.
    Journal of virology, 1991, Volume: 65, Issue:5

    The middle tumor antigen (mT) of polyomavirus is unable to transform a clone of NIH 3T3 cells to anchorage independence (L. Raptis and J.B. Bolen, J. Virol. 63:753-758, 1989). However, this oncogene increased the responsiveness of these cells to the growth factors (alpha-like and beta-type transforming growth factors) produced by cells possessing the whole transforming region of polyomavirus. This resulted in the growth of NIH 3T3 cells, expressing mT under control of the dexamethasone-regulatable mouse mammary tumor virus promoter, in agar medium supplemented with these growth factors upon addition of the inducer. Therefore, mT, a transforming oncogene, is able to enhance the responsiveness of established cells to growth factors, a property previously attributed primarily to myc and other establishment type oncogenes.

    Topics: Animals; Antigens, Polyomavirus Transforming; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Mice; Oncogenes; Phosphorylation; Promoter Regions, Genetic; Transforming Growth Factor alpha; Transforming Growth Factor beta

1991
Liver tumor promotion: effect of phenobarbital on EGF and protein kinase C signal transduction and transforming growth factor-beta 1 expression.
    Digestive diseases and sciences, 1991, Volume: 36, Issue:5

    Phenobarbital (PB) added to the medium of cultured rat hepatocytes alters epidermal growth factor (EGF) dependent mitogenesis in a biphasic manner; PB concentrations less than 1.5 mM are growth stimulatory but higher concentrations significantly inhibit normal hepatocyte proliferation. In contrast, the growth of putative preneoplastic cells is inhibited less by high concentrations of PB. Mechanistic studies designed to test the ability of PB to alter the early events of EGF signal transduction demonstrate that PB neither competes with EGF for binding to the EGF receptor nor alters EGF-induced receptor down-regulation. However, pretreatment with PB (greater than 1 mM) results in a transient inhibition of EGF binding to hepatocytes. The kinetics of this effect are similar to those obtained when hepatocytes are exposed to the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA), a skin tumor promoter and activator of Ca2+/phospholipid-dependent protein kinase C. However, several observations suggest that distinct mechanisms mediate the responses to these two tumor promoters. First, the inhibitory effects of PB and TPA on EGF binding are additive. Also down-regulation of EGF receptors in response to TPA occurs with hepatocytes, A431 epidermal carcinoma cells, HepG2 hepatoma cells, and rat liver epithelial cells, but only hepatocytes are sensitive to PB. Furthermore, translocation of protein kinase C to the membrane occurs in hepatocytes treated with TPA but not in those treated with PB. The chronic treatment of rats with PB further sensitizes hepatocytes to EGF receptor down-regulation by in vitro PB while desensitizing them to EGF receptor down-regulation by TPA. This latter effect is correlated with a decreased ability of TPA to induce translocation of protein kinase C to the membrane. PB significantly increases the intracellular concentration of TGF-beta 1 in periportal hepatocytes but not in putative preneoplastic cells. TGF-beta 1 may therefore have an important function in regulating early stages of cell cycle progression in proliferating hepatocytes.

    Topics: Animals; Binding, Competitive; Cell Transformation, Neoplastic; Diethylnitrosamine; Epidermal Growth Factor; Liver; Liver Neoplasms, Experimental; Phenobarbital; Protein Kinase C; Rats; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured

1991
Expression of growth-related genes during tumor progression in v-raf-transformed rat liver epithelial cells.
    Cancer research, 1990, Dec-01, Volume: 50, Issue:23

    Clonal cell lines were derived from rat liver epithelial cells following their transformation with either v-raf or v-raf/v-myc. Cells transformed with v-raf alone showed reduced tumor incidence and tumor growth rates when implanted into nude mice, compared to cells also expressing the v-myc oncogene. A series of additional clones isolated from a tumor obtained following inoculation of an athymic nude mouse with the v-raf-transformed rat liver epithelial cells displayed an intermediate range of tumor aggressiveness. These findings indicate that unknown genotypic and/or phenotypic changes occur during tumor formation in vivo, which are required in addition to raf activation for complete expression of the malignant phenotype. This in vitro model of tumor progression was used to examine alterations in the expression of genes related to the growth control of liver epithelial cells, which may be involved in the malignant conversion of the preneoplastic cells. A close association was observed between the increased level of expression of the transforming growth factors alpha and beta 1, the decreased expression of extracellular matrix proteins fibronectin and collagen I, and the tumor aggressiveness (latency/growth rate), suggesting a causal role for these factors in the progression of v-raf-transformed rat liver epithelial cells to the fully malignant phenotype.

    Topics: Animals; Blotting, Northern; Blotting, Southern; Blotting, Western; Cell Division; Cell Transformation, Neoplastic; DNA; Down-Regulation; Extracellular Matrix Proteins; Fibronectins; Gene Expression Regulation, Neoplastic; Growth Substances; Liver Neoplasms; Male; Mice; Mice, Nude; Oncogene Proteins v-raf; Rats; Retroviridae Proteins, Oncogenic; RNA; Transformation, Genetic; Transforming Growth Factor alpha; Transforming Growth Factor beta

1990
Transforming growth factor beta 1 in liver carcinogenesis: messenger RNA expression and growth effects.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1990, Volume: 1, Issue:3

    Transforming growth factor beta 1 (TGF-beta 1) is a potent inhibitor of hepatocyte proliferation. Since loss of sensitivity to growth inhibition is thought to contribute to the development of neoplasia, we analyzed the expression of TGF-beta 1 mRNA during hepatocarcinogenesis in vivo and in cultured liver epithelial cells (oval cells) obtained from carcinogen-treated animals. We found that TGF-beta 1 mRNA increases in the liver during carcinogenesis and that, at the early stages of the process, oval cells but not hepatocytes contain the growth factor mRNA. Moreover, immortalized, nontumorigenic oval cells (LE/6 cell line) continued to produce TGF-beta 1 mRNA in culture. TGF-beta 1 message markedly decreased upon cell transformation, but message levels, although generally low, were variable in various tumor cell clones. A consistent feature of the tumorigenic cell lines was a loss of sensitivity to TGF-beta 1 growth inhibition. Tumor cells could bind TGF-beta 1 with similar capacity as normal cells and had the same type of receptors (Mr 280,000, 85,000, and 65,000) capable of binding iodinated TGF-beta 1, suggesting that the loss of sensitivity to TGF-beta 1 in transformed liver epithelial cells involves postreceptor mechanisms. Further studies showed that c-myc is not a target for TGF-beta 1 in liver epithelial cells and that TGF-beta 1 no longer induces fibronectin mRNA in transformed cells. The data presented are consistent with the hypothesis that TGF-beta 1 secreted during liver carcinogenesis may inhibit the proliferation of normal cells while providing a selective advantage for the growth of cells that are "partially transformed" and are unresponsive to the factor.

    Topics: Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Choline Deficiency; Epithelium; Fibronectins; Gene Expression Regulation; Liver; Liver Neoplasms, Experimental; Male; Mice; Mice, Nude; Neoplasm Transplantation; Proto-Oncogene Proteins c-myc; Rats; Rats, Inbred Strains; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; RNA, Messenger; Transforming Growth Factor beta

1990
Sensitivity of the cell cycle to TGF beta 1 does not correlate with transformation of a rat liver epithelial cell line.
    Cell biology and toxicology, 1990, Volume: 6, Issue:4

    The effects of TGF beta 1 on cell cycle events in a rat liver derived epithelial cell line (BL9) and in two in vitro transformants of this line were studied by flow cytometry. Using either ethidium bromide staining or the incorporation of bromodeoxyuridine to evaluate DNA synthesis it was shown that TGF beta 1 prevented the entry of G0/G1 phase BL9 cells into S phase. TGF beta 1 did not exert its inhibitory effect(s) on DNA synthesis by the modulation of early events in the cell cycle. The tumorigenic transformed BL9 cell lines gave contrasting responses to the effects of TGF beta 1. DNA synthesis in a BL9 cell line derived by transfection with an active N-ras oncogene was unaffected by TFG beta 1 and thus appeared refractory to its growth controlling effects. On the other hand cells from a BL9 cell line derived by in vitro transformation with activated aflatoxin B1 retained their sensitivity to the effects of TGF beta 1. Thus the loss of the inhibitory effect of TGF beta 1 on DNA synthesis is not obligatory for the malignant transformation of rat liver epithelial cells.

    Topics: Animals; Cell Cycle; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; DNA; Liver; Rats; Transforming Growth Factor beta

1990
Neoplastic transformation of mouse mammary epithelial cells by deregulated myc expression.
    Cell regulation, 1990, Volume: 1, Issue:11

    A spontaneously immortalized, nontumorigenic mouse mammary epithelial cell line (MMEC) was transfected with an activated myc construct by electroporation. Constitutive expression of myc in MMEC resulted in anchorage independence in soft agar and tumorigenicity in nude mice. The myc-expressing MMEC showed higher saturation density, faster growth rate, and partial abrogation of serum-derived growth factor(s) requirement compared with parent MMEC. Epidermal growth factor or transforming growth factor alpha stimulated the anchorage-independent growth, but not the anchorage-dependent growth, of MMEC-myc cells. Type 1 transforming growth factor beta, on the other hand, inhibited both the anchorage-independent and anchorage-dependent growth of MMEC-myc cells. These results demonstrate that deregulated expression of myc results in neoplastic transformation iin mammary epithelial cells. Accompanying the transformation is altered sensitivity to polypeptide growth factors.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; Female; Gene Expression Regulation, Neoplastic; Genes, myc; Mammary Glands, Animal; Mice; Mice, Nude; Neoplasm Transplantation; Proto-Oncogene Proteins c-myc; Transfection; Transforming Growth Factor alpha; Transforming Growth Factor beta

1990
Transforming growth factor-beta activities in 'in vivo' lines of hormone-dependent and independent mammary adenocarcinomas induced by medroxyprogesterone acetate in BALB/c mice.
    Breast cancer research and treatment, 1990, Volume: 16, Issue:1

    We have determined the presence of transforming growth factor-beta (TGF-beta)-like polypeptides in mammary adenocarcinomas induced by medroxyprogesterone acetate (MPA) in BALB/c mice. In hormone-dependent tumors (HD) from nontreated and MPA-treated mice a high molecular weight (43 kDa) transforming activity was purified by Bio-Gel P-60 chromatography. This TGF was able to confer the neoplastic phenotype on NRK-49F cells without the addition of epidermal growth factor (EGF), though its activity was potentiated by EGF. It did not compete for binding to the EGF receptor, had no mitogenic activity on monolayer cultures of NRK fibroblasts, and was a potent inhibitor of DNA synthesis induced in these cells by EGF and insulin. In HD and hormone-independent tumors (HI) another TGF with a Mr of 13 kDa was isolated. This transforming activity showed the same biological properties as 43 kDa TGF, with the exception that in the absence of EGF it did not stimulate soft agar growth of NRK-49F cells. The synthesis of both factors in 'in vivo' HD tumors seems to be under MPA control, since it is much lower in HD tumors from MPA-treated mice. Further purification of the 13 and 43 kDa TGFs by hydrophobic interaction HPLC demonstrated that each one eluted in a different position, and that their elution profile differed from the TGF-beta from human platelets. The biological activity of the 13 and 43 kDa TGFs was not neutralized by a specific anti-TGF-beta antibody.

    Topics: Adenocarcinoma; Animals; Cell Division; Cell Transformation, Neoplastic; Chromatography, High Pressure Liquid; DNA Replication; Drug Interactions; Epidermal Growth Factor; Fibroblasts; Gene Expression Regulation, Neoplastic; Insulin; Mammary Neoplasms, Experimental; Medroxyprogesterone; Medroxyprogesterone Acetate; Mice; Mice, Inbred BALB C; Neoplasm Proteins; Neoplasms, Hormone-Dependent; Rats; Transforming Growth Factor beta

1990
High frequency of resistance of human squamous carcinoma cells to the anti-proliferative action of transforming growth factor beta.
    Cancer communications, 1990, Volume: 2, Issue:11

    Transforming Growth Factor beta is a potent autocrine inhibitor of the growth of untransformed keratinocytes. We found each of eight human squamous carcinoma cell lines to be refractory to the anti-proliferative action of Transforming Growth Factor beta. Although each of these carcinoma cell lines expressed the 53-65 kDa (type I) and the 280-300 (type III) Transforming Growth Factor beta-receptor proteins, the 73-85 kDa (type II) species was detectable in only one of these cell lines. Furthermore, although Transforming Growth Factor beta-sensitive non-neoplastic mouse keratinocytes expressed type II binding proteins, human keratinocytes did not. Our findings suggest that resistance to the growth-inhibitory actions of Transforming Growth Factor beta is a common feature of human squamous carcinoma cell lines but does not correlate with the expression of cell-surface receptors for this growth factor.

    Topics: Carcinoma, Squamous Cell; Cell Division; Cell Transformation, Neoplastic; DNA; Drug Resistance; Epidermal Growth Factor; Humans; Keratinocytes; Kinetics; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Tumor Cells, Cultured

1990
Altered responsiveness of rat liver epithelial cells to transforming growth factor beta 1 following their transformation with v-raf.
    Cancer research, 1990, Dec-01, Volume: 50, Issue:23

    The effects of transforming growth factor beta (type 1) (TGF-beta 1) on DNA synthesis, cell proliferation, and protein synthesis were examined in a series of v-raf-transformed rat liver epithelial (RLE) cells, which exhibit a range of transformed phenotypes. All of the transformed cells were relatively resistant to the growth-inhibitory effects of TGF-beta 1, compared to normal RLE cells and control cells infected with a helper virus. The more tumorigenic cell lines had very few surface receptors for TGF-beta 1 and showed no increase in the secretion of a number of specific proteins, including fibronectin, following TGF-beta 1 treatment. In contrast, the more normal-looking, less tumorigenic v-raf-transformed cells bound similar amounts of TGF-beta 1 as normal RLE and control cells and showed a similar pattern of TGF-beta 1-stimulated protein secretion. These findings suggest that the effects of TGF-beta 1 on cell proliferation and on the expression of certain secreted proteins are mediated through different mechanisms. Following transformation of RLE cells with v-raf, the signalling pathways controlling TGF-beta 1 growth inhibition are perturbed, while those involved in regulating the synthesis of certain proteins may remain intact. Thus, the escape from the various distinct biological effects of TGF-beta 1 may be an important stage in the progression of neoplastic transformation of RLE cells in vitro.

    Topics: Animals; Blotting, Northern; Cell Division; Cell Transformation, Neoplastic; DNA; Dose-Response Relationship, Drug; Electrophoresis, Gel, Two-Dimensional; Epithelium; Extracellular Matrix Proteins; In Vitro Techniques; Liver; Rats; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; RNA, Messenger; Transformation, Genetic; Transforming Growth Factor beta

1990
[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
Immunoregulatory role of transforming growth factor beta (TGF-beta) in development of killer cells: comparison of active and latent TGF-beta 1.
    The Journal of experimental medicine, 1990, Dec-01, Volume: 172, Issue:6

    Using recombinant DNA technology, we have generated Chinese hamster ovary (CHO) cell lines that synthesize latent transforming growth factor beta 1 (TGF-beta 1) to study immune regulation by TGF-beta 1. In vitro, latent TGF-beta 1 synthesized by transfectants or added exogenously as a purified complex after activation inhibited CTL generation to a similar extent as seen with acid-activated recombinant human (rHu) TGF-beta 1. In vivo, serum from nu/nu mice bearing CHO/TGF-beta 1 tumors contained significant levels of latent TGF-beta 1 in addition to depressed natural killer (NK) activity in spleens which paralleled that seen in C3H/HeJ mice treated with acid-activated rHuTGF-beta 1. rHuTGF-beta 1 treatment of mice receiving heart allografts resulted in significant enhancement of organ graft survival. Because of possible regulated tissue-specific activation, administration of latent rather than active TGF-beta may provide a better route to deliver this powerful immunosuppressive agent in vivo.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cricetinae; Cricetulus; Female; Graft Survival; Heart Transplantation; Humans; Killer Cells, Natural; Mice; Mice, Inbred C57BL; Mice, Nude; Ovary; Recombinant Proteins; T-Lymphocytes, Cytotoxic; Transfection; Transforming Growth Factor beta

1990
Loss of growth inhibitory activity of TGF-beta toward normal human mammary epithelial cells grown within collagen gel matrix.
    Biochemical and biophysical research communications, 1990, Dec-31, Volume: 173, Issue:3

    TGF-beta at concentrations in the range from 0.1 to 10 ng/ml gave significant growth inhibition of nonmalignant human mammary epithelial cells (HMEC) but not of malignant HMEC grown in monolayer cultures in serum-free medium. However, no growth inhibition of the nonmalignant cells was observed when the cells were cultivated within a type-I collagen gel matrix either adhering to a plastic substratum or floating on the medium. Within floating collagen gels, both nonmalignant and malignant HMEC formed a cell mass having radial extensions, and TGF-beta at 1 or 10 ng/ml prevented the formation of extensions only in the nonmalignant HMEC.

    Topics: Breast; Breast Neoplasms; Cell Adhesion; Cell Transformation, Neoplastic; Collagen; Epithelium; Gels; Humans; Plastics; Transforming Growth Factor beta; Tumor Cells, Cultured

1990