transforming-growth-factor-beta has been researched along with Melanoma* in 229 studies
18 review(s) available for transforming-growth-factor-beta and Melanoma
Article | Year |
---|---|
The histone demethylase PHF8 regulates TGFβ signaling and promotes melanoma metastasis.
The contribution of epigenetic dysregulation to metastasis remains understudied. Through a meta-analysis of gene expression datasets followed by a mini-screen, we identified Plant Homeodomain Finger protein 8 (PHF8), a histone demethylase of the Jumonji C protein family, as a previously unidentified prometastatic gene in melanoma. Loss- and gain-of-function approaches demonstrate that PHF8 promotes cell invasion without affecting proliferation in vitro and increases dissemination but not subcutaneous tumor growth in vivo, thus supporting its specific contribution to the acquisition of metastatic potential. PHF8 requires its histone demethylase activity to enhance melanoma cell invasion. Transcriptomic and epigenomic analyses revealed that PHF8 orchestrates a molecular program that directly controls the TGFβ signaling pathway and, as a consequence, melanoma invasion and metastasis. Our findings bring a mechanistic understanding of epigenetic regulation of metastatic fitness in cancer, which may pave the way for improved therapeutic interventions. Topics: Cell Proliferation; Epigenesis, Genetic; Histone Demethylases; Homeodomain Proteins; Humans; Melanoma; Transcription Factors; Transforming Growth Factor beta | 2022 |
The roles of TGF-β and VEGF pathways in the suppression of antitumor immunity in melanoma and other solid tumors.
Immune checkpoint blockade (ICB) has become well-known in cancer therapy, strengthening the body's antitumor immune response rather than directly targeting cancer cells. Therapies targeting immune inhibitory checkpoints, such as PD-1, PD-L1, and CTLA-4, have resulted in impressive clinical responses across different types of solid tumors. However, as with other types of cancer treatments, ICB-based immunotherapy is hampered by both innate and acquired drug resistance. We previously reported the enrichment of gene signatures associated with wound healing, epithelial-to-mesenchymal, and angiogenesis processes in the tumors of patients with innate resistance to PD-1 checkpoint antibody therapy; we termed these the Innate Anti-PD-1 Resistance Signatures (IPRES). The TGF-β and VEGFA pathways emerge as the dominant drivers of IPRES-associated processes. Here, we review these pathways' functions, their roles in immunosuppression, and the currently available therapies that target them. We also discuss recent developments in the targeting of TGF-β using a specific antibody class termed trap antibody. The application of trap antibodies opens the promise of localized targeting of the TGF-β and VEGFA pathways within the tumor microenvironment. Such specificity may offer an enhanced therapeutic window that enables suppression of the IPRES processes in the tumor microenvironment while sparing the normal homeostatic functions of TGF-β and VEGFA in healthy tissues. Topics: Antibodies; Humans; Immunotherapy; Melanoma; Neoplasms; Programmed Cell Death 1 Receptor; Transforming Growth Factor beta; Tumor Microenvironment; Vascular Endothelial Growth Factor A | 2022 |
Exploring major signaling cascades in melanomagenesis: a rationale route for targetted skin cancer therapy.
Although most melanoma cases may be treated by surgical intervention upon early diagnosis, a significant portion of patients can still be refractory, presenting low survival rates within 5 years after the discovery of the illness. As a hallmark, melanomas are highly prone to evolve into metastatic sites. Moreover, melanoma tumors are highly resistant to most available drug therapies and their incidence have increased over the years, therefore leading to public health concerns about the development of novel therapies. Therefore, researches are getting deeper in unveiling the mechanisms by which melanoma initiation can be triggered and sustained. In this context, important progress has been achieved regarding the roles and the impact of cellular signaling pathways in melanoma. This knowledge has provided tools for the development of therapies based on the intervention of signal(s) promoted by these cascades. In this review, we summarize the importance of major signaling pathways (mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)-Akt, Wnt, nuclear factor κ-light-chain-enhancer of activated B cell (NF-κB), Janus kinase (JAK)-signal transducer and activator of transcription (STAT), transforming growth factor β (TGF-β) and Notch) in skin homeostasis and melanoma progression. Available and developing melanoma therapies interfering with these signaling cascades are further discussed. Topics: Carcinogenesis; Humans; Melanoma; Mitogen-Activated Protein Kinases; Molecular Targeted Therapy; NF-kappa B; Signal Transduction; Skin; Skin Neoplasms; STAT Transcription Factors; Transforming Growth Factor beta; Wnt Signaling Pathway | 2018 |
Molecular Aspects of FKBP51 that Enable Melanoma Dissemination.
FKBP51 (FKBP5 Official Symbol) is large molecular weight member of the FK506 binding protein family, a subfamily of the immunophilin proteins. FKBP51 exerts multiple biological functions in the cell, including modulation of steroid hormone response, immune regulation, cell proliferation, regulation of pAkt levels and control of NF-κB activation. Several lines of evidence support a role for this protein in cancer biology, especially in resistance to chemo- and radio-therapy. Recent research studies highlighted functions of FKBP51 in promoting the epithelial to mesenchymal transition (EMT) transdifferentiation program in melanoma. This process, which is classically regulated by Transforming Growth Factor (TGF)-β, enables cancer cells to disseminate from primary tumors and spread to distant locations, acquiring resistance to therapy and self-renewal capability. This last, in turn, is crucial to their subsequent expansion at sites of dissemination. The aim of the present article is to review recent literature data that involve FKBP51 in the mechanisms that switch the TGF-β from a tumor suppressor to a pro-metastatic invader. Topics: Animals; Cell Transdifferentiation; Epithelial-Mesenchymal Transition; Humans; Melanoma; Neoplasm Metastasis; Tacrolimus Binding Proteins; Transforming Growth Factor beta | 2015 |
Role of TGF-β in melanoma.
Human malignant melanoma is highly resistant to chemotherapy and current immunotherapeutic approaches induce long term remission only in the minority of patients. The transforming growth factor-β (TGF-β) has attracted much attention as a therapeutic target because it plays an important and pleiotropic role in melanoma progression. TGF-β is a multifunctional cytokine involved in the regulation of many cellular processes including cell proliferation, differentiation and survival. Resistance to the growth inhibitory effects of TGF-β without alterations of TGF-β signaling molecules is characteristic of cutaneous melanoma. Melanoma produces increasing amounts of TGF-β with disease progression, inhibiting immune responses and providing an optimal microenvironment for undisturbed tumor growth. In addition, TGF-β exerts its tumor promoting functions via direct effects on tumor cell motility and invasiveness and indirectly by modulating tumor stroma and extracellular matrix, supporting angiogenesis and inhibiting immune surveillance. TGF-β acts through multiple intracellular signaling pathways and the outcome of TGF-β signaling is context-dependent. Defining the impact of the different TGF-β signaling pathways on melanoma progression will help to identify suitable therapeutic targets. Here we review the current knowledge of TGF-β in melanoma and discuss recent therapeutic approaches targeting the TGF-β pathway. Topics: Animals; Humans; Immunosuppression Therapy; Melanoma; Neoplasm Invasiveness; Neoplasm Metastasis; Neovascularization, Pathologic; Skin Neoplasms; Transforming Growth Factor beta | 2011 |
TGF-β/SMAD/GLI2 signaling axis in cancer progression and metastasis.
The Hedgehog (HH) and TGF-β signaling pathways represent essential regulators of cell proliferation and differentiation during embryogenesis. Pathway deregulation is a characteristic of various cancers. Recently, evidence for a convergence of these pathways at the level of the GLI2 transcription factor in the context of tumor initiation and progression to metastasis has emerged. This short review summarizes recent knowledge about GLI2 function and mechanisms of action downstream of TGF-β in cancer. Topics: Bone Neoplasms; Breast Neoplasms; Disease Progression; Female; Hedgehog Proteins; Humans; Kruppel-Like Transcription Factors; Melanoma; Neoplasm Metastasis; Neoplasms; Nuclear Proteins; Signal Transduction; Skin Neoplasms; Smad Proteins; Transforming Growth Factor beta; Zinc Finger Protein Gli2 | 2011 |
Resistance to transforming growth factor β-mediated tumor suppression in melanoma: are multiple mechanisms in place?
Resistance to transforming growth factor (TGF) β-mediated tumor suppression in melanoma appears to be a crucial step in tumor aggressiveness since it is usually coupled with the ability of TGFβ to drive the oncogenic process via autocrine and paracrine effects. In this review, we will focus mainly on the mechanisms of escape from TGFβ-induced cell cycle arrest because the mechanisms of resistance to TGFβ-mediated apoptosis are still essentially speculative. As expected, some of these mechanisms can directly affect the function of the main downstream effectors of TGFβ, Smad2 and Smad3, resulting in compromised Smad-mediated antiproliferative activity. Other mechanisms can counteract or overcome TGFβ-mediated cell cycle arrest independently of the Smads. In melanoma, some models of resistance to TGFβ have been suggested and will be described. In addition, we propose additional models of resistance taking into consideration the information available on the dysregulation of fundamental cellular effectors and signaling pathways in melanoma. Topics: Apoptosis; Cell Cycle; Contractile Proteins; Cyclin-Dependent Kinase 4; Cyclin-Dependent Kinase Inhibitor p21; Disease Progression; Filamins; Forkhead Box Protein O1; Forkhead Transcription Factors; Genes, myc; Humans; Intracellular Signaling Peptides and Proteins; Melanoma; Microfilament Proteins; Paired Box Transcription Factors; PAX3 Transcription Factor; Phosphorylation; Proto-Oncogene Proteins; Signal Transduction; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta | 2010 |
Metastasis: alone or together?
Topics: Animals; Carcinoma; Cell Differentiation; Cell Movement; Humans; Melanoma; Models, Biological; Neoplasm Metastasis; Neovascularization, Pathologic; Transforming Growth Factor beta | 2009 |
Immune modulation by melanoma-derived factors.
Melanomas, while the less common of skin cancers, are highly aggressive and once they metastasize usually indicate a poor prognosis. Melanomas are in many cases immunogenic and thus have been a prime target for immunotherapy, which has resulted in objective responses in some patients. To understand why antitumor immunity fails, and for the purpose of discovering new targets to improve therapy, there has been great interest to analyse the antitumor immune responses which exist in these patients, and uncover mechanisms which block tumor-specific immune responses. It is now evident that immunosuppressive cell networks and factors play a major role in the failure of the antitumor immune responses and therapies to eradicate the tumor. In this review, the factors produced by melanomas which can modulate and enhance these suppressive mechanisms are discussed. The roles of immature dendritic cells, neutrophils, T-regulatory cells, myeloid-derived suppressor cells and M2 macrophages or tumor-associated macrophages are described. Furthermore, taking into consideration of the cross-talk which exists among these different cell types and the cycle of immunosuppression which is evident in melanoma cancer patients and animal models, will be important for future therapeutic approaches. Topics: Animals; Chemokine CCL2; Granulocyte-Macrophage Colony-Stimulating Factor; Humans; Immune Tolerance; Immunologic Factors; Interleukins; Melanoma; Models, Immunological; Transforming Growth Factor beta | 2008 |
Transforming growth factor-beta in cutaneous melanoma.
Transforming growth factor-beta (TGF-beta) plays a complex role during carcinogenesis. It may either act as a tumor suppressor through its broad antiproliferative potential or as a tumor promoter either via direct effects on tumor cell aggressiveness or indirectly by modulating stromal responses, angiogenesis and immune surveillance. Increased production of TGF-beta by cancer cells is often associated with tumor grade. Melanoma cells largely escape cell cycle arrest normally induced by TGF-beta in normal melanocytes, yet produce active TGF-beta and are capable of efficient transcriptional responses to the growth factor. In this review, we summarize the current knowledge about the role played by TGF-beta in melanoma progression and hypothesize about the appropriateness of targeting TGF-beta signaling for therapeutic intervention. Topics: Antineoplastic Agents; Extracellular Signal-Regulated MAP Kinases; Humans; Melanoma; Signal Transduction; Skin Neoplasms; Smad Proteins; Transforming Growth Factor beta | 2008 |
Role of nodal signaling and the microenvironment underlying melanoma plasticity.
The incidence of melanoma has increased dramatically over the last 50 yr, and although melanoma accounts for only 10% of all skin cancers, it is responsible for over 80% of skin cancer deaths. Recent studies have uncovered critical molecular events underlying melanocytic transformation and melanomagenesis. Among these noteworthy observations are the acquisition of stem cell-associated proteins, such as the Notch receptors and Nodal, which have also been implicated in melanoma progression. For example, we have demonstrated that Nodal expression is limited to invasive vertical growth phase and metastatic melanoma lesions, and that inhibition of Nodal signaling promotes the reversion of metastatic melanoma cells toward a more differentiated, less invasive non-tumorigenic phenotype. In addition, molecular cross-talk exists between the Notch and Nodal signaling pathways. Interestingly, the acquisition of stem cell-associated plasticity is often acquired via epigenetic mechanisms, and is therefore receptive to reprogramming in response to embryonic microenvironments. Here, we review the concept of melanoma plasticity, with an emphasis on the emerging role of Nodal as a regulator of melanoma tumorigenesis and progression, and present findings related to epigenetic reprogramming. Topics: Epigenesis, Genetic; Humans; Melanins; Melanocytes; Melanoma; Neoplasm Invasiveness; Neoplasm Metastasis; Nodal Protein; Signal Transduction; Transforming Growth Factor beta | 2008 |
Targeting Nodal in malignant melanoma cells.
Metastatic melanoma continues to be a significantly deadly cancer with a cure rate of < 20% and a median survival of 6 - 9 months. The aggressiveness associated with metastatic melanoma is largely attributable to its inherent plasticity, a property that is mediated by the secretion of Nodal, a stem-cell associated protein belonging to the transforming growth factor-beta superfamily. This is supported by the observations that Nodal expression is limited to invasive vertical growth phase and metastatic melanoma lesions, and that inhibition of Nodal signaling promotes the reversion of metastatic melanoma cells toward a more differentiated, less invasive non-tumorigenic phenotype. Hence, due to its restricted expression pattern and function as a melanoma-tumor-promoter, Nodal (and its signaling partners) present unique targets for both immunologic and pharmacologic therapies. Topics: Antineoplastic Agents; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Nodal Protein; Transforming Growth Factor beta | 2007 |
Reprogramming metastatic tumour cells with embryonic microenvironments.
Aggressive tumour cells share many characteristics with embryonic progenitors, contributing to the conundrum of tumour cell plasticity. Recent studies using embryonic models of human stem cells, the zebrafish and the chick have shown the reversion of the metastatic phenotype of aggressive melanoma cells, and revealed the convergence of embryonic and tumorigenic signalling pathways, which may help to identify new targets for therapeutic intervention. This Review will summarize the embryonic models used to reverse the metastatic melanoma phenotype, and highlight the prominent signalling pathways that have emerged as noteworthy targets for future consideration. Topics: Animals; Cell Communication; Cell Differentiation; Cell Line, Tumor; Cell Movement; Chick Embryo; Embryo, Mammalian; Embryo, Nonmammalian; Embryonic Stem Cells; Humans; Melanoma; Models, Animal; Neoplasm Metastasis; Neoplasm Transplantation; Neural Crest; Nodal Protein; Signal Transduction; Transforming Growth Factor beta; Zebrafish | 2007 |
Transforming growth factor-beta and malignant melanoma: molecular mechanisms.
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 |
Targeting the stromal fibroblasts: a novel approach to melanoma therapy.
Classically, cancer is thought of as a genetic disease, where the step-wise acquisition of mutations initiates and drives progression. More recent thinking posits that, although cancers are initiated through genetic mutation, progression is often the result of dynamic interactions between the tumor cells and their surrounding environment. Topics: Cell Adhesion; Cell Communication; Disease Progression; Drug Resistance, Neoplasm; Fibroblasts; Humans; Keratinocytes; Melanoma; Neoplasm Invasiveness; Neovascularization, Pathologic; Phenotype; Skin Neoplasms; Stromal Cells; Transforming Growth Factor beta | 2005 |
Repression of TGF-beta signaling by the oncogenic protein SKI in human melanomas: consequences for proliferation, survival, and metastasis.
Transforming growth factor-beta (TGF-beta ) has dual and paradoxical functions as a tumor suppressor and promoter of tumor progression and metastasis. TGF-Ji-mediated growth inhibition is gradually lost during melanoma tumor progression, but there are no measurable defects at the receptor level. Furthermore, melanoma cells release high levels of TGF-beta to the microenvironment, which upon activation induces matrix deposition, angiogenesis, survival, and transition to more aggressive phenotypes. The SKI and SnoN protein family associate with and repress the activity of Smad2, Smad3, and Smad4, three members of the TGF-fl signaling pathway. SKI also facilitates cell-cycle progression by targeting the RB pathway by at least two ways: it directly associates with RB and represses its activity when expressed at high levels, and indirectly, it represses Smad-mediated induction of p21(Waf-1) This results in increased CDK2 activity, RB phosphorylation,and inactivation. Therefore, high levels of SKI result in lesions to the RB pathway in a manner similar to p16 (INK4a) loss. SKI mRNA and protein levels dramatically increase during human melanoma tumor progression. In addition,the SKI protein shifts from nuclear localization in intraepidermal melanoma cells to nuclear and cytoplasmic in invasive and metastatic melanomas. Here, I discuss the basis for repression of intracellular TGF-beta signaling by SKI, some additional activities of this protein, and propose that by disrupting multiple tumor suppressor pathways, SKI functions as a melanoma oncogene. Topics: Animals; Cell Division; Cell Survival; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; DNA-Binding Proteins; Down-Regulation; Extracellular Matrix; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Mice; Mice, Mutant Strains; Neoplasm Metastasis; Proto-Oncogene Proteins; Repressor Proteins; Research; Signal Transduction; Transforming Growth Factor beta | 2003 |
[The Langerhans cell: from in vitro production to use in cellular immunotherapy].
Dendritic cells constitute a family of antigen presenting cells defined by their morphology and their capacity to initiate primary immune response. Langerhans cells are paradigmatic dendritic cells, described in 1868 by a young medical student, Paul Langerhans in Berlin. Langerhans cells are present with epithelial cells in the epidermis, bronchi and mucosae. After antigenic challenge, Langerhans cells migrate into the T cell areas of proximal lymph nodes where they act as professional antigen-presenting cells. Langerhans cells originate in the bone marrow and CD34+ hematopoïetic progenitors are present in cord blood or circulating blood. They are actively involved in skin lesions of allergic contact dermatitis or atopic dermatitis, in cancer immunosurveillance and are infected by HIV in AIDS. Since 1992, Langerhans cells may be generated in vitro from CD34+ cord blood or circulating blood progenitors by culture with GM-CSF and TNF alpha, as well as from peripheral blood monocytes by culture with GM-CSF, IL4 and TGF beta 1. The possibility to obtain from the blood, the circulating progenitors of dendritic cells and the subsequent possibility to harvest a large number of these cells through in vitro culture using growth factors, have given rise to several very interesting therapeutic perspectives, especially in the field of anti-cancer immunotherapy. In dermatology advanced studies have concerned malignant melanomas. Anti-melanoma immunization trials were performed in patients, through dendritic cells charged with melanoma antigens. Side effects appear to be limited. Injections of antigenically charged dendritic cells were performed subcutaneously, intravenously or in the lymph nodes. Positive clinical responses were obtained with, in some cases, complete remission of the metastasis. These results open a particularly interesting perspective in the field of cancer treatment. Topics: Antigen Presentation; Antigens, Neoplasm; Blood Cells; Cell Differentiation; Cells, Cultured; Clinical Trials as Topic; Dendritic Cells; Fetal Blood; Granulocyte-Macrophage Colony-Stimulating Factor; Hematopoietic Stem Cells; Humans; Immunophenotyping; Interleukin-4; Langerhans Cells; Melanoma; Monocytes; Skin; Transforming Growth Factor beta; Treatment Outcome; Tumor Necrosis Factor-alpha | 2001 |
[Angiogenesis and tumor progression in melanoma].
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 |
7 trial(s) available for transforming-growth-factor-beta and Melanoma
Article | Year |
---|---|
Safety, Antitumor Activity, and Immune Activation of Pegylated Recombinant Human Interleukin-10 (AM0010) in Patients With Advanced Solid Tumors.
Purpose Interleukin-10 (IL-10) stimulates the expansion and cytotoxicity of tumor-infiltrating CD8+ T cells and inhibits inflammatory CD4+ T cells. Pegylation prolongs the serum concentration of IL-10 without changing the immunologic profile. This phase I study sought to determine the safety and antitumor activity of AM0010. Patients and Methods Patients with selected advanced solid tumors were treated with AM0010 in a dose-escalation study, which was followed by a renal cell cancer (RCC) dose-expansion cohort. AM0010 was self-administered subcutaneously at doses of 1 to 40 μg/kg once per day. Primary end points were safety and tolerability; clinical activity and immune activation were secondary end points. Results In the dose-escalation and -expansion cohorts, 33 and 18 patients, respectively, were treated with daily subcutaneous injection of AM0010. AM0010 was tolerated in a heavily pretreated patient population. Treatment-related adverse events (AEs) included anemia, fatigue, thrombocytopenia, fever, and injection site reactions. Grade 3 to 4 nonhematopoietic treatment-related AEs, including rash (n = 2) and transaminitis (n = 1), were observed in five of 33 patients. Grade 3 to 4 anemia or thrombocytopenia was observed in five patients. Most treatment-related AEs were transient or reversible. AM0010 led to systemic immune activation with elevated immune-stimulatory cytokines and reduced transforming growth factor beta in the serum. Partial responses were observed in one patient with uveal melanoma and four of 15 evaluable patients with RCC treated at 20 μg/kg (overall response rate, 27%). Prolonged stable disease of at least 4 months was observed in four patients, including one with colorectal cancer with disease stabilization for 20 months. Conclusion AM0010 has an acceptable toxicity profile with early evidence of antitumor activity, particularly in RCC. These data support the further evaluation of AM0010 both alone and in combination with other immune therapies and chemotherapies. Topics: Adult; Aged; Aged, 80 and over; Anemia; Carcinoma, Renal Cell; Cytokines; Drug Eruptions; Exanthema; Fatigue; Female; Fever; Humans; Injections, Subcutaneous; Interferon-gamma; Interleukin-10; Interleukin-4; Interleukin-8; Kidney Neoplasms; Male; Melanoma; Middle Aged; Neoplasms; Polyethylene Glycols; Recombinant Proteins; Thrombocytopenia; Transforming Growth Factor beta; Uveal Neoplasms; Young Adult | 2016 |
Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma.
In advanced cancers, transforming growth factor-beta (TGFβ) promotes tumor growth and metastases and suppresses host antitumor immunity. GC1008 is a human anti-TGFβ monoclonal antibody that neutralizes all isoforms of TGFβ. Here, the safety and activity of GC1008 was evaluated in patients with advanced malignant melanoma and renal cell carcinoma.. In this multi-center phase I trial, cohorts of patients with previously treated malignant melanoma or renal cell carcinoma received intravenous GC1008 at 0.1, 0.3, 1, 3, 10, or 15 mg/kg on days 0, 28, 42, and 56. Patients achieving at least stable disease were eligible to receive Extended Treatment consisting of 4 doses of GC1008 every 2 weeks for up to 2 additional courses. Pharmacokinetic and exploratory biomarker assessments were performed.. Twenty-nine patients, 28 with malignant melanoma and 1 with renal cell carcinoma, were enrolled and treated, 22 in the dose-escalation part and 7 in a safety cohort expansion. No dose-limiting toxicity was observed, and the maximum dose, 15 mg/kg, was determined to be safe. The development of reversible cutaneous keratoacanthomas/squamous-cell carcinomas (4 patients) and hyperkeratosis was the major adverse event observed. One malignant melanoma patient achieved a partial response, and six had stable disease with a median progression-free survival of 24 weeks for these 7 patients (range, 16.4-44.4 weeks).. GC1008 had no dose-limiting toxicity up to 15 mg/kg. In patients with advanced malignant melanoma and renal cell carcinoma, multiple doses of GC1008 demonstrated acceptable safety and preliminary evidence of antitumor activity, warranting further studies of single agent and combination treatments.. Clinicaltrials.gov NCT00356460. Topics: Adult; Aged; Aged, 80 and over; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Carcinoma, Renal Cell; Female; Humans; Kidney Neoplasms; Male; Melanoma; Middle Aged; Neoplasm Staging; Positron-Emission Tomography; Tomography, X-Ray Computed; Transforming Growth Factor beta; Treatment Outcome | 2014 |
Regulatory T cell frequency in patients with melanoma with different disease stage and course, and modulating effects of high-dose interferon-alpha 2b treatment.
High-dose interferon-alpha 2b (IFN-alpha 2b) is the only approved systemic therapy in the United States for the adjuvant treatment of melanoma. The study objective was to explore the immunomodulatory mechanism of action for IFN-alpha 2b by measuring serum regulatory T cell (Treg), serum transforming growth factor-beta (TGF-beta), interleukin (IL)-10, and autoantibody levels in patients with melanoma treated with the induction phase of the high-dose IFN-alpha 2b regimen.. Patients with melanoma received IFN-alpha 2b administered intravenously (20 MU/m2 each day from day 1 to day 5 for 4 consecutive weeks). Serum Treg levels were measured as whole lymphocytes in CD4+ cells using flow cytometry while TGF-beta, IL-10, and autoantibody levels were measured using enzyme-linked immunosorbent assays.. Twenty-two patients with melanoma received IFN-alpha 2b treatment and were evaluated for Treg levels. Before treatment, Treg levels were significantly higher in patients with melanoma when compared with data from 20 healthy subjects (P = 0.001; Mann-Whitney test). Although a trend for reduction of Treg levels following IFN-α 2b treatment was observed (average decrease 0.29% per week), statistical significance was not achieved. Subgroup analyses indicated higher baseline Treg levels for stage III versus IV disease (P = 0.082), early recurrence versus no recurrence (P = 0.017), deceased versus surviving patients (P = 0.021), and preoperative neoadjuvant versus postoperative adjuvant treatment groups (not significant). No significant effects were observed on the levels of TGF-beta, IL-10, and autoantibodies in patients with melanoma treated with IFN-alpha 2b.. Patients with melanoma in this study showed increased basal levels of Treg that may be relevant to their disease and its progression. Treg levels shifted in patients with melanoma treated with IFN-alpha 2b, although no firm conclusions regarding the role of Tregs as a marker of treatment response or outcome can be made at present. Topics: Adult; Antineoplastic Agents; Autoantibodies; Case-Control Studies; CD4 Antigens; Dose-Response Relationship, Drug; Female; Flow Cytometry; Forkhead Transcription Factors; Humans; Interferon alpha-2; Interferon-alpha; Interleukin-10; Interleukin-2 Receptor alpha Subunit; Male; Melanoma; Neoplasm Staging; Recombinant Proteins; Skin Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Treatment Outcome | 2010 |
Prolonged survival of dendritic cell-vaccinated melanoma patients correlates with tumor-specific delayed type IV hypersensitivity response and reduction of tumor growth factor beta-expressing T cells.
The aim of this work was to assess immunologic response, disease progression, and post-treatment survival of melanoma patients vaccinated with autologous dendritic cells (DCs) pulsed with a novel allogeneic cell lysate (TRIMEL) derived from three melanoma cell lines.. Forty-three stage IV and seven stage III patients were vaccinated four times with TRIMEL/DC vaccine. Specific delayed type IV hypersensitivity (DTH) reaction, ex vivo cytokine production, and regulatory T-cell populations were determined. Overall survival and disease progression rates were analyzed using Kaplan-Meier curves and compared with historical records.. The overall survival for stage IV patients was 15 months. More than 60% of patients showed DTH-positive reaction against the TRIMEL. Stage IV/DTH-positive patients displayed a median survival of 33 months compared with 11 months observed for DTH-negative patients (P = .0014). All stage III treated patients were DTH positive and remained alive and tumor free for a median follow-up period of 48 months (range, 33 to 64 months). DTH-positive patients showed a marked reduction in the proportion of CD4+ transforming growth factor (TGF) beta+ regulatory T cells compared to DTH-negative patients (1.54% v 5.78%; P < .0001).. Our findings strongly suggest that TRIMEL-pulsed DCs provide a standardized and widely applicable source of melanoma antigens, very effective in evoking antimelanoma immune response. To our knowledge, this is the first report describing a correlation between vaccine-induced reduction of CD4+TGFbeta+ regulatory T cells and in vivo antimelanoma immune response associated to improved patient survival and disease stability. Topics: Adult; Cancer Vaccines; Dendritic Cells; Disease Progression; Female; Follow-Up Studies; Humans; Hypersensitivity, Delayed; Male; Melanoma; Middle Aged; Skin Neoplasms; Survival Analysis; T-Lymphocytes, Regulatory; Transforming Growth Factor beta | 2009 |
Interleukin 12 gene therapy of cancer by peritumoral injection of transduced autologous fibroblasts: outcome of a phase I study.
A phase I dose-escalation clinical trial of peritumoral injections of interleukin 12 (IL-12)-transduced autologous fibroblasts was performed in patients with disseminated cancer for whom effective treatment does not exist. The goals of this study were to assess the safety and toxicities as well as the efficacy, and ancillarily the immunomodulatory effects, of peritumoral IL-12 gene transfer. Primary dermal fibroblasts cultured from the patients were transduced with retroviral vector carrying human IL-12 genes (p35 and p40) as well as the neomycin phosphotransferase gene (TFG-hIL-12-Neo). Patients received four injections at intervals of 7 days. Nine patients were enrolled in this dose-escalation study, with secreted IL-12 doses ranging from 300 ng/24 hr for the first three patients to 1000, 3000, and 5000 ng/24 hr for two patients in each subsequent dosage level. Although a definite statement cannot be made, there appears to be perturbation of systemic immunity. Also, the locoregional effects mediated by tumor necrosis factor alpha (TNF-alpha) and CD8+ T cells were observed with tumor regression. Treatment-related adverse events were limited to mild to moderate pain at the injection site; clinically significant toxicities were not encountered. Transient but clear reductions of tumor sizes were observed at the injected sites in four of nine cases, and at noninjected distant sites in one melanoma patient. Hemorrhagic necrosis of tumors was observed in two melanoma patients. These data indicate that gene therapy by peritumoral injection of IL-12-producing autologous fibroblasts is feasible, and promising in patients with advanced cancer. Topics: Adult; CD8-Positive T-Lymphocytes; Female; Fibroblasts; Gene Transfer Techniques; Genetic Therapy; Humans; Immunoenzyme Techniques; Interleukin-12; Killer Cells, Natural; Male; Melanoma; Middle Aged; Retroviridae; Skin Neoplasms; T-Lymphocytes; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha | 2001 |
Vaccination with IL-12 gene-modified autologous melanoma cells: preclinical results and a first clinical phase I study.
Cytokine gene transfer into tumor cells has been shown to mediate tumor regression and antimetastatic effects in several animal models via immunomodulation. Therefore, clinical protocols have been developed to treat cancer patients with cytokine gene-modified tumor cells. We inserted the genes coding for the p35 and p40 chain of interleukin-12 (IL-12) in two independent eukaryotic expression vectors and transduced melanoma cells of 15 different primary tumor cultures with both plasmids by a ballistic gene transfer approach. Secreted IL-12 demonstrated strong bioactivity by inducing interferon-gamma release from peripheral blood lymphocytes upon coculture with cell culture supernatants after IL-12 gene transfer which could at least partly be blocked by IL-12-specific antisera. Further enrichment of transduced tumor cells by magnetic separation directly after gene transfer increased cytokine secretion from a mean of 119 pg in the unsorted to 507 pg IL-12 (24 h/10(8) cells) in the magnetically enriched cell fraction. Irradiation of these cells led to a further elevation of secreted IL-12 (mean 987 pg). Elevated IL-12 levels were detected over 7 days after irradiation in vitro. In a subsequent first clinical phase I study six patients with metastatic melanoma were vaccinated with autologous, interleukin-12 gene-modified tumor cells. Melanoma cells were expanded in vitro from surgically removed metastases, transduced by ballistic gene transfer, irradiated and were then injected subcutaneously (s.c.) at weekly intervals. Clinically, there was no major toxicity except for mild fever. All patients completed more than four s.c. vaccinations over 6 weeks and were eligible for immunological evaluation. Post-vaccination, peripheral mononuclear cells were found to contain an increased number of tumor-reactive proliferative as well as cytolytic cells as determined by a limiting dilution analysis in two patients. Two patients developed DTH reactivity against autologous melanoma cells and one had a minor clinical response. Biopsies taken from that patient's metastases revealed a heavy infiltration of CD4+ and CD8+ T lymphocytes. In conclusion, vaccination induced immunological changes even in a group of advanced, terminally ill patients. These changes can be interpreted as an increased antitumor immune response. Topics: Adult; Biolistics; Cancer Vaccines; Female; Gene Transfer Techniques; Genetic Therapy; Humans; Hypersensitivity, Delayed; Immunohistochemistry; Interleukin-10; Interleukin-12; Killer Cells, Natural; Male; Melanoma; Middle Aged; Polymerase Chain Reaction; T-Lymphocytes; Transforming Growth Factor beta; Tumor Cells, Cultured | 1998 |
In vivo regulation of transforming growth factor beta 1 transcription by immunotherapy: interleukin-2 impairs interferon-alpha-stimulated increase in steady-state mRNA levels of transforming growth factor beta 1.
Recombinant interleukin-2 (rIL-2) in combination with recombinant interferon alpha (rIFN alpha) has been shown to mediate significant antitumoral effects in some patients with advanced renal cell cancer or malignant melanoma. The therapeutic effects may be partially modulated by secondarily induced cytokines, especially with regard to in vivo lymphocyte activation. To investigate possible negative effects on lymphocyte activation during immunotherapy, we designed a study on transcription of transforming growth factor beta 1 (TGF beta 1), a known inhibitor of lymphocyte function, in patients undergoing treatment with daily alternating administration of rIFN alpha and rIL-2. Here we present data on gene expression of TGF beta 1. Kinetic mRNA studies revealed an increase of TGF beta 1 mRNA in peripheral mononuclear cells 12 h after subcutaneous injection of rIFN alpha. The following intravenous rIL-2 administration significantly decreased the amounts of TGF beta 1-specific mRNA. In contrast to the effect of the first dose, subsequent application of rIFN alpha did not enhance TGF beta gene expression during rIFN alpha/IL-2 therapy. The diminished TGF beta 1 gene expression returned to pretreatment levels 1-7 days after the last rIL-2 administration. When concomitant with a decrease in TGF beta 1 transcripts. Our results indicate a complex regulatory effect on secondarily induced cytokines such as TGF beta 1 by immunotherapeutic approaches. The rIL-2-mediated down-regulation of increased TGF beta 1 steady-state mRNA levels following rIFN alpha may represent a positive immune regulatory effect on cytotoxic cells. Furthermore this effect may modulate proliferation of neoplastic tissues. Topics: Autoradiography; Blotting, Northern; Carcinoma, Renal Cell; Cells, Cultured; Densitometry; Humans; Immunotherapy; Injections, Intravenous; Injections, Subcutaneous; Interferon Type I; Interleukin-2; Kidney Neoplasms; Leukocytes, Mononuclear; Melanoma; Recombinant Proteins; RNA, Messenger; Transcription, Genetic; Transforming Growth Factor beta | 1994 |
204 other study(ies) available for transforming-growth-factor-beta and Melanoma
Article | Year |
---|---|
Obstructive sleep apnoea is related to melanoma aggressiveness through paraspeckle protein-1 upregulation.
In patients with obstructive sleep apnoea (OSA), intermittent hypoxia induces overexpression of paraspeckle component (PSPC)1, a master modulator of transforming growth factor (TGF)-β signalling, which promotes cell cancer progression through epithelial-mesenchymal transition (EMT) and acquisition of cancer stem cell (CSC)-like features. However, the persistence of intermittent hypoxia-induced effects on PSPC1, and their consequences in cancer patients are not known. To this effect, circulating PSPC1 levels were compared in patients with cutaneous melanoma with or without OSA, and their relationship with tumour aggressiveness along with the. In 292 cutaneous melanoma patients, sleep studies and serum levels of PSPC1 and TGF-β were evaluated. The effect of PSPC1 on expression of EMT and CSC transcription factors was assessed using melanoma cell lines with patient sera under both normoxia and intermittent hypoxia conditions.. PSPC1 levels were higher in patients with moderate-severe OSA compared with mild OSA or non-OSA patients. Serum levels of PSPC1 were associated with several cutaneous melanoma clinical aggressiveness indicators. Both intermittent hypoxia exposures and serum from OSA patients upregulated TGF-β expression and amplified the expression of transcription factors associated with EMT activation and acquisition of CSC characteristics.. In cutaneous melanoma patients, OSA severity is associated with higher PSPC1 serum levels, which jointly with intermittent hypoxia would enhance the self-reprogramming capabilities of EMT and CSC feature acquisition of melanoma cells, promoting their intrinsic aggressiveness. Topics: Humans; Hypoxia; Melanoma; Melanoma, Cutaneous Malignant; Paraspeckles; RNA-Binding Proteins; Skin Neoplasms; Sleep Apnea, Obstructive; Transforming Growth Factor beta; Up-Regulation | 2023 |
Correlation Between Endoglin and Malignant Phenotype in Human Melanoma Cells: Analysis of hsa-mir-214 and hsa-mir-370 in Cells and Their Extracellular Vesicles.
Endoglin (CD105) is an auxiliary receptor of transforming growth factor (TGF)-β family members that is expressed in human melanomas. It is heterogeneously expressed by primary and metastatic melanoma cells, and endoglin targeting as a therapeutic strategy for melanoma tumors is currently been explored. However, its involvement in tumor development and malignancy is not fully understood. Here, we find that endoglin expression correlates with malignancy of primary melanomas and cultured melanoma cell lines. Next, we have analyzed the effect of ectopic endoglin expression on two miRNAs (hsa-mir-214 and hsa-mir-370), both involved in melanoma tumor progression and endoglin regulation. We show that compared with control cells, overexpression of endoglin in the WM-164 melanoma cell line induces; (i) a significant increase of hsa-mir-214 levels in small extracellular vesicles (EVs) as well as an increased trend in cells; and (ii) significantly lower levels of hsa-mir-370 in the EVs fractions, whereas no significant differences were found in cells. As hsa-mir-214 and hsa-mir-370 are not just involved in melanoma tumor progression, but they can also target endoglin-expressing endothelial cells in the tumor vasculature, these results suggest a complex and differential regulatory mechanism involving the intracellular and extracellular signaling of hsa-mir-214 and hsa-mir-370 in melanoma development and progression. Topics: Endoglin; Endothelial Cells; Extracellular Vesicles; Humans; Melanoma; MicroRNAs; Transforming Growth Factor beta | 2023 |
TGF-β Type I Receptor Signaling in Melanoma Liver Metastases Increases Metastatic Outgrowth.
Despite advances in treatment for metastatic melanoma patients, patients with liver metastasis have an unfavorable prognosis. A better understanding of the development of liver metastasis is needed. The multifunctional cytokine Transforming Growth Factor β (TGF-β) plays various roles in melanoma tumors and metastasis, affecting both tumor cells and cells from the surrounding tumor microenvironment. To study the role of TGF-β in melanoma liver metastasis, we created a model to activate or repress the TGF-β receptor pathway in vitro and in vivo in an inducible manner. For this, we engineered B16F10 melanoma cells to have inducible ectopic expression of a constitutively active (ca) or kinase-inactive (ki) TGF-β receptor I, also termed activin receptor-like kinase (ALK5). In vitro, stimulation with TGF-β signaling and ectopic caALK5 expression reduced B16F10 cell proliferation and migration. Contrasting results were found in vivo; sustained caALK5 expression in B16F10 cells in vivo increased the metastatic outgrowth in liver. Blocking microenvironmental TGF-β did not affect metastatic liver outgrowth of both control and caALK5 expressing B16F10 cells. Upon characterizing the tumor microenvironment of control and caALk5 expressing B16F10 tumors, we observed reduced (cytotoxic) T cell presence and infiltration, as well as an increase in bone marrow-derived macrophages in caALK5 expressing B16F10 tumors. This suggests that caALK5 expression in B16F10 cells induces changes in the tumor microenvironment. A comparison of newly synthesized secreted proteins upon caALK5 expression by B16F10 cells revealed increased secretion of matrix remodeling proteins. Our results show that TGF-β receptor activation in B16F10 melanoma cells can increase metastatic outgrowth in liver in vivo, possibly through remodeling of the tumor microenvironment leading to altered infiltration of immune cells. These results provide insights in the role of TGF-β signaling in B16F10 liver metastasis and could have implications regarding the use of TGF-β inhibitors for the treatment of melanoma patients with liver metastasis. Topics: Cell Line, Tumor; Cytokines; Humans; Liver Neoplasms; Melanoma; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Tumor Microenvironment | 2023 |
EDIL3 as an Angiogenic Target of Immune Exclusion Following Checkpoint Blockade.
Immune checkpoint blockade (ICB) has become the standard of care for several solid tumors. Multiple combinatorial approaches have been studied to improve therapeutic efficacy. The combination of antiangiogenic agents and ICB has demonstrated efficacy in several cancers. To improve the mechanistic understanding of synergies with these treatment modalities, we performed screens of sera from long-term responding patients treated with ipilimumab and bevacizumab. We discovered a high-titer antibody response against EGF-like repeats and discoidin I-like domains protein 3 (EDIL3) that correlated with favorable clinical outcomes. EDIL3 is an extracellular protein, previously identified as a marker of poor prognosis in various malignancies. Our Tumor Immune Dysfunction and Exclusion analysis predicted that EDIL3 was associated with immune exclusion signatures for cytotoxic immune cell infiltration and nonresponse to ICB. Cancer-associated fibroblasts (CAF) were predicted as the source of EDIL3 in immune exclusion-related cells. Furthermore, The Cancer Genome Atlas Skin Cutaneous Melanoma (TCGA-SKCM) and CheckMate 064 data analyses correlated high levels of EDIL3 with increased pan-fibroblast TGFβ response, enrichment of angiogenic signatures, and induction of epithelial-to-mesenchymal transition. Our in vitro studies validated EDIL3 overexpression and TGFβ regulation in patient-derived CAFs. In pretreatment serum samples from patients, circulating levels of EDIL3 were associated with circulating levels of VEGF, and like VEGF, EDIL3 increased the angiogenic abilities of patient-derived tumor endothelial cells (TEC). Mechanistically, three-dimensional microfluidic cultures and two-dimensional transmigration assays with TEC endorsed EDIL3-mediated disruption of the lymphocyte function-associated antigen-1 (LFA-1)-ICAM-1 interaction as a possible means of T-cell exclusion. We propose EDIL3 as a potential target for improving the transendothelial migration of immune cells and efficacy of ICB therapy. Topics: Calcium-Binding Proteins; Cell Adhesion Molecules; Endothelial Cells; Humans; Melanoma; Melanoma, Cutaneous Malignant; Skin Neoplasms; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A | 2023 |
The effects of ALK5 inhibition and simultaneous inhibition or activation of HIF-1α in melanoma tumor growth and angiogenesis.
Hypoxia is the most common signature of the tumor microenvironment that drives tumorigenesis through the complex crosstalk of a family of transcription factors called hypoxia-inducible factors (HIFs), with other intercellular signaling networks. Hypoxia increases transforming growth factor-beta (TGF-β) expression. TGF-β and HIF-1α play critical roles in several malignancies and their interactions in melanoma progression remain unknown. Therefore, the aim of this study was to assess the impact of inhibiting activin receptor-like kinase-5 (ALK5), a TGF-β receptor, on the response to HIF-1α activation or inhibition in melanoma tumor progression.. Tumors were induced in C57BL/6J mice by subcutaneous inoculation with B16F10 melanoma cells. Mice were divided into HIF-1α inhibitor, ALK5 inhibitor (1 mg/kg) and HIF-1α inhibitor (100 mg/kg), ALK5 inhibitor, HIF-1α activator (1000 mg/kg), HIF-1α activator and ALK5 inhibitor, and control groups to receive inhibitors and activators through intraperitoneal injection. The expression of E-cadherin was evaluated by RT-qPCR. Vessel density and platelet-derived growth factor receptor beta (PDGFR)-β+ cells around the vessels were investigated using immunohistochemistry.. The groups receiving HIF-1α inhibitor and activator showed lower and higher tumor growth compared to the control group, respectively. E-cadherin expression decreased in all groups compared to the control group, illustrating the dual function of E-cadherin in the tumor microenvironment. Vascular density was reduced in the groups given HIF-1α inhibitor, ALK5 inhibitor, and ALK5 and HIF-1α inhibitor simultaneously. The percentage of PDGFR-β+ cells was reduced in the presence of HIF-1α inhibitor, ALK5 inhibitor, HIF-1α and ALK5 inhibitors, and upon simultaneous treatment with HIF-1α activator and ALK5 inhibitor.. Despite increased expression and interaction between TGF-β and HIF-1α pathways in some cancers, in melanoma, inhibition of either pathway alone may have a stronger effect on tumor inhibition than simultaneous inhibition of both pathways. The synergistic effects may be context-dependent and should be further evaluated in different cancer types. Topics: Animals; Cadherins; Cell Line, Tumor; Humans; Hypoxia; Hypoxia-Inducible Factor 1, alpha Subunit; Melanoma; Mice; Mice, Inbred C57BL; Transforming Growth Factor beta; Tumor Microenvironment | 2023 |
Melanoma secretion of transforming growth factor-β2 leads to loss of epidermal AMBRA1 threatening epidermal integrity and facilitating tumour ulceration.
For patients with early American Joint Committee on Cancer (AJCC)-stage melanoma the combined loss of the autophagy regulatory protein AMBRA1 and the terminal differentiation marker loricrin in the peritumoral epidermis is associated with a significantly increased risk of metastasis.. The aim of the present study was to evaluate the potential contribution of melanoma paracrine transforming growth factor (TGF)-β signalling to the loss of AMBRA1 in the epidermis overlying the primary tumour and disruption of epidermal integrity.. Immunohistochemistry was used to analyse AMBRA1 and TGF-β2 in a cohort of 109 AJCC all-stage melanomas, and TGF-β2 and claudin-1 in a cohort of 30 or 42 AJCC stage I melanomas, respectively, with known AMBRA1 and loricrin (AMLo) expression. Evidence of pre-ulceration was analysed in a cohort of 42 melanomas, with TGF-β2 signalling evaluated in primary keratinocytes.. Increased tumoral TGF-β2 was significantly associated with loss of peritumoral AMBRA1 (P < 0·05), ulceration (P < 0·001), AMLo high-risk status (P < 0·05) and metastasis (P < 0·01). TGF-β2 treatment of keratinocytes resulted in downregulation of AMBRA1, loricrin and claudin-1, while knockdown of AMBRA1 was associated with decreased expression of claudin-1 and increased proliferation of keratinocytes (P < 0·05). Importantly, we show loss of AMBRA1 in the peritumoral epidermis was associated with decreased claudin-1 expression (P < 0·05), parakeratosis (P < 0·01) and cleft formation in the dermoepidermal junction (P < 0·05).. Collectively, these data suggest a paracrine mechanism whereby TGF-β2 causes loss of AMBRA1 overlying high-risk AJCC early-stage melanomas and reduced epidermal integrity, thereby facilitating erosion of the epidermis and tumour ulceration. Topics: Adaptor Proteins, Signal Transducing; Epidermis; Humans; Melanoma; Skin Neoplasms; Transforming Growth Factor beta; Transforming Growth Factor beta2; Transforming Growth Factors | 2022 |
Hierarchy of TGFβ/SMAD, Hippo/YAP/TAZ, and Wnt/β-catenin signaling in melanoma phenotype switching.
In melanoma, a switch from a proliferative melanocytic to an invasive mesenchymal phenotype is based on dramatic transcriptional reprogramming which involves complex interactions between a variety of signaling pathways and their downstream transcriptional regulators. TGFβ/SMAD, Hippo/YAP/TAZ, and Wnt/β-catenin signaling pathways are major inducers of transcriptional reprogramming and converge at several levels. Here, we report that TGFβ/SMAD, YAP/TAZ, and β-catenin are all required for a proliferative-to-invasive phenotype switch. Loss and gain of function experimentation, global gene expression analysis, and computational nested effects models revealed the hierarchy between these signaling pathways and identified shared target genes. SMAD-mediated transcription at the top of the hierarchy leads to the activation of YAP/TAZ and of β-catenin, with YAP/TAZ governing an essential subprogram of TGFβ-induced phenotype switching. Wnt/β-catenin signaling is situated further downstream and exerts a dual role: it promotes the proliferative, differentiated melanoma cell phenotype and it is essential but not sufficient for SMAD or YAP/TAZ-induced phenotype switching. The results identify epistatic interactions among the signaling pathways underlying melanoma phenotype switching and highlight the priorities in targets for melanoma therapy. Topics: Biomarkers; Biomarkers, Tumor; Cell Cycle Proteins; Cell Proliferation; Computational Biology; Disease Susceptibility; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Hippo Signaling Pathway; Humans; Melanoma; Models, Biological; Neoplasm Grading; Neoplasm Invasiveness; Neoplasm Staging; Phenotype; RNA Interference; RNA, Small Interfering; Signal Transduction; Smad Proteins; Transcription Factors; Transcriptional Coactivator with PDZ-Binding Motif Proteins; Transforming Growth Factor beta; Wnt Signaling Pathway | 2022 |
Higher rates of regional disease but improved outcomes in pediatric versus adult melanoma.
Malignant melanoma is rare in the pediatric population and management is largely extrapolated from adult guidelines. Adult data have shown that immediate completion lymph node dissection (CLND) does not improve overall survival in selected patients with clinically node negative, sentinel lymph node-positive disease. Current nodal management in children is unknown.. The National Cancer Database (NCDB) was queried for patients with melanoma from 2012-2017 and patients categorized as pediatric (≤18 years, n=962) or adult (n=327,987). Factors associated with CLND in children with positive SLNB were evaluated in multivariable analysis. Kaplan-Meier survival analysis was performed.. Compared to adults, children present with thicker primary tumors (T3 or T4 26.5% vs 15.5%, p<0.001), resulting in higher rates of nodal assessment with SLN biopsy or LND (60.2% vs 36.6%, p<0.001) and higher rates of regional nodal disease (35.1% vs 23.4%, p<0.001). Children underwent higher rates of CLND after SLN biopsy (10.4% vs 4.1%) and upfront lymph node dissection (15.2% vs 8.7%). A decreased rate of CLND was noted in 2017 compared to 2012 (odds ratio (OR) 0.16 (p=0.005). CLND was performed more often on multivariable analysis for older pediatric age (>12 years, OR=1.6, p=0.037) and lower extremity primary (OR=0.29, p<0.001). Children with regional nodal disease have improved 3-year overall survival compared to adults (96.5% vs 71.0%, p<0.001).. Children with melanoma have higher rates of nodal disease but better survival than adults. As in adults, there has been a recent increase in close nodal observation rather than CLND for patients with positive SLN. Further study of nodal surveillance for pediatric patients is warranted. Topics: Adult; Child; Humans; Lymph Node Excision; Lymphadenopathy; Melanoma; Sentinel Lymph Node; Sentinel Lymph Node Biopsy; Skin Neoplasms; Syndrome; Transforming Growth Factor beta | 2022 |
MED1 Downregulation Contributes to TGFβ-Induced Metastasis by Inhibiting SMAD2 Ubiquitination Degradation in Cutaneous Melanoma.
Metastasis is the main reason for the high mortality of patients and indeed a difficult task in the treatment of cutaneous melanoma. Therefore, it is of great clinical value to explore the molecular mechanism of cutaneous metastatic melanoma and develop novel therapies. MED1, acting as a factor required for activator-dependent transcription, is reported to be involved in carcinogenesis and progression. In this study, we found that MED1 was highly expressed in patients with cutaneous melanoma. MED1 downregulation could induce cellular epithelial-to-mesenchymal transition and promote migration, invasion, and metastasis of cutaneous melanoma in vivo and in vitro. Further analysis showed that in Med1 knockdown cells, the TGFβ/SMAD2 signaling pathway mediated an increase in epithelial-to-mesenchymal transition phenotype and migration. The opposite results were observed after treatment with TGFβ inhibitors. To further explore the mechanism, we found that MED1 interacted with SMAD2, and MED1 downregulation could protect SMAD2 from degradation by inhibiting SMAD2 ubiquitination. Together, these results suggest that MED1 inhibited TGFβ signaling pathway to reduce cell epithelial-to-mesenchymal transition phenotype and migration through SMAD2 ubiquitination in the metastasis of cutaneous melanoma. Our findings elucidated the role of MED1 in the metastasis of cutaneous melanoma and provided a target for the therapeutic strategies of cutaneous melanoma. Topics: Cell Line, Tumor; Cell Movement; Down-Regulation; Epithelial-Mesenchymal Transition; Humans; Mediator Complex Subunit 1; Melanoma; Melanoma, Cutaneous Malignant; Signal Transduction; Skin Neoplasms; Smad2 Protein; Transforming Growth Factor beta; Ubiquitination | 2022 |
Phosphoinositide-Binding Protein TIPE1 Promotes Alternative Activation of Macrophages and Tumor Progression via PIP3/Akt/TGFβ Axis.
Macrophages perform key and distinct functions in maintaining tissue homeostasis by finely tuning their activation state. Within the tumor microenvironment, macrophages are reshaped to drive tumor progression. Here we report that tumor necrosis factor α-induced protein 8-like 1 (TIPE1) is highly expressed in macrophages and that depletion of TIPE1 impedes alternative activation of macrophages. TIPE1 enhanced activation of the PI3K/Akt pathway in macrophages by directly binding with and regulating the metabolism of phosphatidylinositol 4,5-bisphosphate (PIP2) and phosphatidylinositol 3,4,5-trisphosphate (PIP3). Accordingly, inhibition of the PI3K/Akt pathway significantly attenuated the effect of TIPE1 on macrophage alternative activation. Tumor-associated macrophages (TAM) in human liver cancer and melanoma tissues showed significantly upregulated TIPE1 expression that negatively correlated with patient survival. In vitro and in vivo, TIPE1 knockdown in macrophages retarded the growth and metastasis of liver cancer and melanoma. Furthermore, blockade or depletion of TGFβ signaling in macrophages abrogated the effects of TIPE1 on tumor cell growth and migration. Together, these results highlight that the phosphoinositide-related signaling pathway is involved in reprogramming TAMs to optimize the microenvironment for cancer progression.. This work provides insight into the fine tuning of macrophage polarization and identifies a potential target for macrophage-based antitumor therapy. Topics: Biomarkers, Tumor; Cell Line, Tumor; Humans; Intracellular Signaling Peptides and Proteins; Liver Neoplasms; Macrophages; Melanoma; Phosphatidylinositol 3-Kinases; Phosphatidylinositols; Proto-Oncogene Proteins c-akt; Transforming Growth Factor beta; Tumor Microenvironment | 2022 |
Epidermal melanoma prognostic factors: a link to paracrine transforming growth factor-β signalling.
Topics: Humans; Melanoma; Paracrine Communication; Prognosis; Transforming Growth Factor beta; Transforming Growth Factors | 2022 |
TGFβ activating integrins β6 and β8 are dysregulated in inflammatory skin disease and cutaneous melanoma.
Integrins avβ6 and avβ8 are expressed by keratinocytes and transactivate latent TGFβ. In a murine model, integrin mediated activation of TGFβ has been shown to be critical in maintaining skin homeostasis, specifically playing roles in epidermal retention of Langerhans cells and resident memory cells T cells (Trm).. We examine expression of Integrins β6 and β8 in human skin, inflammatory skin disease, benign nevi, and melanoma and hypothesize that integrin expression is dysregulated in disease.. Using immunohistochemistry, we stained tissue from normal human skin (n = 8), psoriasis (n = 6), atopic dermatitis (n = 6), lichen planus (n = 5), benign nevi (n = 24), and melanoma (n = 25) with anti-integrin β6 and anti-integrin β8 to survey expression pattern. We also performed a retrospective chart review in the melanoma cohort to examine if integrin β6 and β8 expression was associated with increased Breslow depth and worse prognostic staging.. Here, we show that human keratinocytes express integrins β6 and β8, similar to murine keratinocytes. We also found that inflammatory skin conditions have increased Integrin β6, but not Integrin β8 expression. Furthermore, we identified that melanomas have greatly increased expression of integrin β8 compared to nevi. Additionally, high expression of integrin β8 was correlated with greater Breslow depth at diagnosis and with worse prognostic staging.. These findings demonstrate that like murine keratinocytes, human keratinocytes express integrin β6 and β8 under steady state conditions. Moreover, altered integrin expression may participate in the development or maintenance of cutaneous inflammation as well as tumor immune evasion. Topics: Animals; Dermatitis; Humans; Integrin beta Chains; Integrins; Melanoma; Melanoma, Cutaneous Malignant; Mice; Nevus; Retrospective Studies; Skin Neoplasms; Transforming Growth Factor beta | 2022 |
Real-World Adherence to Nodal Surveillance for Sentinel Lymph Node-Positive Melanoma.
Patients with sentinel lymph node-positive (SLN+) melanoma are increasingly undergoing active nodal surveillance over completion lymph node dissection (CLND) since the Second Multicenter Selective Lymphadenectomy Trial (MSLT-II). Adherence to nodal surveillance in real-world practice remains unknown.. In a retrospective cohort of SLN+ melanoma patients who underwent nodal surveillance at a single institution from July 2017 through April 2021, this study evaluated adherence to nodal surveillance ultrasound (US). Adherence to nodal US was compared with adherence to other surveillance methods based on receipt of adjuvant therapy. Early recurrence data were reported using descriptive statistics.. Among 109 SLN+ patients, 37 (34%) received US surveillance at recommended intervals. Of the 72 (66%) non-adherent patients, 16 were lost to follow-up, and 33 had planned follow-up at an outside institution without available records. More patients had a minimum of bi-annual clinic visits (83%) and cross-sectional imaging (53%) compared to those who were adherent with nodal US. The patients who received adjuvant therapy (60%) had fewer ultrasounds (p < 0.01) but more exams (p < 0.01) and a trend toward more cross-sectional imaging (p = 0.06). Of the overall cohort, 26 patients (24%) experienced recurrence at a median follow-up period of 15 months. Of these recurrences, 10 were limited to the SLN basin, and all of these isolated nodal recurrences were resectable.. Pragmatic challenges to real-world delivery of nodal surveillance remain after MSLT-II, and adjuvant therapy appears to be associated with a decreased likelihood of US adherence. Understanding US utility alongside cross-sectional imaging will be critical as increasingly more patients undergo nodal surveillance and adjuvant therapy. Topics: Humans; Lymph Node Excision; Lymph Nodes; Lymphadenopathy; Melanoma; Prognosis; Retrospective Studies; Sentinel Lymph Node; Sentinel Lymph Node Biopsy; Skin Neoplasms; Transforming Growth Factor beta | 2022 |
Human Melanoma-Associated Mast Cells Display a Distinct Transcriptional Signature Characterized by an Upregulation of the Complement Component 3 That Correlates With Poor Prognosis.
Cutaneous melanoma is one of the most aggressive human malignancies and shows increasing incidence. Mast cells (MCs), long-lived tissue-resident cells that are particularly abundant in human skin where they regulate both innate and adaptive immunity, are associated with melanoma stroma (MAMCs). Thus, MAMCs could impact melanoma development, progression, and metastasis by secreting proteases, pro-angiogenic factors, and both pro-inflammatory and immuno-inhibitory mediators. To interrogate the as-yet poorly characterized role of human MAMCs, we have purified MCs from melanoma skin biopsies and performed RNA-seq analysis. Here, we demonstrate that MAMCs display a unique transcriptome signature defined by the downregulation of the FcεRI signaling pathway, a distinct expression pattern of proteases and pro-angiogenic factors, and a profound upregulation of complement component C3. Furthermore, in melanoma tissue, we observe a significantly increased number of C3 Topics: Complement C3; Humans; Mast Cells; Melanoma; Melanoma, Cutaneous Malignant; Peptide Hydrolases; Skin Neoplasms; Transforming Growth Factor beta; Tumor Microenvironment; Up-Regulation | 2022 |
Presence of Dendritic Cell Subsets in Sentinel Nodes of Breast Cancer Patients Is Related to Nodal Burden.
Sentinel lymph nodes (SLNs) are both the first site where breast cancer (BC) metastases form and where anti-tumoral immunity develops. Despite being the most potent antigen-presenting cells, dendritic cells (DCs) located in a nodal tissue can both promote or suppress immune response against cancer in SLNs.. In SLNs excisions obtained from 123 invasive BC patients, we performed immunohistochemistry (IHC) for CD1a, CD1c, DC-LAMP, and DC-SIGN to identify different DCs populations. Then we investigated the numbers of DCs subsets in tumor-free, micrometastatic, and macrometastatic SLNs with the use of a light microscope.. We observed that CD1c+ and DC-SIGN+ DCs were more numerous in SLNs with a larger tumor size. More abundant intratumoral DC-LAMP+ population was related to a higher number of metastatic lymph nodes. Conversely, more abundant CD1a+ DCs were associated with a decreasing nodal burden in SLNs and a lower number of involved lymph nodes. Moreover, densities of the investigated DC populations differed with respect to tumor grade, HER2 overexpression, hormone receptor status, and histologic type of BC.. According to their subtype, DCs are associated with either lower or higher nodal burden in SLNs from invasive BC patients. These relationships appear to be dependent not only on the maturation state of DCs but also on the histological and biological characteristics of the tumor. Topics: Breast Neoplasms; Dendritic Cells; Female; Humans; Lymph Nodes; Lymphadenopathy; Melanoma; Melanoma, Cutaneous Malignant; Sentinel Lymph Node; Sentinel Lymph Node Biopsy; Skin Neoplasms; Transforming Growth Factor beta | 2022 |
National Practice Patterns in the Management of the Regional Lymph Node Basin After Positive Sentinel Lymph Node Biopsy for Cutaneous Melanoma.
Immediate completion lymph node dissection (CLND) for patients with sentinel lymph node (SLN) metastasis from cutaneous melanoma has been replaced largely by ultrasound nodal surveillance since the publication of two landmark trials in 2016 and 2017. National practice patterns of CLND remain poorly characterized.. Patients with a diagnosis of cutaneous melanoma in 2016 and 2018 without clinical nodal disease who underwent sentinel lymph node biopsy (SLNB) were identified from the National Cancer Database (NCDB). Characteristics associated with CLND were analyzed by uni- and multivariate logistic regression. Overall survival (OS) was estimated using Kaplan-Meier and Cox proportional hazards regression analyses.. Of the 3517 patients included in the study, 1405 had disease diagnosed in 2016. The patients with cutaneous melanoma diagnosed in 2016 had a median age of 60 years and a tumor thickness of 2.3 mm compared to 62 years and 2.4 mm, respectively, for the patients with cutaneous melanoma diagnosed in 2018. According to the NCDB, 40 % (n = 559) of the patients underwent CLND in 2016 compared with 6 % (n = 132) in 2018. The factors associated with receipt of CLND in 2018 included younger age (odds ratio [OR], 0.97; 95 % confidence interval [CI], 0.95-0.99; p = 0.001), rural residence (OR, 3.96; 95 % CI, 1.50-10.49; p = 0.006), head/neck tumor location (OR, 1.88; 95 % CI, 1.10-3.23; p = 0.021), and more than one positive SLN (OR, 1.80; 95 % CI, 1.17-2.76; p = 0.007). The 5-year OS did not differ between the patients who received SLNB only and those who underwent CLND (hazard ratio [HR], 0.93; p = 0.54).. The rates of CLND have decreased nationally. However, patients with head/neck primary tumors who live in rural locations are more likely to undergo CLND, highlighting populations for which treatment may be non-uniform with national practice patterns. Topics: Head and Neck Neoplasms; Humans; Lymph Node Excision; Lymphatic Metastasis; Melanoma; Melanoma, Cutaneous Malignant; Middle Aged; Retrospective Studies; Sentinel Lymph Node; Sentinel Lymph Node Biopsy; Skin Neoplasms; Transforming Growth Factor beta | 2022 |
TGF-β-dependent lymphoid tissue residency of stem-like T cells limits response to tumor vaccine.
TGF-β signaling is necessary for CD8 Topics: AIDS Vaccines; Animals; Antigens, Neoplasm; BCG Vaccine; Cancer Vaccines; CD8-Positive T-Lymphocytes; Diphtheria-Tetanus-Pertussis Vaccine; Epitopes; Immunologic Memory; Influenza Vaccines; Lymphoid Tissue; Measles-Mumps-Rubella Vaccine; Melanoma; Mice; Papillomavirus Vaccines; Respiratory Syncytial Virus Vaccines; SAIDS Vaccines; Transforming Growth Factor beta | 2022 |
Effects of Cyclooxygenase-2 in Canine Melanoma-derived Extracellular Vesicles on Tumor Microenvironment
Tumor cell-derived extracellular vesicles (TEVs) promote tumor growth and metastasis; thus, they have drawn the attention of researchers. TEVs regulate the tumor microenvironment by facilitating crosstalk between immune and stromal cells. Macrophages are one of the key components involved in malignant behavior in melanomas. Generally, when activated, macrophages polarize into M1 (pro-inflammatory) or M2 (anti-inflammatory, pro-tumor) phenotypes. However, the role of canine melanoma-derived EVs in macrophage polarization is elusive. In this study, we aimed to analyze the pro- and anti-inflammatory cytokines that are common markers for M1 or M2 macrophages in vitro.. The analysis was performed under coculture conditions of canine melanoma-derived (LMeC) EVs with canine macrophages (DH82). Quantitative reverse transcription polymerase chain reaction, western blotting, and immunofluorescence were used.. Canine melanoma-derived EVs polarized M1 macrophages (inducible nitric oxide synthase, tumor necrosis factor α) into M2 macrophages [cluster of differentiation (CD)206, interleukin-10] and cyclooxygenase-2 is a major factor in macrophage polarization in canine melanoma-derived EVs. Furthermore, we also found that melanoma-derived EVs induced the expression of angiogenic cytokines (vascular endothelial growth factor, transforming growth factor β) in endothelial cells.. Melanoma-derived EVs perform an immunomodulatory function and can be used as targets in anti-inflammatory treatment. Topics: Animals; Cyclooxygenase 2; Cytokines; Dogs; Endothelial Cells; Extracellular Vesicles; Interleukin-10; Melanoma; Nitric Oxide Synthase Type II; Transforming Growth Factor beta; Tumor Microenvironment; Tumor Necrosis Factor-alpha; Vascular Endothelial Growth Factor A | 2022 |
A single-cell analysis reveals tumor heterogeneity and immune environment of acral melanoma.
Acral melanoma is a dismal subtype of melanoma occurring in glabrous acral skin, and has a higher incidence in East Asians. We perform single-cell RNA sequencing for 63,394 cells obtained from 5 acral and 3 cutaneous melanoma samples to investigate tumor heterogeneity and immune environment. We define 5 orthogonal functional cell clusters that are involved in TGF-beta signaling, Type I interferon, Wnt signaling, Cell cycle, and Cholesterol efflux signaling. Signatures of enriched TGF-beta, Type I interferon, and cholesterol efflux signaling are significantly associated with good prognosis of melanoma. Compared with cutaneous melanoma, acral melanoma samples have significantly severe immunosuppressive state including depletion of cytotoxic CD8+ T cells, enrichment of Treg cells, and exhausted CD8+ T cells. PD1 and TIM-3 have higher expression in the exhaustive CD8+ T cells of acral melanoma. Key findings are verified in two independent validation sets. This study contributes to our better understanding of acral melanoma. Topics: Cholesterol; Humans; Interferon Type I; Melanoma; Melanoma, Cutaneous Malignant; Single-Cell Analysis; Skin Neoplasms; Transforming Growth Factor beta | 2022 |
Proteomics Analysis of Lymphatic Metastasis-Related Proteins Using Highly Metastatic Human Melanoma Cells Originated by Sequential in Vivo Implantation.
Metastasis of cancer cells to lymph nodes (LN) is a common modality of metastasis in clinical settings, but the mechanisms involved in lymphatic metastasis remain unclear compared to hematogenous metastasis to bones and the brain. To elucidate the molecular mechanisms responsible for melanoma LN metastasis, we first generated LN metastasis-prone melanoma cells (C8161F2) by the sequential in vivo transplantation of parental melanoma cells (C8161F0). Although the in vitro/in vivo proliferative potential of these melanoma cells were similar, the metastatic potential of the C8161F2 for LNs was significantly enhanced. We then conducted a proteomics analysis to identify the proteins and pathways that contribute to LN metastasis. We identified six proteins (three: up-regulated and three: down-regulated) whose expressions were statistically significantly different by more than 2-fold in the two cell groups. Some of these genes are responsible for the activation of the transforming growth factor-β (TGF-β)-related pathway, a well-known inducer of epithelial-mesenchymal transition (EMT). In addition, a gene ontology analysis revealed that the enhanced cell-cell adhesion appears to be involved in lymphatic metastasis. In conclusion, we established highly lymphatic metastatic melanoma cells, which would be valuable for studies of the molecular mechanisms responsible for lymphatic metastasis. Topics: Animals; Cell Adhesion; Cell Line, Tumor; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Lymph Nodes; Lymphatic Metastasis; Male; Melanoma; Mice; Proteomics; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta; Xenograft Model Antitumor Assays | 2021 |
Knockdown of circular RNA VANGL1 inhibits TGF-β-induced epithelial-mesenchymal transition in melanoma cells by sponging miR-150-5p.
Melanoma is one of the most aggressive and life-threatening skin cancers, and in this research, we aimed to explore the functional role of circular RNA VANGL1 (circVANGL1) in melanoma progression. The expression levels of circVANGL1 were observed to be significantly increased in clinical melanoma tissues and cell lines. Moreover, circVANGL1 knockdown suppressed, while circVANGL1 overexpression promoted the proliferation, migration and invasion abilities of melanoma cells. Further investigations confirmed the direct binding relation between circVANGL1 and miR-150-5p in melanoma, and restoration of miR-150-5p blocked the effects of circVANGL1 overexpression in melanoma cells. We further found that circVANGL1 was up-regulated by TGF-β treatment, and the enhanced EMT of TGF-β-treated melanoma cells was blocked by circVANGL1 knockdown. In conclusion, these results indicated that circVANGL1 might serve as a promising therapeutic target for melanoma. Topics: Apoptosis; Carrier Proteins; Cell Line, Tumor; Cell Movement; Cell Proliferation; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Membrane Proteins; MicroRNAs; RNA, Circular; RNA, Long Noncoding; Skin Neoplasms; Transforming Growth Factor beta; Up-Regulation | 2021 |
Comprehensive upstream and downstream regulatory analyses identify miR-675-3p as a potential prognostic biomarker in melanoma.
This study assessed miR-675-3p-related regulatory mechanisms in melanoma and the clinical relevance of such regulatory activities. We downloaded miRNA mature strand expression RNA-Seq, phenotypic, and DNA methylation data pertaining to the TCGA Melanoma cohort. Differentially expressed miRNAs (DEMs) between metastatic and primary melanoma patient tissues were then identified, and miR-675-3p expression in melanoma patient peripheral blood was confirmed using the GSE20994 GEO dataset, while its expression in melanoma cell lines was evaluated via qRT-RCR. The clinical and prognostic implications of miR-675-3p in melanoma were assessed, and miR-675-3p target genes were identified using bioinformatics tools. Functional roles of this miRNA were explored via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. We identified 3 and 22 miRNAs that were up- and downregulated, respectively, in metastatic melanoma samples relative to primary melanoma samples. Upregulation of miR-675-3p was associated with poorer overall patient survival, tumor histologic grade, and Clark's level. Consistently, miR-675-3p was also overexpressed in the peripheral blood of melanoma patients relative to healthy controls, and in melanoma cell lines relative to control cells. Gene regulatory networks indicated that 32 transcription factors control miR-675-3p expression, and that it, in turn, regulates 10 target genes. KEGG analyses indicated that these genes were associated with cell cycle, transcriptional misregulation in cancer, TGF-beta signaling, and HIF-1 signaling pathways. Gain-of-function assays revealed that miR-675-3p could promote cell proliferation via accelerating cell cycle progression. Western blotting results indicated that miR-675-3p could active TGF-beta and HIF-1 signaling. Through upstream and downstream analyses of miR-675-3p-related regulatory activity, we confirmed that this miRNA participates in key melanoma-related processes and offers value as a prognostic biomarker in melanoma patients. Topics: Aged; Biomarkers, Tumor; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Data Analysis; Datasets as Topic; Female; Gene Expression; Gene Expression Regulation, Neoplastic; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Male; Melanoma; MicroRNAs; Prognosis; Signal Transduction; Transforming Growth Factor beta | 2021 |
Tumor suppressive microRNA-485-5p targets PRRX1 in human skin melanoma cells, regulating epithelial-mesenchymal transition and apoptosis.
Melanoma is one of the most aggressive skin cancers. Existing evidence has reported the aberrant expression of microRNAs (miRNAs) in melanoma, but their putative targets and underlying downstream effects remain to be further understood. Herein, we explored the suppressive role of miR-485-5p in melanoma progression. Initial bioinformatics analyses showed that the PRRX1 gene was differentially expressed in melanoma, while miR-485-5p was predicted to be a potential regulatory miRNA binding to PRRX1 mRNA. We confirmed that PRRX1 was upregulated, while miR-485-5p was downregulated in human melanoma samples compared with adjacent normal skin tissues. We then showed that PRRX1 was a target gene of miR-485-5p by dual-luciferase reporter gene assay. Moreover, a reduction in the expression of PRRX1 and downregulation of important proteins of the transforming growth factor-beta (TGFβ) signaling pathway was observed after miR-485-5p overexpression. Furthermore, miR-485-5p overexpression or PRRX1 knockdown suppressed epithelial-mesenchymal transition, cell viability, migration, and invasion, and promoted cell apoptosis in melanoma cells. Our study demonstrates the tumor-suppressive functions of miR-485-5p in the development of human melanoma, providing a potential target for therapy. Topics: Adult; Aged; Apoptosis; Cell Line, Tumor; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Homeodomain Proteins; Humans; Male; Melanoma; MicroRNAs; Middle Aged; Transforming Growth Factor beta | 2021 |
pH-Triggered Copper-Free Click Reaction-Mediated Micelle Aggregation for Enhanced Tumor Retention and Elevated Immuno-Chemotherapy against Melanoma.
Topics: Cell Line, Tumor; Click Chemistry; Doxorubicin; Humans; Hydrogen-Ion Concentration; Immunotherapy; Melanoma; Micelles; Polyethylene Glycols; Signal Transduction; Smad3 Protein; Transforming Growth Factor beta | 2021 |
Blockade of transforming growth factor β2 by anti-sense oligonucleotide improves immunotherapeutic potential of IL-2 against melanoma in a humanized mouse model.
IL-2 is a potent cytokine that activates natural killer cells and CD8+ cytotoxic T lymphocytes (CTLs) and has been approved for the treatment of metastatic renal cell carcinoma and metastatic melanoma. However, the medical use of IL-2 is restricted because of its narrow therapeutic window and potential side effects, including the expansion of regulatory T cells (Tregs).. In this study, the authors investigated the complementary effects of transforming growth factor-β2 (TGF-β2) anti-sense oligodeoxynucleotide (TASO) on the immunotherapeutic potential of IL-2 in a melanoma-bearing humanized mouse model.. The authors observed that the combination of TASO and IL-2 facilitated infiltration of CTLs into the tumor, thereby potentiating the tumor killing function of CTLs associated with increased granzyme B expression. In addition, TASO attenuated the increase in Tregs by IL-2 in the peripheral blood and spleen and also inhibited infiltration of Tregs into the tumor, which was partly due to decreased CCL22. Alteration of T-cell constituents at the periphery by TGF-β2 inhibition combined with IL-2 might be associated with the synergistic augmentation of serum pro-inflammatory cytokines (such as interferon γ and tumor necrosis factor α) and decreased ratio of Tregs to CTLs in tumor tissues, which consequently results in significant inhibition of tumor growth CONCLUSIONS: These results indicate that the application of TASO improves IL-2-mediated anti-tumor immunity, thus implying that blockade of TGF-β2 in combination with IL-2 may be a promising immunotherapeutic strategy for melanoma. Topics: Animals; Carcinoma, Renal Cell; Immunotherapy; Interleukin-2; Kidney Neoplasms; Melanoma; Mice; Oligonucleotides, Antisense; Transforming Growth Factor beta; Transforming Growth Factor beta2; Transforming Growth Factors | 2021 |
ZNF224 is a mediator of TGF-β pro-oncogenic function in melanoma.
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 |
Immunosuppressive activity is attenuated by Astragalus polysaccharides through remodeling the gut microenvironment in melanoma mice.
Astragalus polysaccharides (APS), the main effective component of Astragalus membranaceus, can inhibit tumor growth, but the underlying mechanisms remain unclear. Previous studies have suggested that APS can regulate the gut microenvironment, including the gut microbiota and fecal metabolites. In this work, our results showed that APS could control tumor growth in melanoma-bearing mice. It could reduce the number of myeloid-derived suppressor cells (MDSC), as well as the expression of MDSC-related molecule Arg-1 and cytokines IL-10 and TGF-β, so that CD8 Topics: Animals; Anti-Bacterial Agents; Arginase; Astragalus Plant; Bifidobacterium; CD8-Positive T-Lymphocytes; Drug Combinations; Fecal Microbiota Transplantation; Feces; Gastrointestinal Microbiome; Immune Tolerance; Interleukin-10; Interleukin-1beta; Interleukin-6; Lactobacillus; Male; Melanoma; Mice; Mice, Inbred C57BL; Myeloid-Derived Suppressor Cells; Polysaccharides; RNA, Ribosomal, 16S; Transforming Growth Factor beta; Tumor Microenvironment | 2021 |
Promotion effects of mono-2-ethyhexyl phthalate (MEHP) on migration and invasion of human melanoma cells via activation of TGF-β signals.
Malignant melanoma is one of the most leading forms of skin cancer associated with a low patient survival rate. There is an urgent need to illustrate risk factors that can trigger the motility of melanoma cancer cells. Our present study revealed that mono-(2-ethylhexyl)phthalate (MEHP) exposure can significantly increase the in vitro migration and invasion of WM983A and A375 cells. Among the tested cytokines, MEHP can increase the expression of transforming growth factor β (TGF-β). Inhibition of TGF-β via its neutralization antibody can attenuate MEHP-induced cell migration and invasion. Further, upregulation of TGF-β mediated MEHP-induced activation of Smad signals and upregulation of Snail in melanoma cells. Blocking the TGF-β/Smad signal pathway can attenuate MEHP-induced cell migration. Estrogen receptor α (ERα) was essential for MEHP-induced expression of TGF-β. In addition, MEHP can increase the expression of ERα in melanoma cells. Collectively, our study found that MEHP can stimulate the progression of melanoma via TGF-β signals. SIGNIFICANCE: Mono-(2-ethylhexyl)phthalate (MEHP) is the active and most toxic metabolite of di(2-ethylhexyl)phthalate (DEHP). Our present study revealed that MEHP can trigger the in vitro migration and invasion of melanoma cells via upregulation of TGF-β/Snail signals. It revealed that daily exposure to MEHP might be a risk factor for melanoma patients. Topics: Antineoplastic Agents; Cell Movement; Cell Proliferation; Dose-Response Relationship, Drug; Drug Screening Assays, Antitumor; Humans; Melanoma; Melanoma, Cutaneous Malignant; Phthalic Acids; Signal Transduction; Skin Neoplasms; Structure-Activity Relationship; Transforming Growth Factor beta; Tumor Cells, Cultured | 2020 |
Autophagy inhibition potentiates the anti-EMT effects of alteronol through TGF-β/Smad3 signaling in melanoma cells.
Accumulating evidence demonstrated that alteronol, a novel compound that has a similar structure with paclitaxel, exerts anticancer effects against diversified tumors. However, whether alteronol induces autophagy and the relationship between its anticancer effects and autophagy in melanoma remains elusive. In this study, we show that alteronol induces not only anti-proliferation activity and apoptosis but also autophagy in A375 and UACC62 cells. In addition, alteronol inhibits A375 and UACC62 cells invasion and migration by preventing the epithelial-mesenchymal transition (EMT). Blocking autophagy enhances alteronol-induced apoptosis and anti-EMT effects in vitro and in vivo. Mechanistically, we find that alteronol significantly inhibits Akt/mTOR and TGFβ/Smad3 pathways, and co-treatment with autophagy inhibitor 3-MA further potentiate these effects. Our results suggest that alteronol induces cyto-protective autophagy in melanoma cells through inhibition of Akt/mTOR pathway, thus attenuates apoptosis and promotes melanoma cell EMT through TGF-β/Smad3 pathway. Combination with alteronol and autophagy inhibitor 3-MA may be a potential treatment for melanoma as it not only significantly inhibited tumor growth but also suppressed tumor invasion and migration as anti-metastasis agent. Topics: Animals; Autophagy; Epithelial-Mesenchymal Transition; Humans; Male; Melanoma; Mice; Naphthoquinones; Signal Transduction; Transforming Growth Factor beta | 2020 |
Host dysbiosis negatively impacts IL-9-producing T-cell differentiation and antitumour immunity.
Host-microbiota interactions shape T-cell differentiation and promote tumour immunity. Although IL-9-producing T cells have been described as potent antitumour effectors, their role in microbiota-mediated tumour control remains unclear.. We analysed the impact of the intestinal microbiota on the differentiation of colonic lamina propria IL-9-producing T cells in germ-free and dysbiotic mice. Systemic effects of the intestinal microbiota on IL-9-producing T cells and the antitumour role of IL-9 were analysed in a model of melanoma-challenged dysbiotic mice.. We show that germ-free mice have lower frequency of colonic lamina propria IL-9-producing T cells when compared with conventional mice, and that intestinal microbiota reconstitution restores cell frequencies. Long-term antibiotic treatment promotes host dysbiosis, diminishes intestinal IL-4 and TGF-β gene expression, decreases the frequency of colonic lamina propria IL-9-producing T cells, increases the susceptibility to tumour development and reduces the frequency of IL-9-producing T cells in the tumour microenvironment. Faecal transplant restores intestinal microbiota diversity, and the frequency of IL-9-producing T cells in the lungs of dysbiotic animals, restraining tumour burden. Finally, recombinant IL-9 injection enhances tumour control in dysbiotic mice.. Host-microbiota interactions are required for adequate differentiation and antitumour function of IL-9-producing T cells. Topics: Animals; Anti-Bacterial Agents; Cell Differentiation; Cell Line, Tumor; Dysbiosis; Fecal Microbiota Transplantation; Gastrointestinal Microbiome; Germ-Free Life; Interleukin-4; Interleukin-9; Male; Melanoma; Mice; Mucous Membrane; Neoplasm Transplantation; T-Lymphocytes; Transforming Growth Factor beta; Tumor Microenvironment | 2020 |
Targeting Extracellular Matrix Remodeling Restores BRAF Inhibitor Sensitivity in BRAFi-resistant Melanoma.
The extracellular matrix (ECM) is an intriguing, yet understudied component of therapy resistance. Here, we investigated the role of ECM remodeling by the collagenase, MT1-MMP, in conferring resistance of v-Raf murine sarcoma viral oncogene homolog B1 (BRAF)-mutant melanoma to BRAF inhibitor (BRAFi) therapy.. Publicly available RNA-sequencing data and reverse phase protein array were used to determine the relevance of MT1-MMP upregulation in BRAFi-resistant melanoma in patients, patient-derived xenografts, and cell line-derived tumors. Short hairpin RNA (shRNA)-mediated knockdown of MT1-MMP, inhibition via the selective MT1-MMP/MMP2 inhibitor, ND322, or overexpression of MT1-MMP was used to assess the role of MT1-MMP in mediating resistance to BRAFi.. MT1-MMP was consistently upregulated in posttreatment tumor samples derived from patients upon disease progression and in melanoma xenografts and cell lines that acquired resistance to BRAFi. shRNA- or ND322-mediated inhibition of MT1-MMP synergized with BRAFi leading to resensitization of resistant cells and tumors to BRAFi. The resistant phenotype depends on the ability of cells to cleave the ECM. Resistant cells seeded in MT1-MMP uncleavable matrixes were resensitized to BRAFi similarly to MT1-MMP inhibition. This is due to the inability of cells to activate integrinβ1 (ITGB1)/FAK signaling, as restoration of ITGB1 activity is sufficient to maintain resistance to BRAFi in the context of MT1-MMP inhibition. Finally, the increase in MT1-MMP in BRAFi-resistant cells is TGFβ dependent, as inhibition of TGFβ receptors I/II dampens MT1-MMP overexpression and restores sensitivity to BRAF inhibition.. BRAF inhibition results in a selective pressure toward higher expression of MT1-MMP. MT1-MMP is pivotal to an ECM-based signaling pathway that confers resistance to BRAFi therapy. Topics: Animals; Arginine; Cell Line, Tumor; Drug Resistance, Neoplasm; Focal Adhesion Kinase 1; Gene Expression Regulation, Neoplastic; Heterografts; Humans; Integrin beta1; Matrix Metalloproteinase 14; Melanoma; Mice; Protein Kinase Inhibitors; Proto-Oncogene Proteins B-raf; Signal Transduction; Sulfones; Transforming Growth Factor beta; Vemurafenib | 2020 |
Targeting PSMD14 inhibits melanoma growth through SMAD3 stabilization.
Although melanoma therapy is improved by novel molecular targeted reagents, including vemurafenib, aberrant proliferation and early metastasis remain obstacles for melanoma; therefore, novel target molecules for melanoma need to be identified. In this study, we focused on deubiquitinating enzymes, which regulate protein stability through ubiquitin-proteasome systems, and identified 26S proteasome non-ATPase regulatory subunit 14 (PSMD14) as a molecule related to melanoma growth using siRNA library screening. Similar to a previous report, PSMD14 knockdown strongly induced p21 expression and inhibited RB phosphorylation in melanoma. After in silico analysis, TGF-β signaling was identified as a negatively correlated gene set with PSMD14 expression. Although TGF-β signaling is also related to the invasive phenotype of melanoma, PSMD14 knockdown suppressed melanoma migration and reduced SLUG expression, suggesting that targeting PSMD14 suppresses both growth and migration. Furthermore, SMAD3 expression increased in nucleus and SMAD3 degradation was delayed after PSMD14 knockdown. Thus, our present study suggests that targeting PSMD14 can inhibit melanoma growth and migration through either SMAD3 accumulation or SLUG reduction, respectively. Topics: Cell Line, Tumor; Cell Proliferation; Gene Knockdown Techniques; Humans; Melanoma; Phosphorylation; Proteasome Endopeptidase Complex; RNA, Small Interfering; Signal Transduction; Skin Neoplasms; Smad3 Protein; Trans-Activators; Transforming Growth Factor beta | 2020 |
Regulatory T Cells Induce Metastasis by Increasing Tgf-β and Enhancing the Epithelial–Mesenchymal Transition.
Malignant melanoma is the most aggressive form of skin cancer; a substantial percentage of patients present with distant metastases. However, the mechanism of metastasis is not well understood. Here, we demonstrate that the administration of exogenous regulatory T cells (Tregs) into melanoma tumor-bearing mice results in a significant increase in lung metastasis. An increase in the invasive and metastatic phenotype of melanoma was mediated by cell-to-cell contact between melanoma cells and Tregs, which elevated the expression level of transforming growth factor-β (TGF-β) and the subsequent induction of the epithelial-to-mesenchymal transition (EMT).B16-BL6 melanoma tumors co-cultured with Tregs showed a larger population of migrating cells compared to B16-BL6 tumors cultured without Tregs. Additionally, the injection of exogenous Tregs into B16-BL6 melanoma tumors led to the recruitment and infiltration of endogenous Tregs into tumor tissues, thus increasing the overall Treg percentage in the tumor infiltrating lymphocyte population. Collectively, our findings propose novel mechanisms in which exogenous Treg-dependent upregulation of TGF-β and mesenchymal markers is important for augmenting the migration capacity and invasiveness of melanoma, thereby contributing to the metastasis. Topics: Animals; Cell Line, Tumor; Cell Movement; Epithelial-Mesenchymal Transition; Lung Neoplasms; Lymphocytes, Tumor-Infiltrating; Male; Melanoma; Melanoma, Cutaneous Malignant; Mice; Mice, Inbred C57BL; Skin Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Up-Regulation | 2019 |
SMAD7 is a novel independent predictor of survival in patients with cutaneous melanoma.
Overexpression of SMAD7-a hallmark inhibitor of transforming growth factor β (TGFβ) signaling-has been documented and related with adverse prognosis in a number of epithelial malignancies, suggesting that it may be responsible for resistance to TGFβ-induced growth arrest of cancer cells. The involvement of SMAD7 in development and progression of malignant melanoma is unclear, and its expression has not been characterized so far at the protein level in clinical melanoma tissue samples. We evaluated SMAD7 expression in 205 skin melanoma primary tumors by immunohistochemistry and correlated the findings with clinicopathological profiles of patients. Melanocytic SMAD7 was evidenced in 204 cases, and the expression pattern was predominantly nuclear. High expression of SMAD7 was positively associated with several features of tumor aggressiveness, for example, presence of ulceration (P < 0.001), higher tumor thickness (P < 0.001), and mitotic rate (P < 0.001), but not presence of regional or distant metastases. Moreover, high SMAD7 expression independently predicted unfavorable outcome: melanoma-specific survival (hazard ratio = 3.16, P < 0.001) and recurrence-free survival (hazard ratio = 2.88, P < 0.001). Taken together, our results underline the importance of TGFβ signaling in cancer and define SMAD7 as a marker of aggressive tumor behavior and adverse clinical outcomes in melanoma patients. Topics: Adolescent; Adult; Aged; Aged, 80 and over; Female; Humans; Immunohistochemistry; Male; Melanocytes; Melanoma; Middle Aged; Signal Transduction; Skin Neoplasms; Smad7 Protein; Transforming Growth Factor beta; Young Adult | 2019 |
MicroRNA-92 Expression in CD133
In addition to being refractory to treatment, melanoma cancer stem cells (CSC) are known to suppress host antitumor immunity, the underlying mechanisms of which need further elucidation. In this study, we established a novel role for miR-92 and its associated gene networks in immunosuppression. CSCs were isolated from the B16-F10 murine melanoma cell line based on expression of the putative CSC marker CD133 (Prominin-1). CD133 Topics: AC133 Antigen; Animals; Female; Humans; Immune Tolerance; Integrin alpha Chains; Melanoma; Melanoma, Experimental; Mice; Mice, Inbred C57BL; MicroRNAs; Neoplastic Stem Cells; Transforming Growth Factor beta; Tumor Microenvironment | 2019 |
Interleukin-37 is highly expressed in regulatory T cells of melanoma patients and enhanced by melanoma cell secretome.
Immune suppression is one of the 10 hallmarks of cancer. Interleukin-37 (IL-37), a member of the IL-1 family, inhibits both innate and adaptive immunity, and has been shown to modulate immune responses in various disease conditions. Yet, IL-37 has rarely been investigated in cancer patients, and its biological role in cancer remains to be elucidated. In this study, we investigated the gene expression of IL-37 in age- and sex-matched blood samples of healthy individuals and melanoma patients, and demonstrated upregulation of IL-37 messenger RNA (mRNA) in the blood samples of melanoma patients. By further analyzing immune cell subsets responsible for the upregulated IL-37 expression, we discovered that IL-37 mRNA was highly expressed in T cells and granulocytes, with the highest expression in regulatory T (T Topics: Biomarkers, Tumor; Cells, Cultured; Female; Humans; Interleukin-1; Male; Melanoma; RNA, Messenger; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Up-Regulation | 2019 |
Nodal Facilitates Differentiation of Fibroblasts to Cancer-Associated Fibroblasts that Support Tumor Growth in Melanoma and Colorectal Cancer.
Fibroblasts become cancer-associated fibroblasts (CAFs) in the tumor microenvironment after activation by transforming growth factor-β (TGF-β) and are critically involved in cancer progression. However, it is unknown whether the TGF superfamily member Nodal, which is expressed in various tumors but not expressed in normal adult tissue, influences the fibroblast to CAF conversion. Here, we report that Nodal has a positive correlation with α-smooth muscle actin (α-SMA) in clinical melanoma and colorectal cancer (CRC) tissues. We show the Nodal converts normal fibroblasts to CAFs, together with Snail and TGF-β signaling pathway activation in fibroblasts. Activated CAFs promote cancer growth in vitro and tumor-bearing mouse models in vivo. These results demonstrate that intercellular crosstalk between cancer cells and fibroblasts is mediated by Nodal, which controls tumor growth, providing potential targets for the prevention and treatment of tumors. Topics: Actins; Animals; Cancer-Associated Fibroblasts; Cell Differentiation; Cell Line; Colorectal Neoplasms; Female; Humans; Melanoma; Mice; Mice, Nude; Nodal Protein; Proliferating Cell Nuclear Antigen; RNA Interference; RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta; Transplantation, Heterologous | 2019 |
Transcriptional repression of the tyrosinase-related protein 2 gene by transforming growth factor-β and the Kruppel-like transcription factor GLI2.
Tyrosinase-Related Protein 2 (TRP2) is an enzyme involved in melanogenesis, that also exerts proliferative, anti-apoptotic and immunogenic functions in melanoma cells. TRP2 transcription is regulated by the melanocytic master transcription factor MITF. GLI2, a transcription factor that acts downstream of Hedgehog signaling, is also a direct transcriptional target of the TGF-β/SMAD pathway that contributes to melanoma progression and exerts transcriptional antagonistic activities against MITF.. To characterize the molecular events responsible for TGF-β and GLI2 repression of TRP2 expression.. In silico promoter analysis, transient cell transfection experiments with 5'-end TRP2 promoter deletion constructs, chromatin immuno-precipitation, and site-directed promoter mutagenesis were used to dissect the molecular mechanisms of TRP2 gene regulation by TGF-β and GLI2.. We demonstrate that TGF-β and GLI2-specific TRP2 repression involves direct mechanisms that occur in addition to MITF downregulation by TGF-β and GLI2. We identify two functional GLI2 binding sites within the TRP2 promoter that are critical for TGF-β and GLI2 responsiveness, one of them overlapping a CREB binding site. GLI2 and CREB competing for the same cis-element is associated with opposite transcriptional outcome.. Our results further refine the understanding of how TGF-β and GLI2 control the phenotypic plasticity of melanoma cells. In particular, we identify critical GLI2-binding cis-elements within the TRP2 promoter region that allow for its transcriptional repression independently from MITF concomitant downregulation. Topics: Cell Line, Tumor; Computational Biology; Gene Expression Regulation, Neoplastic; Humans; Intramolecular Oxidoreductases; Melanoma; Mutagenesis, Site-Directed; Nuclear Proteins; Promoter Regions, Genetic; Skin Neoplasms; Transcription, Genetic; Transforming Growth Factor beta; Zinc Finger Protein Gli2 | 2019 |
Adipocytes sensitize melanoma cells to environmental TGF-β cues by repressing the expression of miR-211.
Transforming growth factor-β (TGF-β) superfamily members are critical signals in tissue homeostasis and pathogenesis. Melanoma grows in the epidermis and invades the dermis before metastasizing. This disease progression is accompanied by increased sensitivity to microenvironmental TGF-β. Here, we found that skin fat cells (adipocytes) promoted metastatic initiation by sensitizing melanoma cells to TGF-β. Analysis of melanoma clinical samples revealed that adipocytes, usually located in the deeper hypodermis layer, were present in the upper dermis layer within proximity to in situ melanoma cells, an observation that correlated with disease aggressiveness. In a coculture system, adipocytes secreted the cytokines IL-6 and TNF-α, which induced a proliferative-to-invasive phenotypic switch in melanoma cells by repressing the expression of the microRNA miR-211. In a xenograft model, miR-211 exhibited a dual role in melanoma progression, promoting cell proliferation while inhibiting metastatic spread. Bioinformatics and molecular analyses indicated that miR-211 directly targeted and repressed the translation of Topics: Adipocytes; Animals; Cell Proliferation; Coculture Techniques; Disease Progression; Gene Expression Regulation, Neoplastic; Humans; Interleukin-6; Ligands; Melanoma; Mice; MicroRNAs; Neoplasm Metastasis; Neoplasm Transplantation; NIH 3T3 Cells; Phenotype; Signal Transduction; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha | 2019 |
Mechanism of MicroRNA-708 Targeting BAMBI in Cell Proliferation, Migration, and Apoptosis in Mice With Melanoma via the Wnt and TGF-β Signaling Pathways.
The aim of this study was to evaluate the mechanisms involved with miRNA-708 and its targeting of bone morphogenetic protein and activin membrane-bound inhibitor in cell proliferation, migration, and apoptosis in mice with melanoma via the Wnt and transforming growth factor β signaling pathways.. Sixty mice were recruited of which 40 were subsequently assigned into the experimental group (22 mice were successfully established as melanoma model and 18 mice used in tumor xenograft), and the normal control group consisted of 20 mice. B16 cells were assigned to the normal, blank, and negative control, miR-708 mimics, miR-708 inhibitors, si-BAMBI, and miR-708 inhibitors + si-bone morphogenetic protein and activin membrane-bound inhibitor groups. Western blotting and reverse transcription quantitative polymerase chain reaction were employed to detect the expression levels within the tissues and cell lines. TCF luciferase reporter (TOP-FLASH) or a control vector (FOP-FLASH) was applied to detect the activity of the Wnt signaling pathway. MTT3-(4,5-Dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide assay, flow cytometry, scratch test, and Transwell assay were conducted, respectively, for cell proliferation, apoptosis, migration, and invasion, while tumor xenograft procedures were performed on the nude mice recruited for the study.. Compared to the normal control group, the model group displayed increased expressions of bone morphogenetic protein and activin membrane-bound inhibitor, Wnt10B, P53, and Bcl-2; TOPflash activity; β-catenin expression; cell proliferation; migration; and invasion capabilities while decreased expressions of miR-708, vascular endothelial growth factor, Fas, Bax, Caspase-3, and cleaved Caspase-3 and apoptosis rate. Compared to the blank and negative control groups, the miR-708 mimics and small-interfering RNA-bone morphogenetic protein and activin membrane-bound inhibitor groups exhibited decreases expressions of bone morphogenetic protein and activin membrane-bound inhibitor, Wnt10B, P53, and Bcl-2 and decreased proliferation, migration, and invasion capabilities, while increases in the apoptosis rate, expressions of vascular endothelial growth factor, Fas, Bax, Caspase-3, and cleaved Caspase-3; however, downregulated levels of TOPflash activity and β-catenin expression were recorded. The miR-708 inhibitors group displayed an opposite trend.. Downregulation of miR-708-targeted bone morphogenetic protein and activin membrane-bound inhibitor inhibits the proliferation and migration of melanoma cells through the activation of the transforming growth factor β pathway and the suppression of Wnt pathway. Topics: Animals; Apoptosis; Biomarkers; Cell Cycle; Cell Line; Cell Line, Tumor; Cell Movement; Cell Proliferation; Disease Models, Animal; Gene Expression Regulation, Neoplastic; Genes, Reporter; Humans; Immunohistochemistry; Male; Melanoma; Membrane Proteins; Mice; MicroRNAs; RNA Interference; Transforming Growth Factor beta; Wnt Signaling Pathway; Xenograft Model Antitumor Assays | 2018 |
Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFβ enhance the efficacy of cancer immunotherapy.
Topics: Animals; Antibodies; B7-H1 Antigen; Breast Neoplasms; Cohort Studies; CTLA-4 Antigen; Female; Forkhead Transcription Factors; Humans; Immunotherapy; Lymphocytes, Tumor-Infiltrating; Melanoma; Mice; Mice, Inbred NOD; Receptors, Transforming Growth Factor beta; T-Lymphocytes, Regulatory; Th1 Cells; Th17 Cells; Transforming Growth Factor beta | 2018 |
Stromal Fibroblasts Mediate Anti-PD-1 Resistance via MMP-9 and Dictate TGFβ Inhibitor Sequencing in Melanoma.
Although anti-PD-1 therapy has improved clinical outcomes for select patients with advanced cancer, many patients exhibit either primary or adaptive resistance to checkpoint inhibitor immunotherapy. The role of the tumor stroma in the development of these mechanisms of resistance to checkpoint inhibitors remains unclear. We demonstrated that pharmacologic inhibition of the TGFβ signaling pathway synergistically enhanced the efficacy of anti-CTLA-4 immunotherapy but failed to augment anti-PD-1/PD-L1 responses in an autochthonous model of BRAF Topics: Animals; B7-H1 Antigen; CTLA-4 Antigen; Drug Resistance, Neoplasm; Female; Fibroblasts; Immunotherapy; Male; Matrix Metalloproteinase 9; Melanoma; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Transgenic; Proto-Oncogene Proteins B-raf; Pyrazoles; Quinolines; Transforming Growth Factor beta; Tumor Microenvironment; Xenograft Model Antitumor Assays | 2018 |
Seeking Synergy of Checkpoint Blockade through TGFβ Inhibition.
Immunotherapy, including checkpoint blockade, is revolutionizing the treatment of cancers, but efficacy has, however, been limited to a subset of patients and can be of limited duration due to primary and acquired resistance to treatment. An article in this issue shows that inhibiting TGFβ can overcome resistance to blockade of one immune checkpoint, but not another, unless one follows where the mechanism leads. Topics: CTLA-4 Antigen; Fibroblasts; Humans; Melanoma; Programmed Cell Death 1 Receptor; Transforming Growth Factor beta | 2018 |
Integrin β1 activation induces an anti-melanoma host response.
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 |
Neokestose suppresses the growth of human melanoma A2058 cells via inhibition of the nuclear factor‑κB signaling pathway.
Neokestose has superior prebiotic effects compared with the commercial fructooligosaccharides (FOS). In addition, the branched structure of neokestose, a type of neo‑FOS, confers improved chemical stability compared with conventional FOS; therefore, the investigation of the branched structure by the present study may be of high biomedical value. The present study aimed to determine whether neokestose may suppress growth of the A2058 melanoma cell line. The cells were initially treated with neokestose; subsequently, in vitro cytotoxicity was assessed using MTT, and cell cycle progression and apoptosis were detected using flow cytometry. The protein expression levels of cyclin D1, phosphorylated (p)‑inhibitor of κB (IκB) and nuclear factor‑κB (NF‑κB) were determined using western blotting. Treatment with neokestose led to a dose‑dependent inhibition of cell viability. Flow cytometry data indicated that neokestose increased the sub‑G1 cell population, and induced early and late apoptosis. Western blot analysis revealed that neokestose treatment reduced the expression levels of p‑IκB and cyclin D1. These findings suggest that neokestose treatment may induce suppression of A2058 melanoma cell viability via inhibition of the NF‑κB pathway. The present findings support the requirement for further investigation into the potential use of neokestose as an additional or chemopreventive therapeutic agent for the treatment of melanoma. Topics: Apoptosis; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Cell Survival; Cyclin D1; Gene Expression; Humans; Melanoma; NF-kappa B; Phosphorylation; Signal Transduction; Transforming Growth Factor beta; Trisaccharides | 2017 |
UV radiation promotes melanoma dissemination mediated by the sequential reaction axis of cathepsins-TGF-β1-FAP-α.
Ultraviolet radiation (UVR) is the major risk factor for development of malignant melanoma. Fibroblast activation protein (FAP)-α is a serine protease expressed on the surface of activated fibroblasts, promoting tumour invasion through extracellular matrix (ECM) degradation. The signalling mechanism behind the upregulation of FAP-α is not yet completely revealed.. Expression of FAP-α was analysed after UVR exposure in in vitro co-culture systems, gene expression arrays and artificial skin constructs. Cell migration and invasion was studied in relation to cathepsin activity and secretion of transforming growth factor (TGF)-β1.. Fibroblast activation protein-α expression was induced by UVR in melanocytes of human skin. The FAP-α expression was regulated by UVR-induced release of TGF-β1 and cathepsin inhibitors prevented such secretion. In melanoma cell culture models and in a xenograft tumour model of zebrafish embryos, FAP-α mediated ECM degradation and facilitated tumour cell dissemination.. Our results provide evidence for a sequential reaction axis from UVR via cathepsins, TGF-β1 and FAP-α expression, promoting cancer cell dissemination and melanoma metastatic spread. Topics: Animals; Cathepsins; Cell Line, Tumor; Cell Movement; Cellular Senescence; Coculture Techniques; Culture Media, Conditioned; Down-Regulation; Endopeptidases; Fibroblasts; Gelatinases; Gene Expression; Gene Silencing; Humans; Keratinocytes; Melanocytes; Melanoma; Membrane Proteins; Neoplasm Transplantation; Nevus; Primary Cell Culture; Serine Endopeptidases; Signal Transduction; Skin; Skin Neoplasms; Skin, Artificial; Transcriptome; Transforming Growth Factor beta; Ultraviolet Rays; Up-Regulation; Zebrafish | 2017 |
C-myc overexpression drives melanoma metastasis by promoting vasculogenic mimicry via c-myc/snail/Bax signaling.
c-Myc is a well-characterized proto-oncogene that induces cellular transformation and modulates programmed cell death. While recent studies have demonstrated high expression of c-Myc protein in advanced and metastatic melanoma, the clinical and biological implications remain to be fully elucidated. In this study, we investigated the effect of c-Myc overexpression in melanoma tumorigenesis. Clinicopathological analysis demonstrated that c-Myc expression positively correlated with the formation of vasculogenic mimicry (VM) and linearly patterned programmed cell necrosis (LPPCN). Clinically, high c-Myc expression was significantly associated with distant metastasis and poor prognosis, while biologically, c-Myc overexpression led to significant increases in cell motility, invasiveness and metastasis. Moreover, c-Myc induced the formation of VM and promoted the expression of epithelial-mesenchymal transition (EMT)-associated protein Snail both in vivo and in vitro. High expression of c-Myc increased Bax expression in hypoxic conditions and induced cell apoptosis. Taken together, we conclude that c-Myc overexpression promotes the formation of VM by EMT and LPPCN in melanoma. Our improved understanding of the clinical and biological effects of c-Myc overexpression in melanoma highlights the incomplete understanding of this oncogene, and indicates that c-Myc is a potential therapeutic target of this disease.. High c-Myc expression is associated with tumor metastasis and poor prognosis in human melanoma. c-Myc upregulates Snail expression to promote EMT via the TGF-β/Snail/Ecadherin signal pathway. c-Myc leads to cell death by upregulating Bax expression causing a lower Bcl2/Bax ratio under severe hypoxic conditions. c-Myc promotes vasculogenic mimicry and linearly patterned programmed cell necrosis. Topics: Adult; Aged; Animals; bcl-2-Associated X Protein; Cadherins; Cell Line, Tumor; Cell Movement; Cell Proliferation; Disease Models, Animal; Epithelial-Mesenchymal Transition; Female; Gene Expression; Heterografts; Humans; Immunohistochemistry; Male; Melanoma; Mice; Middle Aged; Neoplasm Metastasis; Neoplasm Staging; Neovascularization, Pathologic; Prognosis; Proto-Oncogene Mas; Proto-Oncogene Proteins c-bcl-2; Proto-Oncogene Proteins c-myc; Signal Transduction; Snail Family Transcription Factors; Transforming Growth Factor beta | 2017 |
MicroRNA-125a promotes resistance to BRAF inhibitors through suppression of the intrinsic apoptotic pathway.
Melanoma patients with BRAF Topics: Apoptosis; bcl-2 Homologous Antagonist-Killer Protein; Cell Line, Tumor; Drug Resistance, Neoplasm; Gene Expression Regulation, Neoplastic; Humans; Melanoma; MicroRNAs; Protein Kinase Inhibitors; Proto-Oncogene Proteins B-raf; Signal Transduction; Transforming Growth Factor beta | 2017 |
Melanoma Cells Block PEDF Production in Fibroblasts to Induce the Tumor-Promoting Phenotype of Cancer-Associated Fibroblasts.
Loss of pigment epithelium-derived factor (PEDF, SERPINF1) in cancer cells is associated with poor prognosis and metastasis, but the contribution of stromal PEDF to cancer evolution is poorly understood. Therefore, we investigated the role of fibroblast-derived PEDF in melanoma progression. We demonstrate that normal dermal fibroblasts expressing high PEDF levels attenuated melanoma growth and angiogenesis in vivo, whereas PEDF-depleted fibroblasts exerted tumor-promoting effects. Accordingly, mice with global PEDF knockout were more susceptible to melanoma metastasis. We also demonstrate that normal fibroblasts in close contact with PEDF-null melanoma cells lost PEDF expression and tumor-suppressive properties. Further mechanistic investigations underlying the crosstalk between tumor and stromal cells revealed that melanoma cells produced PDGF-BB and TGFβ, which blocked PEDF production in fibroblasts. Notably, cancer-associated fibroblasts (CAF) isolated from patient-derived tumors expressed markedly low levels of PEDF. Treatment of patient CAF and TGFβ-treated normal fibroblasts with exogenous PEDF decreased the expression of CAF markers and restored PEDF expression. Finally, expression profiling of PEDF-depleted fibroblasts revealed induction of IL8, SERPINB2, hyaluronan synthase-2, and other genes associated with tumor promotion and metastasis. Collectively, our results demonstrate that PEDF maintains tumor-suppressive functions in fibroblasts to prevent CAF conversion and illustrate the mechanisms by which melanoma cells silence stromal PEDF to promote malignancy. Cancer Res; 76(8); 2265-76. ©2016 AACR. Topics: Animals; Cell Line, Tumor; Cell Proliferation; Eye Proteins; Fibroblasts; Humans; Melanoma; Mice; Mice, Inbred BALB C; Mice, Nude; Nerve Growth Factors; Serpins; Transforming Growth Factor beta | 2016 |
Inactivation of TGFβ receptors in stem cells drives cutaneous squamous cell carcinoma.
Melanoma patients treated with oncogenic BRAF inhibitors can develop cutaneous squamous cell carcinoma (cSCC) within weeks of treatment, driven by paradoxical RAS/RAF/MAPK pathway activation. Here we identify frequent TGFBR1 and TGFBR2 mutations in human vemurafenib-induced skin lesions and in sporadic cSCC. Functional analysis reveals these mutations ablate canonical TGFβ Smad signalling, which is localized to bulge stem cells in both normal human and murine skin. MAPK pathway hyperactivation (through Braf(V600E) or Kras(G12D) knockin) and TGFβ signalling ablation (through Tgfbr1 deletion) in LGR5(+ve) stem cells enables rapid cSCC development in the mouse. Mutation of Tp53 (which is commonly mutated in sporadic cSCC) coupled with Tgfbr1 deletion in LGR5(+ve) cells also results in cSCC development. These findings indicate that LGR5(+ve) stem cells may act as cells of origin for cSCC, and that RAS/RAF/MAPK pathway hyperactivation or Tp53 mutation, coupled with loss of TGFβ signalling, are driving events of skin tumorigenesis. Topics: Animals; Antineoplastic Agents; Biopsy; Carcinogenesis; Carcinoma, Squamous Cell; Cell Line, Tumor; DNA Mutational Analysis; Exome Sequencing; Female; Humans; Indoles; Male; Melanoma; Mice; Mice, Inbred Strains; Mutation; Neoplasms, Experimental; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins B-raf; Proto-Oncogene Proteins p21(ras); Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Skin Neoplasms; Stem Cells; Sulfonamides; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Vemurafenib | 2016 |
USF1 prompt melanoma through upregulating TGF-β signaling pathway.
The activation of TGF-β signaling contributes to abnormal EMT process and upstream stimulatory family1 (USF1) was recently found to activate the expression of TGF-β. However, the specific role of USF1 in melanoma has never been explored.. The expression of USF1 was analyzed using real-time PCR and Western blot. The changes of cell morphology were observed under a microscope. Cell migration was determined using in vitro scratch test. A specific siRNA was applied to knockdown of USF1.. The mRNA and protein levels of USF1 were significantly enhanced in melanoma cell lines, 1205-Lu, DO4, WM3211, and WM278, compared with normal human melanocytes PIG1. Overexpression of USF1 induced demonstrated an elongated and spindle-shaped morphology in the 1205-Lu cells. Meanwhile, USF1 induced the expression of α-SMA, Vimentin and Fibronectin, while the epithelial marker, E-cadherin (E-cad), was significantly decreased. Furthermore, in vitro scratch test demonstrated that overexpression of USF1 markedly enhanced 1205-Lu cell migration capacity at 24 h and 48 h. More importantly, knockdown of USF1 could partially reverse TGF-β1-treatment-induced changes of EMT markers as well as cell morphological changes.. We first demonstrate that overexpression of USF1 prompts the EMT process through the accumulation of TGF-β1 production in melanoma cells. Topics: Antigens, CD; Cadherins; Cell Movement; Fibronectins; Gene Expression Regulation; Humans; Melanoma; Skin Neoplasms; Transforming Growth Factor beta; Transforming Growth Factor beta1; Upstream Stimulatory Factors | 2016 |
Varying effects of EGF, HGF and TGFβ on formation of invadopodia and invasiveness of melanoma cell lines of different origin.
The understanding of melanoma malignancy mechanisms is essential for patient survival, because melanoma is responsible for ca. 75% of deaths related to skin cancers. Enhanced formation of invadopodia and extracellular matrix (ECM) degradation are two important drivers of cell invasion, and actin dynamics facilitate protrusive activity by providing a driving force to push through the ECM. We focused on the influence of epidermal growth factor (EGF), hepatocyte growth factor (HGF) and transforming growth factor β (TGFβ) on melanoma cell invasiveness, since they are observed in the melanoma microenvironment. All three factors stimulated invasion of A375 and WM1341D cells derived from primary tumor sites. In contrast, only EGF and HGF stimulated invasion of WM9 and Hs294T cells isolated from lymph node metastases. Enhanced formation of invadopodia and ECM degradation underlie the increased amount of invasive cells after stimulation with the tested agents. Generally, a rise in invasive potential was accompanied by a decrease in actin polymerization state (F:G ratio). The F:G ratio remained unchanged or was even increased in metastatic cell lines treated with TGFβ. Our findings indicate that the effects of stimulation with EGF, HGF and TGFβ on melanoma cell invasiveness could depend on melanoma cell progression stage. Topics: Cell Line, Tumor; Cell Membrane Structures; Epidermal Growth Factor; Hepatocyte Growth Factor; Humans; Melanoma; Neoplasm Invasiveness; Transforming Growth Factor beta; Tumor Microenvironment | 2016 |
Divergence(s) in nodal signaling between aggressive melanoma and embryonic stem cells.
The significant role of the embryonic morphogen Nodal in maintaining the pluripotency of embryonic stem cells is well documented. Interestingly, the recent discovery of Nodal's re-expression in several aggressive and metastatic cancers has highlighted its critical role in self renewal and maintenance of the stem cell-like characteristics of tumor cells, such as melanoma. However, the key TGFβ/Nodal signaling component(s) governing Nodal's effects in metastatic melanoma remain mostly unknown. By employing receptor profiling at the mRNA and protein level(s), we made the novel discovery that embryonic stem cells and metastatic melanoma cells share a similar repertoire of Type I serine/threonine kinase receptors, but diverge in their Type II receptor expression. Ligand:receptor crosslinking and native gel binding assays indicate that metastatic melanoma cells employ the heterodimeric TGFβ receptor I/TGFβ receptor II (TGFβRI/TGFβRII) for signal transduction, whereas embryonic stem cells use the Activin receptors I and II (ACTRI/ACTRII). This unexpected receptor usage by tumor cells was tested by: neutralizing antibody to block its function; and transfecting the dominant negative receptor to compete with the endogenous receptor for ligand binding. Furthermore, a direct biological role for TGFβRII was found to underlie vasculogenic mimicry (VM), an endothelial phenotype contributing to vascular perfusion and associated with the functional plasticity of aggressive melanoma. Collectively, these findings reveal the divergence in Nodal signaling between embryonic stem cells and metastatic melanoma that can impact new therapeutic strategies targeting the re-emergence of embryonic pathways. Topics: Activin Receptors, Type I; Activin Receptors, Type II; Activins; Blotting, Western; Cells, Cultured; Embryonic Stem Cells; Humans; Melanoma; Nodal Protein; Protein Serine-Threonine Kinases; Real-Time Polymerase Chain Reaction; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta | 2015 |
PI3K signalling is required for a TGFβ-induced epithelial-mesenchymal-like transition (EMT-like) in human melanoma cells.
Epithelial to mesenchymal transition (EMT) is a programme defined in epithelial cells and recognized as playing a critical role in cancer progression. Although melanoma is not a cancer of epithelial cells, hallmarks of EMT have been described to play a critical role in melanoma progression. Here, we demonstrate that long-term TGFβ exposure can induce a dedifferentiated EMT-like state resembling a previously described invasive phenotype (EMT-like). TGFβ-induced EMT-like is marked by the downregulation of melanocyte differentiation markers, such as MITF, and the upregulation of mesenchymal markers, such as N-cadherin, and an increase in melanoma cell migration and cell invasion. Pharmacological interference shows the dependency of TGFβ-induced EMT-like on the activation of the PDGF signalling pathway and the subsequent activation of PI3K in human melanoma cells. Together, the data provide novel insights into the transcriptional plasticity of melanoma cells that might contribute to tumor progression in patients and propose avenues to therapeutic interventions. Topics: Cell Differentiation; Cell Line, Tumor; Cell Movement; Disease Progression; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Neovascularization, Pathologic; Phenotype; Phosphatidylinositol 3-Kinases; Platelet-Derived Growth Factor; RNA, Small Interfering; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured | 2015 |
Inhibitor of differentiation-4 (Id4) stimulates pigmentation in melanoma leading to histiocyte infiltration.
TGF-β and the inhibitors of differentiation (Id) are linked. Smad7 and other TGF-β inhibitors can potently suppress melanomagenesis; however, little work examining Ids has been reported in melanoma, particularly for Id4. Here, we report that Id4, but not Id2 or Id3 expression, surprisingly, activated robust melanin production in xenografts of previously amelanotic (lacking pigment) 1205Lu/Smad7 (S7) cells. Fontana-Masson stain and de-novo expression of MART-1 and tyrosinase proteins confirmed melanin production. Additionally, pigment-laden CD163+ mouse histiocytes with areas of extensive necrosis were found throughout S7/Id4 tumors, but not in parental 1205Lu, S7/Id2 or S7Id3-derived tumors. Mechanistic investigation revealed increased nuclear M-microphthalmia-associated transcription factor (MITF) and MART-1 promoter activation following Id4 expression in 1205Lu and WM852 melanoma cells, suggesting broader implications for Id4 in melanin synthesis. In human tumors, melanin colocalized with Id4 expression establishing a correlation. Current chemotherapeutics for melanoma are only marginally effective. Immunotherapy provides the most promise, yet the role of innate immunity is poorly understood. Here, TGF-β suppression followed by Id4 expression results in extensive melanin synthesis and robust histiocyte recruitment following tumorigenesis, a novel role for Id4. Our results suggest that TGF-β suppression coupled with pigment overproduction triggers an innate immune response resulting in tumor necrosis. Topics: Animals; Antigens, CD; Antigens, Differentiation, Myelomonocytic; Cell Proliferation; Genetic Vectors; Histiocytes; Humans; Immunity, Innate; Inhibitor of Differentiation Proteins; Keratinocytes; MART-1 Antigen; Melanins; Melanoma; Mice; Mice, Nude; Neoplasm Transplantation; Pigmentation; Promoter Regions, Genetic; Receptors, Cell Surface; Retroviridae; Skin Neoplasms; Transforming Growth Factor beta | 2015 |
Role of IL-10 and TGF-β in melanoma.
IL-10 and TGF-β are immunosuppressive cytokines expressed in tumors including melanoma and, therefore, deemed major cause for failing antitumor immune responses. Re-evaluating their role, we compared their expression by quantitative RT-PCR in melanoma and skin of healthy individuals, tested their induction in dendritic cells and T cells co-cultured with tumor cells, and their effects on the immune cells. Both cytokines as well as their receptors were expressed in melanoma at significantly lower levels than in healthy skin. Consequently, the expressions of IL-10-responsive SOCS-3 and TGF-β-responsive Smad-7 were low in tumors but high in healthy skin. T cells co-cultured with tumor cells developed an anergic state without increased IL-10 or TGF-β expression. In vitro tumor-induced immature dendritic cells produced high IL-10 levels and less efficiently induced T-cell proliferation. Nonetheless, they could be induced to mature, and blocking IL-10 did not alter the capacity of the resulting mature dendritic cells to stimulate T cells. Mature dendritic cells co-cultured with tumor cells produced increased IL-10 but decreased TGF-β and more efficiently induced T-cell proliferation. The lack of correlation of IL-10 and TGF-β with immune deficits in situ and in vitro suggests re-evaluating their roles in cancer. Topics: Cell Proliferation; Coculture Techniques; Dendritic Cells; Gene Expression; Humans; Interleukin-10; Melanoma; Skin; Skin Neoplasms; Smad7 Protein; Suppressor of Cytokine Signaling 3 Protein; Suppressor of Cytokine Signaling Proteins; T-Lymphocytes; Transforming Growth Factor beta; Tumor Cells, Cultured | 2015 |
EWI-2 negatively regulates TGF-β signaling leading to altered melanoma growth and metastasis.
In normal melanocytes, TGF-β signaling has a cytostatic effect. However, in primary melanoma cells, TGF-β-induced cytostasis is diminished, thus allowing melanoma growth. Later, a second phase of TGF-β signaling supports melanoma EMT-like changes, invasion and metastasis. In parallel with these "present-absent-present" TGF-β signaling phases, cell surface protein EWI motif-containing protein 2 (EWI-2 or IgSF8) is "absent-present-absent" in melanocytes, primary melanoma, and metastatic melanoma, respectively, suggesting that EWI-2 may serve as a negative regulator of TGF-β signaling. Using melanoma cell lines and melanoma short-term cultures, we performed RNAi and overexpression experiments and found that EWI-2 negatively regulates TGF-β signaling and its downstream events including cytostasis (in vitro and in vivo), EMT-like changes, cell migration, CD271-dependent invasion, and lung metastasis (in vivo). When EWI-2 is present, it associates with cell surface tetraspanin proteins CD9 and CD81 - molecules not previously linked to TGF-β signaling. Indeed, when associated with EWI-2, CD9 and CD81 are sequestered and have no impact on TβR2-TβR1 association or TGF-β signaling. However, when EWI-2 is knocked down, CD9 and CD81 become available to provide critical support for TβR2-TβR1 association, thus markedly elevating TGF-β signaling. Consequently, all of those TGF-β-dependent functions specifically arising due to EWI-2 depletion are reversed by blocking or depleting cell surface tetraspanin proteins CD9 or CD81. These results provide new insights into regulation of TGF-β signaling in melanoma, uncover new roles for tetraspanins CD9 and CD81, and strongly suggest that EWI-2 could serve as a favorable prognosis indicator for melanoma patients. Topics: Animals; Antigens, CD; Benzamides; Cell Line, Tumor; Cell Movement; Cell Proliferation; Dioxoles; HEK293 Cells; Humans; Melanoma; Membrane Proteins; Mice; Mice, Inbred C57BL; Mice, SCID; Neoplasm Invasiveness; Neoplasm Transplantation; Nerve Tissue Proteins; Prognosis; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Nerve Growth Factor; Receptors, Transforming Growth Factor beta; RNA Interference; RNA, Small Interfering; Signal Transduction; Tetraspanin 24; Tetraspanin 28; Tetraspanin 29; Transforming Growth Factor beta; Transplantation, Heterologous | 2015 |
Friends or foes: IL-10 and TGF-β in melanoma.
Topics: Humans; Interleukin-10; Melanoma; Skin Neoplasms; Transforming Growth Factor beta | 2015 |
Inhibition of uPAR-TGFβ crosstalk blocks MSC-dependent EMT in melanoma cells.
The capacity of cancer cells to undergo epithelial-to-mesenchymal transition (EMT) is now considered a hallmark of tumor progression, and it is known that interactions between cancer cells and mesenchymal stem cells (MSCs) of tumor microenvironment may promote this program. Herein, we demonstrate that MSC-conditioned medium (MSC-CM) is a potent inducer of EMT in melanoma cells. The EMT profile acquired by MSC-CM-exposed melanoma cells is characterized by an enhanced level of mesenchymal markers, including TGFβ/TGFβ-receptors system upregulation, by increased invasiveness and uPAR expression, and in vivo tumor growth. Silencing TGFβ in MSC is found to abrogate ability of MSC to promote EMT characteristics in melanoma cells, together with uPAR expression, and this finding is strengthened using an antagonist peptide of TGFβRIII, the so-called P17. Finally, we demonstrate that the uPAR antisense oligonucleotide (uPAR aODN) may inhibit EMT of melanoma cells either stimulated by exogenous TGFβ or MSC-CM. Thus, uPAR upregulation in melanoma cells exposed to MSC-medium drives TGFβ-mediated EMT. On the whole, TGFβ/uPAR dangerous liaison in cancer cell/MSC interactions may disclose a new strategy to abrogate melanoma progression.. Mesenchymal stem cell (MSC)-conditioned medium induces EMT-like profile in melanoma. MSC-derived TGFβ promotes uPAR and TGFβ/TGFβ-receptor upregulation in melanoma. TGFβ gene silencing in MSCs downregulates uPAR expression and EMT in melanoma. uPAR downregulation prevents MSC-induced EMT-like profile in melanoma cells. Inhibition of the dangerous TGFβ/uPAR relationship might abrogate melanoma progression. Topics: Animals; Cell Line, Tumor; Culture Media, Conditioned; Epithelial-Mesenchymal Transition; Humans; Melanoma; Mesenchymal Stem Cells; Mice; Neoplasm Transplantation; Receptors, Transforming Growth Factor beta; Receptors, Urokinase Plasminogen Activator; Transforming Growth Factor beta; Transplantation, Heterologous; Tumor Microenvironment | 2015 |
Immunomodulatory pathways regulate expression of a spliced FKBP51 isoform in lymphocytes of melanoma patients.
FKBP51 (gene FKBP5) is an immunophilin capable of immunosuppression expressed in melanoma and lymphocytes. We found increased levels of a spliced FKBP5 variant in the PBMCs of 124 patients with melanoma. This variant encodes for an unknown isoform (FKBP51s). We hypothesized that FKBP51s resulted from tumour interaction with immune cells, through PDL-1/PD-1. To address this issue, we performed melanoma/PBMC cocultures. Furthermore, the immunohistochemistry of 76 melanoma specimens served to investigate whether FKBP51s stained tumour infiltrating lymphocytes. Our results showed that PBMCs expressed FKBP51s when cocultured with melanoma. Tumour PDL-1 knockdown or anti-PD-1 reduced FKBP51s expression in cocultured PBMCs. IHC showed a strong FKBP51s signal in tumour infiltrating lymphocytes, and lymphocytes of the invasion front of the tumour, along with melanoma PDL-1 expression. When overexpressed in melanoma, FKBP51s facilitated PDL-1 expression on the cell surface. In conclusion, our study shows that FKBP51s marks the PBMCs of patients with melanoma and is exploited by the tumour to immunomodulate through PDL-1. Topics: Adolescent; Adult; Aged; Aged, 80 and over; Alternative Splicing; B7-H1 Antigen; Female; Gene Expression Regulation, Neoplastic; Humans; Immunologic Factors; Lymphocytes; Lymphocytes, Tumor-Infiltrating; Male; Melanoma; Middle Aged; Protein Isoforms; Tacrolimus Binding Proteins; Transforming Growth Factor beta; Up-Regulation; Young Adult | 2015 |
The BRAF(V600E) inhibitor, PLX4032, increases type I collagen synthesis in melanoma cells.
Vertical growth phase (VGP) melanoma is frequently metastatic, a process mediated by changes in gene expression, which are directed by signal transduction pathways in the tumor cells. A prominent signaling pathway is the Ras-Raf-Mek-Erk MAPK pathway, which increases expression of genes that promote melanoma progression. Many melanomas harbor a mutation in this pathway, BRAF(V600E), which constitutively activates MAPK signaling and expression of downstream target genes that facilitate tumor progression. In BRAF(V600E) melanoma, the small molecule inhibitor, vemurafenib (PLX4032), has revolutionized therapy for melanoma by inducing rapid tumor regression. This compound down-regulates the expression of many genes. However, in this study, we document that blocking the Ras-Raf-Mek-Erk MAPK pathway, either with an ERK (PLX4032) or a MEK (U1026) signaling inhibitor, in BRAF(V600E) human and murine melanoma cell lines increases collagen synthesis in vitro and collagen deposition in vivo. Since TGFß signaling is a major mediator of collagen synthesis, we examined whether blocking TGFß signaling with a small molecule inhibitor would block this increase in collagen. However, there was minimal reduction in collagen synthesis in response to blocking TGFß signaling, suggesting additional mechanism(s), which may include activation of the p38 MAPK pathway. Presently, it is unclear whether this increased collagen synthesis and deposition in melanomas represent a therapeutic benefit or an unwanted "off target" effect of inhibiting the Ras-Raf-Erk-Mek pathway. Topics: Animals; Antineoplastic Agents; Cell Line, Tumor; Collagen Type I; Extracellular Signal-Regulated MAP Kinases; Gene Expression Regulation, Neoplastic; Humans; Indoles; MAP Kinase Signaling System; Melanoma; Mice; Mitogen-Activated Protein Kinase Kinases; Mutation; Proto-Oncogene Proteins B-raf; raf Kinases; ras Proteins; Skin Neoplasms; Sulfonamides; Transforming Growth Factor beta; Vemurafenib; Xenograft Model Antitumor Assays | 2015 |
Lin28B promotes melanoma growth by mediating a microRNA regulatory circuit.
It has been increasingly recognized that microRNAs (miRNAs) are often dysregulated in various human malignancies and can function as oncogenes or tumor-suppressors. However, the potential roles of miRNAs and components of the miRNA biogenesis pathway remain poorly defined in melanoma. Here, we systematically profiled miRNA expression in human melanocytes and melanoma cells, and identified a prominent function of miR-125a-5p in suppressing melanoma growth. Mechanistically, we discovered that Lin28B, a well-characterized inhibitor of let-7 miRNA biogenesis, was a direct target of miR-125a-5p in melanoma. We showed that the Lin28B was aberrantly expressed in a large proportion of melanoma patients and was functionally required for melanoma progression. We further demonstrated the involvement of let-7-dependent mechanism downstream of Lin28B, resulting in the activation of transforming growth factor-β signaling cascade. Collectively, our data implicate Lin28B as a novel oncogene in melanomagenesis by mediating a miRNA regulatory circuit. Topics: Animals; Cell Line, Tumor; Down-Regulation; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; HEK293 Cells; Humans; Melanocytes; Melanoma; Mice; Mice, Inbred BALB C; Mice, Nude; MicroRNAs; RNA-Binding Proteins; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta; Tumor Suppressor Proteins | 2015 |
Molecular profiling of CD8 T cells in autochthonous melanoma identifies Maf as driver of exhaustion.
T cells infiltrating neoplasms express surface molecules typical of chronically virus-stimulated T cells, often termed "exhausted" T cells. We compared the transcriptome of "exhausted" CD8 T cells infiltrating autochthonous melanomas to those of naïve and acutely stimulated CD8 T cells. Despite strong similarities between transcriptional signatures of tumor- and virus-induced exhausted CD8 T cells, notable differences appeared. Among transcriptional regulators, Nr4a2 and Maf were highly overexpressed in tumor-exhausted T cells and significantly upregulated in CD8 T cells from human melanoma metastases. Transduction of murine tumor-specific CD8 T cells to express Maf partially reproduced the transcriptional program associated with tumor-induced exhaustion. Upon adoptive transfer, the transduced cells showed normal homeostasis but failed to accumulate in tumor-bearing hosts and developed defective anti-tumor effector responses. We further identified TGFβ and IL-6 as main inducers of Maf expression in CD8 T cells and showed that Maf-deleted tumor-specific CD8 T cells were much more potent to restrain tumor growth in vivo. Therefore, the melanoma microenvironment contributes to skewing of CD8 T cell differentiation programs, in part by TGFβ/IL-6-mediated induction of Maf. Topics: Animals; CD8-Positive T-Lymphocytes; Cell Differentiation; DNA Primers; Flow Cytometry; Gene Expression Profiling; Homeodomain Proteins; Interleukin-6; Luciferases; Melanoma; Mice; Mice, Transgenic; Proto-Oncogene Proteins c-maf; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; Transforming Growth Factor beta; Tumor Microenvironment | 2015 |
CDKN2B Loss Promotes Progression from Benign Melanocytic Nevus to Melanoma.
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 |
BRAF Inhibition Generates a Host-Tumor Niche that Mediates Therapeutic Escape.
The current study defines a fibroblast-derived niche that facilitates the therapeutic escape of melanoma cells from BRAF inhibition. Vemurafenib treatment led to the release of transforming growth factor-β (TGF-β) from the melanoma cells that increased the differentiation state of the fibroblasts, an affect associated with fibronectin deposition, increase in α-smooth muscle actin expression, and the release of neuregulin (NRG). At the same time, vemurafenib directly activated the fibroblasts through paradoxical stimulation of the mitogen-activated protein kinase pathway, causing them to secrete hepatocyte growth factor (HGF). Treatment with the BRAF/MEK inhibitor combination reversed the release of HGF. Adhesion of melanoma cells to fibronectin was critical in amplifying the fibroblast-derived NRG- and HGF-mediated PI3K/AKT (phosphatidylinositol 3'-kinase/AKT) survival signaling in the melanoma cells following BRAF inhibition. In coculture studies, combination treatment with inhibitors of BRAF/MET/HER kinase was ineffective at reversing the fibroblast-mediated therapeutic escape from BRAF inhibition. Instead, it was noted that combined BRAF/PI3K inhibition overcame fibroblast-mediated drug resistance in vitro and was associated with enhanced antitumor effects in an in vivo xenograft model. Thus, we show that melanoma cells and fibroblasts remodel their microenvironment in response to BRAF inhibition and that these adaptations allow tumor cells to evade therapy through increased PI3K/AKT survival signaling. Topics: Cell Line, Tumor; Fibronectins; Humans; Indoles; Melanoma; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Protein Kinase Inhibitors; Proto-Oncogene Proteins B-raf; Proto-Oncogene Proteins c-akt; Sulfonamides; Transforming Growth Factor beta; Vemurafenib | 2015 |
Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses.
Activated T cells engage aerobic glycolysis and anabolic metabolism for growth, proliferation, and effector functions. We propose that a glucose-poor tumor microenvironment limits aerobic glycolysis in tumor-infiltrating T cells, which suppresses tumoricidal effector functions. We discovered a new role for the glycolytic metabolite phosphoenolpyruvate (PEP) in sustaining T cell receptor-mediated Ca(2+)-NFAT signaling and effector functions by repressing sarco/ER Ca(2+)-ATPase (SERCA) activity. Tumor-specific CD4 and CD8 T cells could be metabolically reprogrammed by increasing PEP production through overexpression of phosphoenolpyruvate carboxykinase 1 (PCK1), which bolstered effector functions. Moreover, PCK1-overexpressing T cells restricted tumor growth and prolonged the survival of melanoma-bearing mice. This study uncovers new metabolic checkpoints for T cell activity and demonstrates that metabolic reprogramming of tumor-reactive T cells can enhance anti-tumor T cell responses, illuminating new forms of immunotherapy. Topics: Animals; Calcium; CD4-Positive T-Lymphocytes; Endoplasmic Reticulum; Glycolysis; Hexokinase; Immunotherapy; Lymphocytes, Tumor-Infiltrating; Melanoma; Mice; Monitoring, Immunologic; NFATC Transcription Factors; Phosphoenolpyruvate; Receptors, Antigen, T-Cell; Sarcoplasmic Reticulum Calcium-Transporting ATPases; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment | 2015 |
Dual Roles of RNF2 in Melanoma Progression.
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 |
Extracellular acidity strengthens mesenchymal stem cells to promote melanoma progression.
Mesenchymal stem cells (MSC) participate to tumor stroma development and several evidence suggests that they play a role in facilitating cancer progression. Because melanoma often shows extracellular pH low enough to influence host cell as tumor cell behavior, the aim of this study is to elucidate whether acidity affects cross talk between MSC and melanoma cells to disclose new liaisons promoting melanoma progression, and to offer new therapeutic opportunities. We found that MSC grown in a low pH medium (LpH-MSC) stimulate melanoma xenografts more than MSC grown in a standard pH medium. LpH-MSC express a higher level of TGFβ that is instrumental of epithelial-to-mesenchymal transition (EMT)-like phenotype induction in melanoma cells. LpH-MSC profile also shows a switching to an oxidative phosphorylation metabolism that was accompanied by a forced glycolytic pathway of melanoma cells grown in LpH-MSC-conditioned medium. Metformin, an inhibitor of mitochondrial respiratory chain was able to reconvert oxidative metabolism and abrogate TGFβ expression in LpH-MSC. In addition, esomeprazole, a proton pump inhibitor activated in acidosis, blocked TGFβ expression in LpH-MSC through the downregulation of IkB. Both agents, metformin and esomeprazole, inhibited EMT profile in melanoma cells grown in LpH-MSC medium, and reduced glycolytic markers. Thus, acidosis of tumor microenvironment potentiates the pro-tumoral activity of MSC and orchestrates for a new potential symbiosis, which could be target to limit melanoma progression. Topics: Cell Line, Tumor; Cell Movement; Cell Proliferation; Disease Progression; Epithelial-Mesenchymal Transition; Esomeprazole; Humans; Hydrogen-Ion Concentration; Melanoma; Mesenchymal Stem Cells; Transforming Growth Factor beta | 2015 |
TGF-β-Induced Transcription Sustains Amoeboid Melanoma Migration and Dissemination.
Cell migration underlies metastatic dissemination of cancer cells, and fast "amoeboid" migration in the invasive fronts of tumors is controlled by high levels of actomyosin contractility. How amoeboid migration is regulated by extracellular signals and sustained over time by transcriptional changes is not fully understood. Transforming growth factor β (TGF-β) is well known to promote epithelial-to-mesenchymal transition (EMT) and contribute to metastasis, but melanocytes are neural crest derivatives that have undergone EMT during embryonic development. Surprisingly, we find that in melanoma, TGF-β promotes amoeboid features such as cell rounding, membrane blebbing, high levels of contractility, and increased invasion. Using genome-wide transcriptomics, we find that amoeboid melanoma cells are enriched in a TGF-β-driven signature. We observe that downstream of TGF-β, SMAD2 and its adaptor CITED1 control amoeboid behavior by regulating the expression of key genes that activate contractile forces. Moreover, CITED1 is highly upregulated during melanoma progression, and its high expression is associated with poor prognosis. CITED1 is coupled to a contractile-rounded, amoeboid phenotype in a panel of 16 melanoma cell lines, in mouse melanoma xenografts, and in 47 human melanoma patients. Its expression is also enriched in the invasive fronts of lesions. Functionally, we show how the TGF-β-SMAD2-CITED1 axis promotes different steps associated with progression: melanoma detachment from keratinocytes, 2D and 3D migration, attachment to endothelial cells, and in vivo lung metastatic initial colonization and outgrowth. We propose a novel mechanism by which TGF-β-induced transcription sustains actomyosin force in melanoma cells and thereby promotes melanoma progression independently of EMT. Topics: Actomyosin; Animals; Apoptosis Regulatory Proteins; Cell Line, Tumor; Cell Movement; Cell Shape; Epithelial-Mesenchymal Transition; Humans; Keratinocytes; Melanocytes; Melanoma; Mice; Mice, Nude; Neoplasm Metastasis; Nuclear Proteins; Smad2 Protein; Trans-Activators; Transcription Factors; Transcriptional Activation; Transforming Growth Factor beta; Up-Regulation | 2015 |
Id2, Id3 and Id4 overcome a Smad7-mediated block in tumorigenesis, generating TGF-β-independent melanoma.
The role for the inhibitors of differentiation (Ids) proteins in melanomagenesis has been poorly explored. In other cell types, Ids have been shown to contribute to cell proliferation, migration and angiogenesis and, along with a number of other genes, are direct downstream targets of the transforming growth factor (TGF)-β pathway. Expression of Smad7, which suppress TGF-β signaling, or synthetic TGF-β inhibitors, was shown to potently suppress melanomagenesis. We found that endogenous Id2, Id3 and Id4 expression was elevated in 1205Lu versus 1205Lu cells constitutively expressing Smad7, indicating Ids may play a role in melanomagenesis. Therefore, the effects of Tet-inducible expression of Id2, Id3 or Id4 along with Smad7 in TGF-β-dependent 1205Lu human melanoma cells were explored in vitro and in vivo. 1205Lu cells formed subcutaneous tumors in athymic mice, whereas cells expressing Smad7 failed to form tumors. However, 1205Lu cells expressing Smad7 along with doxycycline-induced Id2, Id3 or Id4 were able to overcome the potent tumorigenic block mediated by S7, to varying degrees. Conversely, Id small interfering RNA knockdown suppressed anchorage-independent growth of melanoma. Histology of tumors from 1205Lu cells expressing Smad7 + Id4 revealed an average of 31% necrosis, compared with 5.2% in tumors from 1205Lu with vector only. Downstream, Ids suppressed cyclin-dependent kinase inhibitors, and re-upregulated invasion and metastasis-related genes matrix metalloproteinase 2 (MMP2), MMP9, CXCR4 and osteopontin, shown previously to be downregulated in response to Smad7. This study shows that Id2, Id3 and Id4 are each able to overcome TGF-β dependence, and establish a role for Ids as key mediators of TGF-β melanomagenesis. Topics: Base Sequence; Cell Line, Tumor; Cell Proliferation; DNA Primers; Humans; Inhibitor of Differentiation Protein 2; Inhibitor of Differentiation Proteins; Melanoma; Neoplasm Metastasis; Neoplasm Proteins; Reverse Transcriptase Polymerase Chain Reaction; Smad7 Protein; Transforming Growth Factor beta; Up-Regulation | 2014 |
TGF-β-induced (TGFBI) protein in melanoma: a signature of high metastatic potential.
Tumor-produced extracellular matrix (ECM) proteins can be key elements in tumor growth and metastasis. Transforming growth factor beta-inducible (TGFBI) protein is a secreted ECM component that can have dual function in cancer, acting as tumor suppressor or promoter. Although TGFBI is expressed in human melanoma cells, the exact role it might have in melanoma metastasis remains elusive. Assessing the expression and secretion of TGFBI, we show that human metastatic melanomas express and secrete significantly higher amounts of TGFBI, compared with nevus lesions and primary melanoma tumors. Intravenous injection of highly metastatic human melanoma cells expressing shRNA that targets TGFBI assigns a critical role for TGFBI in the formation of melanoma distal metastases in nude mice. In vivo assays demonstrate that TGFBI silencing does not interfere with melanoma cells' dissemination to distal sites but rather with their proliferation and outgrowth within new microenvironment. In line, TGFBI silencing increases melanoma cells motility/invasion/extravasation in vitro but interferes with their progression through the cell cycle, drastically reducing their proliferation. Furthermore, we show that TGFBI is a regulator of cyclins and cyclin-dependent kinases in melanoma. Collectively, our data describe a mechanism of melanoma metastatic outgrowth via promotion of growth/survival by the ECM protein TGFBI. Topics: Animals; Apoptosis; Biopsy; Cell Adhesion; Cell Cycle; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Survival; Extracellular Matrix Proteins; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Gene Silencing; Humans; Lung Neoplasms; Melanoma; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasm Transplantation; RNA, Small Interfering; Skin Neoplasms; Transforming Growth Factor beta | 2014 |
Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma.
Treatment of BRAF(V600E) mutant melanoma by small molecule drugs that target the BRAF or MEK kinases can be effective, but resistance develops invariably. In contrast, colon cancers that harbour the same BRAF(V600E) mutation are intrinsically resistant to BRAF inhibitors, due to feedback activation of the epidermal growth factor receptor (EGFR). Here we show that 6 out of 16 melanoma tumours analysed acquired EGFR expression after the development of resistance to BRAF or MEK inhibitors. Using a chromatin-regulator-focused short hairpin RNA (shRNA) library, we find that suppression of sex determining region Y-box 10 (SOX10) in melanoma causes activation of TGF-β signalling, thus leading to upregulation of EGFR and platelet-derived growth factor receptor-β (PDGFRB), which confer resistance to BRAF and MEK inhibitors. Expression of EGFR in melanoma or treatment with TGF-β results in a slow-growth phenotype with cells displaying hallmarks of oncogene-induced senescence. However, EGFR expression or exposure to TGF-β becomes beneficial for proliferation in the presence of BRAF or MEK inhibitors. In a heterogeneous population of melanoma cells having varying levels of SOX10 suppression, cells with low SOX10 and consequently high EGFR expression are rapidly enriched in the presence of drug, but this is reversed when the drug treatment is discontinued. We find evidence for SOX10 loss and/or activation of TGF-β signalling in 4 of the 6 EGFR-positive drug-resistant melanoma patient samples. Our findings provide a rationale for why some BRAF or MEK inhibitor-resistant melanoma patients may regain sensitivity to these drugs after a 'drug holiday' and identify patients with EGFR-positive melanoma as a group that may benefit from re-treatment after a drug holiday. Topics: Animals; Antineoplastic Agents; Cell Proliferation; Cellular Senescence; Drug Resistance, Neoplasm; ErbB Receptors; Female; Flow Cytometry; Gene Expression Regulation, Neoplastic; Gene Library; Humans; Indoles; Melanoma; Mice; Mitogen-Activated Protein Kinase Kinases; Protein Kinase Inhibitors; Proto-Oncogene Proteins B-raf; Receptor Protein-Tyrosine Kinases; Receptor, Platelet-Derived Growth Factor beta; RNA, Small Interfering; Signal Transduction; SOXE Transcription Factors; Sulfonamides; Transforming Growth Factor beta; Vemurafenib | 2014 |
Cells to surgery quiz: june 2014.
Topics: Antineoplastic Agents; Brain Neoplasms; Diagnosis, Differential; Extracellular Matrix Proteins; G1 Phase; Humans; Lung Neoplasms; Male; Melanoma; Middle Aged; Neoplasm Metastasis; Phospholipases A2; S Phase; Skin Neoplasms; Transforming Growth Factor beta | 2014 |
LIF mediates proinvasive activation of stromal fibroblasts in cancer.
Signaling crosstalk between tumor cells and fibroblasts confers proinvasive properties to the tumor microenvironment. Here, we identify leukemia inhibitory factor (LIF) as a tumor promoter that mediates proinvasive activation of stromal fibroblasts independent of alpha-smooth muscle actin (α-SMA) expression. We demonstrate that a pulse of transforming growth factor β (TGF-β) establishes stable proinvasive fibroblast activation by inducing LIF production in both fibroblasts and tumor cells. In fibroblasts, LIF mediates TGF-β-dependent actomyosin contractility and extracellular matrix remodeling, which results in collective carcinoma cell invasion in vitro and in vivo. Accordingly, carcinomas from multiple origins and melanomas display strong LIF upregulation, which correlates with dense collagen fiber organization, cancer cell collective invasion, and poor clinical outcome. Blockade of JAK activity by Ruxolitinib (JAK inhibitor) counteracts fibroblast-dependent carcinoma cell invasion in vitro and in vivo. These findings establish LIF as a proinvasive fibroblast producer independent of α-SMA and may open novel therapeutic perspectives for patients with aggressive primary tumors. Topics: Actins; Actomyosin; Animals; Carcinogenesis; Carcinoma; Cell Line, Tumor; Cell Movement; Extracellular Matrix; Female; Fibroblasts; Humans; Janus Kinases; Leukemia Inhibitory Factor; Melanoma; Mice; Mice, Inbred BALB C; Neoplasm Invasiveness; Nitriles; Pyrazoles; Pyrimidines; Signal Transduction; Transforming Growth Factor beta; Tumor Microenvironment; Up-Regulation | 2014 |
Inhibition of tumor-derived prostaglandin-e2 blocks the induction of myeloid-derived suppressor cells and recovers natural killer cell activity.
Increased frequencies of myeloid-derived suppressor cells (MDSC) correlate with poor prognosis in patients with cancers. Tumor-derived prostaglandin-E2 (PGE2) plays an important role in inducing MDSCs. However, the detailed mechanisms of this induction remain unknown. To develop targeted therapies for MDSCs, we sought to investigate the molecular basis of PGE2-regulated accumulation of MDSCs and their functional consequence on natural killer (NK) cell activity.. The effects of PGE2 in inducing phenotypic, signaling, and functional alternations on monocytes were analyzed in vitro. Suppression of NK-cell activity by PGE2-treated monocytes was compared with that of freshly isolated CD14(+)HLA-DR(low/-) monocytic MDSCs (moMDSC) from patients with melanoma. In addition, to explore the in vivo relevance of targeting PGE2 to reduce MDSC-mediated suppression of NK cells, we established a murine model, where tumor cells were disabled from cyclooxygenase-2 (COX-2) production.. Patient-derived moMDSCs inhibited NK-cell activity through the production of TGFβ. In vitro, binding of PGE2 to EP2 and EP4 receptors on monocytes activated the p38MAPK/ERK pathway and resulted in elevated secretion of TGFβ. Similar to moMDSCs, PGE2-treated monocytes potently suppressed NK-cell activity through production of TGFβ. Furthermore, silencing COX-2 in murine 4T1 tumor cells reduced the accumulation of CD11b(+)Gr1(+) MDSCs in the spleen, resulting in concomitant improved in vivo clearance of NK-cell sensitive YAC-1 cells.. Our results reveal an indispensable role of tumor-derived PGE2 in inducing MDSCs and suggest a favorable outcome of combining COX-2-targeted therapy and adoptive NK-cell transfer in patients with cancer. Topics: Adoptive Transfer; Animals; Antigen Presentation; Apoptosis; Blotting, Western; Cell Cycle; Cell Proliferation; Cyclooxygenase 2; Dinoprostone; Female; Flow Cytometry; HLA-DR Antigens; Humans; Killer Cells, Natural; Leukocytes, Mononuclear; Lipopolysaccharide Receptors; Lymphocyte Activation; Mammary Neoplasms, Animal; Melanoma; Mice; Mice, Nude; Monocytes; Myeloid Cells; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured; Xenograft Model Antitumor Assays | 2014 |
Melanoma-initiating cells exploit M2 macrophage TGFβ and arginase pathway for survival and proliferation.
M2 macrophages promote tumor growth and metastasis, but their interactions with specific tumor cell populations are poorly characterized. Using a mouse model of spontaneous melanoma, we showed that CD34- but not CD34+ tumor-initiating cells (TICs) depend on M2 macrophages for survival and proliferation. Tumor-associated macrophages (TAMs) and macrophage-conditioned media protected CD34- TICs from chemotherapy in vitro. In vivo, while inhibition of CD115 suppressed the macrophage-dependent CD34- TIC population, chemotherapy accelerated its development. The ability of TICs to respond to TAMs was acquired during melanoma progression and immediately preceded a surge in metastatic outgrowth. TAM-derived transforming growth factor-β (TGFβ) and polyamines produced via the Arginase pathway were critical for stimulation of TICs and synergized to promote their growth. Topics: Animals; Arginase; Cell Proliferation; Cell Survival; Disease Models, Animal; Female; Macrophages; Male; Melanoma; Mice; Mice, Mutant Strains; Neoplastic Stem Cells; Signal Transduction; Transforming Growth Factor beta | 2014 |
Nestin depletion induces melanoma matrix metalloproteinases and invasion.
Matrix metalloproteinases (MMPs) are key biological mediators of processes as diverse as wound healing, embryogenesis, and cancer progression. Although MMPs may be induced through multiple signaling pathways, the precise mechanisms for their regulation in cancer are incompletely understood. Because cytoskeletal changes are known to accompany MMP expression, we sought to examine the potential role of the poorly understood cytoskeletal protein, nestin, in modulating melanoma MMPs. Nestin knockdown (KD) upregulated the expression of specific MMPs and MMP-dependent invasion both through extracellular matrix barriers in vitro and in peritumoral connective tissue of xenografts in vivo. The development of three-dimensional melanospheres that in vitro partially recapitulate noninvasive tumorigenic melanoma growth was inhibited by nestin KD, although ECM invasion by aberrant melanospheres that did form was enhanced. Mechanistically, nestin KD-dependent melanoma invasion was associated with intracellular redistribution of phosphorylated focal adhesion kinase and increased melanoma cell responsiveness to transforming growth factor-beta, both implicated in pathways of melanoma invasion. The results suggest that the heretofore poorly understood intermediate filament, nestin, may serve as a novel mediator of MMPs critical to melanoma virulence. Topics: Animals; Cell Line, Tumor; Female; Focal Adhesion Protein-Tyrosine Kinases; Humans; Matrix Metalloproteinases; Melanoma; Mice; Neoplasm Invasiveness; Nestin; Transforming Growth Factor beta | 2014 |
GLI2 cooperates with ZEB1 for transcriptional repression of CDH1 expression in human melanoma cells.
In melanoma cells, high expression of the transcription factor GLI2 is associated with increased invasive potential and loss of E-cadherin expression, an event reminiscent of the epithelial-to-mesenchymal transition (EMT). Herein, we provide evidence that GLI2 represses E-cadherin gene (CDH1) expression in melanoma cells via distinct mechanisms, enhancing transcription of the EMT-activator ZEB1 and cooperative repression of CDH1 gene transcription via direct binding of both GLI2 and ZEB1 to two closely positioned Kruppel-like factor-binding sites within the CDH1 promoter. GLI2 silencing rescued CDH1 expression except in melanoma cell lines in which the CDH1 promoter was hypermethylated. Proximity ligation assays identified GLI2-ZEB1 complexes in melanoma cell nuclei, proportional to endogenous GLI2 and ZEB1 expression, and whose accumulation was enhanced by the classical EMT inducer TGF-β. These data identify GLI2 as a critical modulator of the cadherin switch in melanoma, a molecular process that is critical for metastatic spread of the disease. Topics: Antigens, CD; Base Sequence; Binding Sites; Cadherins; Cell Line, Tumor; Cell Nucleus; DNA Methylation; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Gene Silencing; Homeodomain Proteins; Humans; Kruppel-Like Transcription Factors; Melanoma; Models, Biological; Molecular Sequence Data; Nuclear Proteins; Promoter Regions, Genetic; Protein Binding; Repressor Proteins; Skin Neoplasms; Snail Family Transcription Factors; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Zinc Finger E-box-Binding Homeobox 1; Zinc Finger Protein Gli2 | 2013 |
Novel WWP2 ubiquitin ligase isoforms as potential prognostic markers and molecular targets in cancer.
The WWP2 E3 ubiquitin ligase has previously been shown to regulate TGFβ/Smad signalling activity linked to epithelial-mesenchymal transition (EMT). Whilst inhibitory I-Smad7 was found to be the preferred substrate for full-length WWP2-FL and a WWP2-C isoform, WWP2-FL also formed a stable complex with an N-terminal WWP2 isoform (WWP2-N) in the absence of TGFβ, and rapidly stimulated activating Smad2/3 turnover. Here, using stable knockdown experiments we show that specific depletion of individual WWP2 isoforms impacts differentially on Smad protein levels, and in WWP2-N knockdown cells we unexpectedly find spontaneous expression of the EMT marker vimentin. Re-introduction of WWP2-N into WWP2-N knockout cells also repressed TGFβ-induced vimentin expression. In support of the unique role for WWP2-N in regulating TGFβ/Smad functional activity, we then show that a novel V717M-WWP2 mutant in the MZ7-mel melanoma cell line forms a stable complex with the WWP2-N isoform and promotes EMT by stabilizing Smad3 protein levels. Finally, we report the first analysis of WWP2 expression in cancer cDNA panel arrays using WWP2 isoform-specific probes and identify unique patterns of WWP2 isoform abundance associated with early/advanced disease stages. WWP2-N is significantly downregulated in stage IIIC melanoma and up-regulated in stage II/III prostate cancer, and we also find isolated examples of WWP2-FL and WWP2-C overexpression in early-stage breast cancer. Together, these data suggest that individual WWP2 isoforms, and particularly WWP2-N, could play central roles in tumourigenesis linked to aberrant TGFβ-dependent signalling function, and also have potential as both prognostic markers and molecular therapeutic targets. Topics: Apoptosis; Biomarkers, Tumor; Blotting, Western; Breast Neoplasms; Case-Control Studies; Cell Proliferation; Epithelial-Mesenchymal Transition; Female; Gene Expression Profiling; Humans; Immunoprecipitation; Luciferases; Male; Melanoma; Neoplasm Staging; Oligonucleotide Array Sequence Analysis; Prognosis; Prostatic Neoplasms; Protein Isoforms; RNA, Small Interfering; Smad3 Protein; Transforming Growth Factor beta; Tumor Cells, Cultured; Ubiquitin-Protein Ligases | 2013 |
Chitinase 3-like 1 regulates cellular and tissue responses via IL-13 receptor α2.
Members of the 18 glycosyl hydrolase (GH 18) gene family have been conserved over species and time and are dysregulated in inflammatory, infectious, remodeling, and neoplastic disorders. This is particularly striking for the prototypic chitinase-like protein chitinase 3-like 1 (Chi3l1), which plays a critical role in antipathogen responses where it augments bacterial killing while stimulating disease tolerance by controlling cell death, inflammation, and remodeling. However, receptors that mediate the effects of GH 18 moieties have not been defined. Here, we demonstrate that Chi3l1 binds to interleukin-13 receptor α2 (IL-13Rα2) and that Chi3l1, IL-13Rα2, and IL-13 are in a multimeric complex. We also demonstrate that Chi3l1 activates macrophage mitogen-activated protein kinase, protein kinase B/AKT, and Wnt/β-catenin signaling and regulates oxidant injury, apoptosis, pyroptosis, inflammasome activation, antibacterial responses, melanoma metastasis, and TGF-β1 production via IL-13Rα2-dependent mechanisms. Thus, IL-13Rα2 is a GH 18 receptor that plays a critical role in Chi3l1 effector responses. Topics: Animals; Apoptosis; Chitinase-3-Like Protein 1; Glycoproteins; Humans; Inflammasomes; Interleukin-13; Interleukin-13 Receptor alpha2 Subunit; Lung Neoplasms; Macrophages; MAP Kinase Signaling System; Melanoma; Mice; Mice, Inbred C57BL; Oxidative Stress; Protein Binding; Transforming Growth Factor beta; Wnt Signaling Pathway | 2013 |
Triptolide downregulates Treg cells and the level of IL-10, TGF-β, and VEGF in melanoma-bearing mice.
Regulatory T cells play a key role in suppressing tumor immunity. Triptolide, a major active component isolated from the Chinese medicinal herb Tripterygium wilfordii, has been proven to possess multiple antitumor activities. Here, we investigated the effect of triptolide on regulatory T cells and on the level of IL-10, transforming growth factor-β, and vascular endothelial growth factor in tumor-bearing mice. Fifty male C57BL/6 mice were randomly grouped as follows: normal control group, model group with B16-F10 cells implanted, and three treatment groups with cyclophosphamide, triptolide-high dose, triptolide-low dose. The proportion of regulatory T cells in the spleen and axillary lymph nodes was evaluated by flow cytometric analysis. Production of cytokines IL-10, transforming growth factor-β, and vascular endothelial growth factor in serum was measured using enzyme-labeled immunosorbent assay kits. The mRNA levels of Foxp3, IL-10, and transforming growth factor-β in the spleen and vascular endothelial growth factor in tumor tissue were detected by real-time PCR. The results showed that triptolide significantly decreased the proportion of regulatory T cells and lowered the Foxp3 level in the spleen and axillary lymph nodes of tumor-bearing mice. Production of IL-10 and transforming growth factor-β in peripheral blood, and the mRNA level of IL-10 and transforming growth factor-β in the spleen were also decreased. Additionally, triptolide could remarkably inhibit production of vascular endothelial growth factor in tumor-bearing mice. In conclusion, our study demonstrated that triptolide might inhibit tumor growth by inhibiting regulatory T cells and some cytokines such as IL-10 and transforming growth factor-β, as well as production of vascular endothelial growth factor. Topics: Animals; Antineoplastic Agents, Phytogenic; Disease Models, Animal; Diterpenes; Down-Regulation; Drugs, Chinese Herbal; Epoxy Compounds; Forkhead Transcription Factors; Interleukin-10; Lymph Nodes; Male; Melanoma; Mice, Inbred C57BL; Phenanthrenes; Phytotherapy; RNA, Messenger; Spleen; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tripterygium; Vascular Endothelial Growth Factor A | 2013 |
TGF-beta inhibits human cutaneous melanoma cell migration and invasion through regulation of the plasminogen activator system.
Over the past decades, the incidence of cutaneous melanoma in developed countries has increased faster than any other cancer. Although most patients have localized disease at the time of diagnosis and are cured by surgical excision of the primary tumor, melanoma can be highly malignant and the survival dramatically decreases for advanced stage melanomas. It is thus necessary to understand the progression of this disease. Cell migration and invasion promote tumor metastasis, the major cause of melanoma cancer morbidity and death. In this study, we investigated the role of the TGFβ/Smad signaling pathway in melanoma tumor progression and found TGFβ to potently inhibit both cell migration and invasion in human melanoma cell lines, established from different patients. Furthermore, we elucidated the molecular mechanisms by which TGFβ exerts its effects and found the plasminogen activation system (PAS) to play a central role in the regulation of these effects. We found TGFβ to strongly up-regulate the Plasminogen Activator Inhibitor-1 (PAI-1) in melanoma cells, leading to reduced plasmin generation and activity and, in turn to inhibition of cell migration and invasion. Together, our results define TGFβ as a potent suppressor of tumor progression in cutaneous melanoma, inhibiting both cell migration and invasion. Topics: Adult; Aged; Cell Movement; Cell Survival; Female; Fibrinolysin; Humans; Male; Melanoma; Middle Aged; Plasminogen Activator Inhibitor 1; Plasminogen Activators; RNA Interference; RNA, Small Interfering; Signal Transduction; Skin Neoplasms; Smad Proteins; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation | 2013 |
The chick embryo as an experimental system for melanoma cell invasion.
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 |
Demethylation of the FOXP3 gene in human melanoma cells precludes the use of this epigenetic mark for quantification of Tregs in unseparated melanoma samples.
The human suppressive T cells that stably express transcription factor FOXP3, or regulatory T cells (Tregs), are thought to suppress antitumor immune responses. The most specific marker for human Tregs is the demethylation of CpG dinucleotides located in the first intron of FOXP3 (FOXP3i1). FOXP3i1 is completely methylated in other hematopoietic cells, including nonsuppressive T cells that transiently express FOXP3 after activation. Previously, we and others reported estimations of the frequency of Tregs in the blood of melanoma patients using a FOXP3i1 methylation-specific qPCR assay. Here, we attempted to quantify Tregs inside tumor samples using this assay. However, we found demethylated FOXP3i1 sequences in the melanoma cells themselves. This demethylation was not associated with substantial FOXP3 mRNA or protein expression, even though the demethylation extended to the promoter and terminal regions of the gene in some melanoma cells. Our results imply that analyzing Treg frequencies by quantification of demethylated FOXP3i1 will require that tumor-infiltrating T cells be separated from melanoma cells. Topics: Biomarkers, Tumor; Blotting, Western; Cell Line, Tumor; Colorectal Neoplasms; CpG Islands; DNA Methylation; Epigenesis, Genetic; Female; Forkhead Transcription Factors; Gene Expression Regulation, Neoplastic; Humans; Immunohistochemistry; Introns; Lung Neoplasms; Male; Melanoma; Phosphorylation; Promoter Regions, Genetic; Reverse Transcriptase Polymerase Chain Reaction; Smad2 Protein; T-Lymphocytes; T-Lymphocytes, Regulatory; Transforming Growth Factor beta | 2012 |
Transforming growth factor beta-induced (TGFBI) is an anti-adhesive protein regulating the invasive growth of melanoma cells.
Melanoma is a malignancy characterized by high invasive/metastatic potential, with no efficient therapy after metastasis. Understanding the molecular mechanisms underlying the invasive/metastatic tendency is therefore important. Our genome-wide gene expression analyses revealed that human melanoma cell lines WM793 and especially WM239 (vertical growth phase and metastatic cells, respectively) overexpress the extracellular matrix (ECM) protein transforming growth factor β induced (TGFBI). In adhesion assays, recombinant TGFBI was strongly anti-adhesive for both melanoma cells and skin fibroblasts. TGFBI further impaired the adhesion of melanoma cells to the adhesive ECM proteins fibronectin, collagen-I, and laminin, known to interact with it. Unexpectedly, WM239 cells migrated/invaded more effectively in three-dimensional collagen-I and Matrigel cultures after knockdown of TGFBI by shRNA expression. However, in the physiological subcutaneous microenvironment in nude mice, after TGFBI knockdown, these cells showed markedly impaired tumor growth and invasive capability; the initially formed small tumors later underwent myxoid degeneration and completely regressed. By contrast, the expanding control tumors showed intense TGFBI staining at the tumor edges, co-localizing with the fibrillar fibronectin/tenascin-C/periostin structures that characteristically surround melanoma cells at invasion fronts. Furthermore, TGFBI was found in similar fibrillar structures in clinical human melanoma metastases as well, co-localizing with fibronectin. These data imply an important role for TGFBI in the ECM deposition and invasive growth of melanoma cells, rendering TGFBI a potential target for therapeutic interventions. Topics: Actin Cytoskeleton; Animals; Cell Adhesion; Cell Movement; Extracellular Matrix; Extracellular Matrix Proteins; Female; Fibroblasts; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Genome-Wide Association Study; Humans; Integrin beta1; Melanoma; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Invasiveness; Neoplasm Proteins; Neoplasm Transplantation; Recombinant Proteins; Skin; Talin; Thymosin; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation | 2012 |
Antitumor efficacy of viable tumor vaccine modified by heterogenetic ESAT-6 antigen and cytokine IL-21 in melanomatous mouse.
The goal of this study was to investigate whether glycosylphosphatidylinositol (GPI)-anchored 6 kDa early secreted antigenic target (ESAT-6) and IL-21-producing B16F10/ESAT-6-GPI-IL-21 viable vaccine would induce antitumor efficacy. Mice were immunized with B16F10/ESAT-6-GPI-IL-21 vaccine and challenged by B16F10 cells 2 weeks later. Antitumor efficacy and mechanisms of the vaccine were analyzed. Vaccination with the viable B16F10/ESAT-6-GPI-IL-21 vaccine resulted in an increase of IFN-γ level and the CD8(+)CTL cytotoxicity, a decrease in TGF-β generation and increase in the expression of miR-200c that serves as melanoma suppressor by directly targeting zinc-finger E-box binding homeobox 1 to inhibit epithelial-mesenchymal transition and block tumor metastasis. The vaccine significantly inhibited the melanoma growth, reduced the lung melanoma nodules, and prolonged the mouse survival compared with the controls. These findings highlighted IL-21 as an immune adjuvant in an engineered viable tumor vaccine to reinforce heterogenetic antigen ESAT-6 immune tolerance break to induce powerful antitumor efficacy in mice. Topics: Animals; Antigens, Bacterial; Bacterial Proteins; Cancer Vaccines; CD8-Positive T-Lymphocytes; Cell Line, Tumor; Interferon-gamma; Interleukins; Melanoma; Mice; Transforming Growth Factor beta; Vaccination | 2012 |
Expression of microphthalmia-associated transcription factor (MITF), which is critical for melanoma progression, is inhibited by both transcription factor GLI2 and transforming growth factor-β.
The melanocyte-specific transcription factor M-MITF is involved in numerous aspects of melanoblast lineage biology including pigmentation, survival, and migration. It plays complex roles at all stages of melanoma progression and metastasis. We established previously that GLI2, a Kruppel-like transcription factor that acts downstream of Hedgehog signaling, is a direct transcriptional target of the TGF-β/SMAD pathway and contributes to melanoma progression, exerting antagonistic activities against M-MITF to control melanoma cell invasiveness. Herein, we dissected the molecular mechanisms underlying both TGF-β and GLI2-driven M-MITF gene repression. Using transient cell transfection experiments with M-MITF promoter constructs, chromatin immunoprecipitation, site-directed mutagenesis, and electrophoretic mobility shift assays, we identified a GLI2 binding site within the -334/-296 region of the M-MITF promoter, critical for GLI2-driven transcriptional repression. This region is, however, not needed for inhibition of M-MITF promoter activity by TGF-β. We determined that TGF-β rapidly repressed protein kinase A activity, thus reducing both phospho-cAMP-response element-binding protein (CREB) levels and CREB-dependent transcription of the M-MITF promoter. Increased GLI2 binding to its cognate cis-element, associated with reduced CREB-dependent transcription, allowed maximal inhibition of the M-MITF promoter via two distinct mechanisms. Topics: Base Sequence; Cell Line, Tumor; Chromatin Immunoprecipitation; Disease Progression; DNA Primers; DNA, Neoplasm; Electrophoretic Mobility Shift Assay; Gene Expression Regulation; Humans; Kruppel-Like Transcription Factors; Melanoma; Microphthalmia-Associated Transcription Factor; Molecular Sequence Data; Nuclear Proteins; Promoter Regions, Genetic; Regulatory Sequences, Nucleic Acid; Transcription, Genetic; Transforming Growth Factor beta; Zinc Finger Protein Gli2 | 2012 |
TGF-β signaling, activated stromal fibroblasts, and cysteine cathepsins B and L drive the invasive growth of human melanoma cells.
Accumulating evidence indicates that interactions between cancer cells and stromal cells are important for the development/progression of many cancers. Herein, we found that the invasive growth of melanoma cells in three-dimensional-Matrigel/collagen-I matrices is dramatically increased on their co-culture with embryonic or adult skin fibroblasts. Studies with fluorescent-labeled cells revealed that the melanoma cells first activate the fibroblasts, which then take the lead in invasion. To identify the physiologically relevant invasion-related proteases involved, we performed genome-wide microarray analyses of invasive human melanomas and benign nevi; we found up-regulation of cysteine cathepsins B and L, matrix metalloproteinase (MMP)-1 and -9, and urokinase- and tissue-type plasminogen activators. The mRNA levels of cathepsins B/L and plasminogen activators, but not MMPs, correlated with metastasis. The invasiveness/growth of the melanoma cells with fibroblasts was inhibited by cell membrane-permeable inhibitors of cathepsins B/L, but not by wide-spectrum inhibitors of MMPs. The IHC analysis of primary melanomas and benign nevi revealed cathepsin B to be predominantly expressed by melanoma cells and cathepsin L to be predominantly expressed by the tumor-associated fibroblasts surrounding the invading melanoma cells. Finally, cathepsin B regulated TGF-β production/signaling, which was required for the activation of fibroblasts and their promotion of the invasive growth of melanoma cells. These data provide a basis for testing inhibitors of TGF-β signaling and cathepsins B/L in the therapy of invasive/metastatic melanomas. Topics: Adult; Cathepsin B; Cathepsin L; Cell Line, Tumor; Cell Proliferation; Embryo, Mammalian; Fibroblasts; Fluorescent Dyes; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Matrix Metalloproteinases; Melanoma; Neoplasm Invasiveness; Nevus; Oligonucleotide Array Sequence Analysis; Protease Inhibitors; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Skin; Skin Neoplasms; Stromal Cells; Transforming Growth Factor beta | 2012 |
Non-canonical Smads phosphorylation induced by the glutamate release inhibitor, riluzole, through GSK3 activation in melanoma.
Riluzole, an inhibitor of glutamate release, has shown the ability to inhibit melanoma cell xenograft growth. A phase 0 clinical trial of riluzole as a single agent in patients with melanoma resulted in involution of tumors associated with inhibition of both the mitogen-activated protein kinase (MAPK) and phophoinositide-3-kinase/AKT (PI3K/AKT) pathways in 34% of patients. In the present study, we demonstrate that riluzole inhibits AKT-mediated glycogen synthase kinase 3 (GSK3) phosphorylation in melanoma cell lines. Because we have demonstrated that GSK3 is involved in the phosphorylation of two downstream effectors of transforming growth factor beta (TGFβ), Smad2 and Smad3, at their linker domain, our aim was to determine whether riluzole could induce GSK3β-mediated linker phosphorylation of Smad2 and Smad3. We present evidence that riluzole increases Smad2 and Smad3 linker phosphorylation at the cluster of serines 245/250/255 and serine 204 respectively. Using GSK3 inhibitors and siRNA knock-down, we demonstrate that the mechanism of riluzole-induced Smad phosphorylation involved GSK3β. In addition, GSK3β could phosphorylate the same linker sites in vitro. The riluzole-induced Smad linker phosphorylation is mechanistically different from the Smad linker phosphorylation induced by TGFβ. We also demonstrate that riluzole-induced Smad linker phosphorylation is independent of the expression of the metabotropic glutamate receptor 1 (GRM1), which is one of the glutamate receptors whose involvement in human melanoma has been documented. We further show that riluzole upregulates the expression of INHBB and PLAU, two genes associated with the TGFβ signaling pathway. The non-canonical increase in Smad linker phosphorylation induced by riluzole could contribute to the modulation of the pro-oncogenic functions of Smads in late stage melanomas. Topics: Animals; Cell Line, Tumor; Gene Expression Regulation, Neoplastic; Glutamic Acid; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Humans; Melanoma; Mice; Phosphorylation; Riluzole; Signal Transduction; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta; Transplantation, Heterologous; Up-Regulation | 2012 |
Critical role of endoglin in tumor cell plasticity of Ewing sarcoma and melanoma.
Tumor cell plasticity enables certain types of highly malignant tumor cells to dedifferentiate and engage a plastic multipotent embryonic-like phenotype, which enables them to 'adapt' during tumor progression and escape conventional therapeutic strategies. This plastic phenotype of aggressive cancer cells enables them to express endothelial cell-specific markers and form tube-like structures, a phenotype that has been linked to aggressive behavior and poor prognosis. We demonstrate here that the transforming growth factor (TGF)-β co-receptor endoglin, an endothelial cell marker, is expressed by tumor cells and its expression correlates with tumor cell plasticity in two types of human cancer, Ewing sarcoma and melanoma. Moreover, endoglin expression was significantly associated with worse survival of Ewing sarcoma patients. Endoglin knockdown in tumor cells interferes with tumor cell plasticity and reduces invasiveness and anchorage-independent growth in vitro. Ewing sarcoma and melanoma cells with reduced endoglin levels showed reduced tumor growth in vivo. Mechanistically, we provide evidence that endoglin, while interfering with TGF-β signaling, is required for efficient bone morphogenetic protein, integrin, focal adhesion kinase and phosphoinositide-3-kinase signaling in order to maintain tumor cell plasticity. The present study delineates an important role of endoglin in tumor cell plasticity and progression of aggressive tumors. Topics: Animals; Antigens, CD; Base Sequence; Bone Morphogenetic Proteins; Cell Division; Cell Line, Tumor; DNA Primers; Endoglin; Female; Focal Adhesion Protein-Tyrosine Kinases; Gene Knockdown Techniques; Humans; Immunohistochemistry; Melanoma; Mice; Mice, Nude; Neoplasm Invasiveness; Phosphatidylinositol 3-Kinases; Receptors, Cell Surface; Sarcoma, Ewing; Signal Transduction; Transforming Growth Factor beta | 2011 |
Efficient TGF-β/SMAD signaling in human melanoma cells associated with high c-SKI/SnoN expression.
SKI and SnoN proteins have been shown to inhibit TGF-β signaling, acting both as transcriptional co-repressors in the cell nucleus, and as sequestrators of SMAD proteins in the cytoplasm. TGF-β, on the other hand, induces rapid, proteasome-mediated, degradation of both proteins. How elevated SKI and SnoN protein levels co-exist with active autocrine TGF-β signaling in cancer cells is yet to be understood.. In this study, we found elevated SKI and SnoN protein levels in a panel of melanoma cell lines, as compared to normal melanocytes. There was no correlation between SKI protein content and the capacity of melanoma cells to invade Matrigel™, to form subcutaneous tumors, or to metastasize to bone after intracardiac inoculation into nude mice. Nor did we find a correlation between SKI expression and histopathological staging of human melanoma. TGF-β induced a rapid and dose-dependent degradation of SKI protein, associated with SMAD3/4 specific transcriptional response and induction of pro-metastatic target genes, partially prevented by pharmacologic blockade of proteasome activity. SKI knockdown in 1205Lu melanoma cells did not alter their invasive capacity or transcriptional responses to TGF-β, and did not allow p21 expression in response to TGF-β or reveal any growth inhibitory activity of TGF-β.. Despite high expression in melanoma cells, the role of SKI in melanoma remains elusive: SKI does not efficiently interfere with the pro-oncogenic activities of TGF-β, unless stabilized by proteasome blockade. Its highly labile nature makes it an unlikely target for therapeutic intervention. Topics: Animals; Cell Line; Cell Line, Tumor; Cell Proliferation; Cyclin-Dependent Kinase Inhibitor p21; DNA-Binding Proteins; Gene Knockdown Techniques; Humans; Intracellular Signaling Peptides and Proteins; Leupeptins; Melanoma; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasm Transplantation; Proteasome Inhibitors; Proto-Oncogene Proteins; RNA Interference; Skin Neoplasms; Smad Proteins; Transcriptional Activation; Transforming Growth Factor beta; Up-Regulation | 2011 |
Expansion of CD16-negative natural killer cells in the peripheral blood of patients with metastatic melanoma.
Altered natural killer (NK) cell function is a component of the global immune dysregulation that occurs in advanced malignancies. Another condition associated with altered NK homeostasis is normal pregnancy, where robust infiltration with CD16- CD9+ NK cells can be identified in decidual tissues, along with a concomitant expansion of CD16- NK cells in the maternal peripheral blood. In metastatic melanoma, we identified a similar expansion of peripheral blood CD16- NK cells (median 7.4% in 41 patients with melanoma compared with 3.0% in 29 controls, P < .001). A subset of NK cells in melanoma patients also expresses CD9, which is characteristically expressed only on NK cells within the female reproductive tract. Expansion of CD16- NK cells was associated with elevated plasma transforming growth factor-beta (TGF-β levels (median 20 ng/ml, Spearman's ρ = 0.81, P = .015)). These findings suggest the possibility of exploring anti-TGF-β therapy to restore NK function in melanoma. Topics: Antigens, CD; Case-Control Studies; Female; Humans; Killer Cells, Natural; Male; Melanoma; Membrane Glycoproteins; Neoplasm Metastasis; Pregnancy; Receptors, IgG; Skin Neoplasms; Tetraspanin 29; Transforming Growth Factor beta | 2011 |
Interferon-alpha induces reversible DNA demethylation of the interferon-induced transmembrane protein-3 core promoter in human melanoma cells.
The interferon (IFN)-α response gene interferon-induced transmembrane protein 3 (IFITM3) has antiproliferative properties in a number of biological systems. In the human melanoma cell line D10, IFITM3 is constitutively expressed and we show that the core promoter is significantly hypomethylated compared to ME15 cells, where IFITM3 is tightly controlled. We demonstrate that treatment of ME15 cells with the demethylating agent 5'-aza-2'-deoxycytidine enhances IFITM3 expression after IFN-α treatment. In a time-course experiment, we show that IFN-α induces demethylation of specific CpG sites of the IFITM3 core promoter 6 h after stimulation and that promoter methylation is precisely re-set to the naïve state 24 h after stimulation. This cyclable modification of methylation requires costimulation with tumor growth factor-beta or expression of the calcium binding protein S100A2, which are known cofactors for enhancement of antiproliferative activity in ME15 cells. Thus, the transcriptional response to IFN-α can be enhanced by promoter demethylation of a subset of inducible genes such as IFITM3. This epigenetic modulation might be crucial to augment the immune response under critical conditions in vivo. Topics: Azacitidine; Biomarkers, Pharmacological; Cell Growth Processes; Cell Line, Tumor; Chemotactic Factors; Decitabine; DNA Methylation; DNA Modification Methylases; Gene Expression Regulation, Neoplastic; Humans; Interferon-alpha; Melanoma; Membrane Proteins; Promoter Regions, Genetic; RNA-Binding Proteins; S100 Proteins; Transforming Growth Factor beta; Transgenes | 2011 |
Constitutive Smad linker phosphorylation in melanoma: a mechanism of resistance to transforming growth factor-β-mediated growth inhibition.
Melanoma cells are resistant to transforming growth factor-β (TGFβ)-induced cell-cycle arrest. In this study, we investigated a mechanism of resistance involving a regulatory domain, called linker region, in Smad2 and Smad3, main downstream effectors of TGFβ. Melanoma cells in culture and tumor samples exhibited constitutive Smad2 and Smad3 linker phosphorylation. Treatment of melanoma cells with the MEK1/2 inhibitor, U0126, or the two pan-CDK and GSK3 inhibitors, Flavopiridol and R547, resulted in decreased linker phosphorylation of Smad2 and Smad3. Overexpression of the linker phosphorylation-resistant Smad3 EPSM mutant in melanoma cells resulted in an increase in expression of p15(INK4B) and p21(WAF1) , as compared with cells transfected with wild-type (WT) Smad3. In addition, the cell numbers of EPSM Smad3-expressing melanoma cells were significantly reduced compared with WT Smad3-expressing cells. These results suggest that the linker phosphorylation of Smad3 contributes to the resistance of melanoma cells to TGFβ-mediated growth inhibition. Topics: Cell Line, Tumor; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p21; Drug Resistance, Neoplasm; Flavonoids; Humans; MAP Kinase Kinase 1; MAP Kinase Kinase 2; Melanoma; Mutation; Phosphorylation; Piperidines; Protein Kinase Inhibitors; Protein Structure, Tertiary; Pyrimidines; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta | 2011 |
Foxp3 expression in melanoma cells as a possible mechanism of resistance to immune destruction.
The forkhead transcription factor Foxp3 is the only definitive marker of CD4(+)CD25(+) regulatory T cells (Tregs) and has been identified as a key regulator in the development and function of Tregs. Foxp3 expression has been reported in a variety of solid tumors, including melanoma. In this study, we validated Foxp3 expression in both tumor-infiltrating Tregs and melanoma cells by performing immunohistochemical analysis of human melanoma tissue sections. Further, we assessed Foxp3 expression in melanoma cell lines by performing flow cytometry, confocal microscopic analysis, reverse transcription-polymerase chain reaction (RT-PCR), and Western blotting. Inhibition of Foxp3 expression in melanoma cells using small interfering RNA (siRNA) resulted in downregulation of B7-H1 and transforming growth factor (TGF)-β expression; in contrast, Foxp3 overexpression resulted in the upregulation of the expression of these proteins. Coculture of Foxp3-expressing melanoma cells with naive CD4(+)CD25(-) T cells resulted in strong inhibition of T-cell proliferation. This antiproliferative effect was partially abrogated by specific inhibition of Foxp3 expression and was effectively enhanced by overexpression of Foxp3. We observed an attenuated antiproliferative effect even when melanoma cells and T cells in the coculture were separated using Transwell inserts. These findings indicated that melanoma cells could have Foxp3-dependent Treg-like suppressive effects on T cells and suggested that the mimicking of Treg function by melanoma cells may represent a possible mechanism of tumor resistance to immune destruction in the melanoma tumor microenvironment. Topics: Antigens, CD; B7-H1 Antigen; CD4 Antigens; Cell Line, Tumor; Cell Proliferation; Coculture Techniques; Forkhead Transcription Factors; Gene Expression Regulation, Neoplastic; Humans; Immunohistochemistry; Interleukin-2 Receptor alpha Subunit; Melanoma; RNA, Small Interfering; Skin Neoplasms; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Transgenes; Tumor Escape | 2011 |
Prognostic significance of RUNX3 expression in human melanoma.
RUNX3 is a tumor suppressor that plays important roles in cell proliferation, apoptosis, and metastasis. The authors investigated the role of RUNX3 in melanoma pathogenesis and analyzed the prognostic impact of RUNX3 expression in a large series of melanoma patients.. Two sets of tissue microarrays were constructed, including 440 cases of melanomas (202 for the training set and 238 for the validation set) and 88 cases of nevi (25 normal nevi and 63 dysplastic nevi). RUNX3 expression was evaluated by immunohistochemistry.. Positive RUNX3 expression was observed in 56%, 54%, 33%, and 24% of the biopsies in normal nevi, dysplastic nevi, primary melanoma, and melanoma metastases, respectively. Significant differences for positive nuclear RUNX3 staining were observed between dysplastic nevi and primary melanomas (P = .002, chi-square test), between dysplastic nevi and melanoma metastases (P < .001, chi-square test), and between primary melanoma and melanoma metastases (P = .045, chi-square test). Loss of RUNX3 expression was correlated with a worse 5-year survival of melanoma patients in both training and validation sets. Furthermore, loss of RUNX3 expression was also correlated with a poor 5-year disease-specific survival in primary melanoma (P = .001) and metastatic melanoma patients (P = .008). Multivariate Cox regression analysis revealed that positive RUNX3 expression is an independent prognostic factor to predict melanoma patient outcome.. Our findings indicate that RUNX3 plays an important role in melanoma pathogenesis and may serve as a promising prognostic marker for melanoma. Topics: Adult; Aged; Core Binding Factor Alpha 3 Subunit; Disease Progression; Female; Humans; Male; Melanoma; Middle Aged; Prognosis; Proportional Hazards Models; Tissue Array Analysis; Transforming Growth Factor beta | 2011 |
GLI2 and M-MITF transcription factors control exclusive gene expression programs and inversely regulate invasion in human melanoma cells.
We recently identified GLI2, the most active of GLI transcription factors, as a direct TGF-β/SMAD target, whose expression in melanoma cells is associated with increased invasiveness and metastatic capacity. In this work, we provide evidence that high GLI2 expression is inversely correlated with that of the melanocyte-specific transcription factor M-microphthalmia transcription factor (M-MITF) and associated transcriptional program. GLI2-expressing cell lines were characterized by the loss of M-MITF-dependent melanocytic differentiation markers and reduced pigmentation. The balance between M-MITF and GLI2 expression did not correlate with the presence or absence of BRAF-activating mutations, but rather was controlled by two distinct pathways: the TGF-β pathway, which favors GLI2 expression, and the protein kinase A (PKA)/cAMP pathway, which pushes the balance toward high M-MITF expression. Furthermore, overexpression and knockdown experiments demonstrated that GLI2 and M-MITF reciprocally repress each other's expression and control melanoma cell invasion in an opposite manner. These findings thus identify GLI2 as a critical transcription factor antagonizing M-MITF function to promote melanoma cell phenotypic plasticity and invasive behavior. Topics: Animals; Cell Line, Tumor; Cell Proliferation; Cyclic AMP; Cyclic AMP-Dependent Protein Kinases; Gene Expression Regulation, Neoplastic; Humans; Kruppel-Like Transcription Factors; Melanoma; Mice; Mice, Nude; Microphthalmia-Associated Transcription Factor; Neoplasm Invasiveness; Nuclear Proteins; Pigmentation; Signal Transduction; Skin Neoplasms; Smad Proteins; Subcutaneous Tissue; Transforming Growth Factor beta; Xenograft Model Antitumor Assays; Zinc Finger Protein Gli2 | 2011 |
SKI promotes Smad3 linker phosphorylations associated with the tumor-promoting trait of TGFbeta.
The transcriptional co-regulator SKI is a potent inhibitor of TGFbeta-growth inhibitory signals. SKI binds to receptor-activated Smads in the nucleus, forming repressor complexes containing HDACs, mSin3, NCoR, and other protein partners. Alternatively, SKI binds to activated Smads in the cytoplasm, preventing their nuclear translocation. SKI is necessary for anchorage-independent growth of melanoma cells in vitro, and most important, for human melanoma xenograft growth in vivo. We recently identified a novel role of SKI in TGFbeta signaling. SKI promotes the switch of Smad3 from repressor of proliferation to activator of oncogenesis by facilitating phosphorylations in the linker domain. High levels of endogenous SKI are required by the tumor promoting trait of TGFbeta to induce expression of the plasminogen-activator inhibitor-1 (PAI-1), sustained expression of C-Myc and for aborting upregulation of p21(Waf-1). Here we discuss how SKI diversifies and amplifies its functions by associating with multiple protein partners and by promoting Smad3 linker phosphorylation(s) in response to TGFbeta signaling in melanoma cells. Topics: Animals; Cyclin-Dependent Kinase Inhibitor p21; DNA-Binding Proteins; Humans; Melanoma; Mice; Phosphorylation; Plasminogen Activator Inhibitor 1; Protein Binding; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-myc; RNA Interference; Signal Transduction; Smad3 Protein; Transforming Growth Factor beta; Transplantation, Heterologous; Up-Regulation | 2010 |
Expression of the embryonic morphogen Nodal in cutaneous melanocytic lesions.
Nodal, a potent embryonic morphogen in the transforming growth factor-beta family, is a proposed key regulator of melanoma tumorigenicity. However, there has been no systematic study of Nodal expression in melanocytic lesions. We investigated Nodal expression by immunohistochemistry in 269 melanocytic lesions, including compound nevi, dysplastic nevi, congenital nevi, Spitz nevi, melanoma in situ, malignant melanoma including the variant desmoplastic melanoma, and metastatic melanoma. We found that the Nodal expression was significantly increased in malignant lesions (including melanoma in situ, malignant melanoma, and metastatic melanoma) compared with compound nevi, Spitz nevi, and dysplastic nevi. Surprisingly, congenital nevi expressed a level of Nodal comparable with malignant lesions, whereas desmoplastic melanoma showed lower expression than nondesmoplastic malignant melanoma (P<0.05). Deep melanoma (Breslow depth >1 mm) displayed a higher percentage of Nodal-positive tumor cells than did superficial melanoma (Breslow depth < or =1 mm), although there was no statistical difference in the overall staining intensity (P=0.18). Melanomas in situ showed a lower level of Nodal expression than did deep melanomas and metastatic melanomas (P<0.05). The low expression of Nodal in normal and dysplastic nevi, and its increasing expression with the progression of malignant lesions, are suggestive of a role for Nodal in melanoma progression. Topics: Biomarkers, Tumor; Dysplastic Nevus Syndrome; Humans; Immunohistochemistry; Melanoma; Nevus, Pigmented; Nodal Protein; Precancerous Conditions; Skin Neoplasms; Transforming Growth Factor beta | 2010 |
Co-regulated expression of matrix metalloproteinase-2 and transforming growth factor-beta in melanoma development and progression.
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 |
The ups and downs of transcription factors in melanoma.
Topics: Bone Neoplasms; Cell Line, Tumor; Cell Survival; Chromosomes, Human, Pair 10; Collagen Type I; Gene Expression Regulation, Neoplastic; Hedgehog Proteins; Humans; Kruppel-Like Factor 6; Kruppel-Like Transcription Factors; Melanoma; Neoplasm Invasiveness; Nuclear Proteins; Proto-Oncogene Proteins; Signal Transduction; Skin Neoplasms; Transcription, Genetic; Transforming Growth Factor beta; Up-Regulation; Zinc Finger Protein Gli2 | 2010 |
Evaluation of molecular markers of mesenchymal phenotype in melanoma.
The epithelial to mesenchymal transition is a developmental process allowing epithelial cells to dedifferentiate into cells displaying mesenchymal phenotypes. The pathological role of epithelial to mesenchymal transition has been implicated in invasion and metastasis for numerous carcinomas, yet limited data exist addressing whether mesenchymal transition (MT) occurs in malignant melanoma cells. Our group developed an in-vitro three-dimensional culture system to address MT in melanoma cells upon transforming growth factor-β/ tumor necrosis factor-α treatment. Loss of E-cadherin is one of the best indicators of MT in epithelial cells. Not surprisingly, E-cadherin was expressed in only three of 12 (25%) melanoma cell lines and all three mesenchymal proteins, N-cadherin, vimentin, and fibronectin, were expressed by seven (58%) melanoma cell lines. However, after cytokine treatment, two or more mesenchymal proteins were elevated in nine (75%) melanoma cell lines. Data support the transforming growth factor-β production by melanoma cells which may induce/support MT. Evaluation of E-cadherin, N-cadherin, and Snail expression in melanoma tissue samples are consistent with an inverse coupling of E-cadherin and N-cadherin expression, however, there are also examples suggesting a more complex control of their expression. These results indicate that malignant melanoma cell lines are susceptible to MT after cytokine treatment and highlight the importance of understanding the effects of cytokines on melanoma to undergo MT. Topics: Antigens, CD; Biomarkers, Tumor; Cadherins; Cell Line, Tumor; Epithelial-Mesenchymal Transition; Female; Humans; Immunohistochemistry; Lung Neoplasms; Male; Melanoma; Phenotype; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Necrosis Factor-alpha | 2010 |
The oncoprotein c-ski functions as a direct antagonist of the transforming growth factor-{beta} type I receptor.
The oncoprotein c-Ski has been implicated in the negative regulation of transforming growth factor-β (TGF-β) signaling owing to its ability to repress Smad transcriptional activity via recruitment of a transcriptional corepressor complex containing histone deacetylases. However, c-Ski has also been shown to localize to the cytoplasm, raising the interesting possibility that it might disable TGF-β signaling through alternative mechanisms. Here, we provide evidence that c-Ski can restrict TGF-β signaling by interacting directly with the activated TGF-β type I receptor (TβRI). We explored the physiologic relevance of the c-Ski/TβRI interaction and found that it can culminate in a constitutive association of TβRI with a nonfunctional R-Smad/Smad4 complex. Based on these findings, we hypothesize that the interaction between c-Ski and TβRI might interfere with nuclear translocation of the R-Smad/Smad4 complex, thereby attenuating TGF-β signaling. Such a mechanism may play a crucial role in tumor progression, because many tumors that express high levels of c-Ski also display impaired nuclear accumulation of Smads. Topics: Adenocarcinoma; Breast Neoplasms; Cell Nucleus; Cytoplasm; DNA-Binding Proteins; Female; Humans; Immunoenzyme Techniques; Intracellular Signaling Peptides and Proteins; Lung Neoplasms; Melanoma; Phosphorylation; Protein Serine-Threonine Kinases; Protein Transport; Proto-Oncogene Proteins; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Signal Transduction; Skin; Smad Proteins, Receptor-Regulated; Smad2 Protein; Smad3 Protein; Tissue Array Analysis; Transforming Growth Factor beta; Tumor Cells, Cultured | 2010 |
Regulation of melanin synthesis by the TGF-beta family in B16 melanoma cells.
Effects of representative members of the transforming growth factor-beta (TGF-beta) family, TGF-beta1, activin A and BMP-2, on melanin content and expression of pigment-producing enzymes were examined in B16 melanoma cells. Treatment with TGF-beta1 or activin A but not with BMP-2 significantly decreased melanin content and expression of Tyrosinase and Tyrp-1, suggesting an inhibitory effect of TGF-beta1 and activin A on melanin synthesis. TGF-beta1 completely inhibited melanin synthesis induced by alpha-melanin stimulating hormone (alpha-MSH), whereas activin A only slightly did. As compared with parental B16 cells, the inhibitory effects of TGF-beta1 and activin A on melanin content were relative smaller in B16 F10 cells, a subline of B16 cells that contain more pigment. The present study indicates that in addition to TGF-beta, activin negatively regulates melanogenesis in the absence of alpha-MSH, but that the activity in the presence of alpha-MSH was slightly different between TGF-beta and activin. Topics: Activins; Bone Morphogenetic Protein 2; Cell Line, Tumor; Gene Expression Regulation, Neoplastic; Humans; Melanins; Melanocyte-Stimulating Hormones; Melanoma; Membrane Glycoproteins; Monophenol Monooxygenase; Oxidoreductases; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2009 |
Clinical significance of the expression of c-Ski and SnoN, possible mediators in TGF-beta resistance, in primary cutaneous melanoma.
Loss of TGF-beta growth control is considered as a hallmark of several human neoplasms including melanoma. Resistance of cancer cells to TGF-beta has been linked to mutations in proteins involved in the TGF-beta pathway. In melanoma such mutations have not been observed. C-Ski and SnoN, two structurally and functionally highly homologous proteins, are known as negative regulators in the TGF-beta signaling pathway. C-Ski and SnoN expression levels and subcellular localization have been associated with clinicopathological parameters and tumour progression in several human malignancies. In melanoma cell lines, high c-Ski and SnoN expression levels have been described.. The objective of this study was to evaluate the clinical value of c-Ski and SnoN expression in primary cutaneous melanoma.. We evaluated c-Ski and SnoN expression by immunohistochemical staining in 120 primary melanomas. Possible associations between c-Ski and SnoN staining patterns and clinicopathological parameters were analyzed.. Nuclear c-Ski expression was significantly associated with thicker and ulcerated tumours. The percentage of SnoN positivity was higher in ulcerated tumours and in the sentinel node positive group.. These results suggest that c-Ski and SnoN, mediators in TGF-beta resistance, might be implicated in melanoma growth and progression. Topics: Cell Proliferation; Disease Progression; DNA-Binding Proteins; Female; Humans; Intracellular Signaling Peptides and Proteins; Male; Melanoma; Middle Aged; Proto-Oncogene Proteins; Retrospective Studies; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta | 2009 |
Increased survival from stage IV melanoma associated with fewer regulatory T Cells.
Melanoma often elicits a profound immune response, and this response has been exploited by various immune therapies. These immunotherapies ultimately fail, however, and advanced melanoma is uniformly fatal, suggesting the development of an immune escape mechanism. In this study, markers of immune escape including regulatory T cells (T(regs)), dendritic cells (DCs), and TGF-beta were evaluated in 14 Stage IV melanoma patients and correlated with survival.. Peripheral blood mononuclear cells were isolated from Stage IV melanoma patients and analyzed for T(regs) and DCs by flow cytometry using fluorescent CD3, CD4, CD25, Lin, HLA-DR, CD11c, and CD123 antibodies. Serum TGF-beta levels were evaluated by ELISA from these patients. Clinical data were extracted from the patients' medical records.. Stage IV melanoma patients with shorter survival (less than 24 mo) had a significantly higher proportion of T(regs) than those with longer survival (15% versus 8%, respectively, P = 0.004). The numbers of DCs and the serum TGF-beta levels were not significantly different in these two groups. There was an inverse relationship between the percentage of T(regs) and survival, although this did not reach statistical significance (r = -0.35, P = 0.22). There was also an inverse relationship between peripheral T(regs) and DCs. When patients were divided into groups of greater than or less than 7% T(regs), the number of total DCs was higher in the patients with fewer T(regs) than in those with more T(regs), but this did not reach statistical significance (16,535 versus 12,126 total DCs/mL, P = 0.52).. In Stage IV melanoma patients, a high percentage of T(regs) appears to be associated with shorter survival. The inverse relationship of the number of DCs and T(regs) in these patients may provide an insight to the origin of this observation. Topics: Antigens, CD; Dendritic Cells; Enzyme-Linked Immunosorbent Assay; Flow Cytometry; HLA-DR Antigens; Humans; Leukocytes, Mononuclear; Lymphocyte Count; Melanoma; Survival Rate; Survivors; T-Lymphocytes, Regulatory; Transforming Growth Factor beta | 2009 |
Comparison of stable human Treg and Th clones by transcriptional profiling.
From cancerous and non-cancerous patients, we derived stable clones of CD4(+) Treg, defined as clones that expressed high CD25 at rest, were anergic in vitro, and suppressed the proliferation of co-cultured CD4(+) cells. A conserved region of FOXP3 intron 1 was demethylated in all Treg clones, whereas it was methylated in non-regulatory Th and CTL clones. In our panel of human clones, this stable epigenetic mark correlated better with suppressive activity than did FOXP3 mRNA or protein expression. We used expression microarrays to compare Treg and Th clones after activation, which is required for suppressive function. The transcriptional profile that is specific of activated Treg clones includes a TGF-beta signature. Both activated Treg and Th clones produced the latent form of TGF-beta. However, SMAD2 phosphorylation was observed after activation in the Treg but not in the Th clones, indicating that only activated Treg clones produced the bioactive form of TGF-beta. A TGF-beta signature was also displayed by a Th clone "suppressed" by a Treg clone. In conclusion, the hallmark of our panel of activated human Treg clones is to produce bioactive TGF-beta which has autocrine actions on Tregs and can have paracrine actions on other T cells. Topics: Clone Cells; DNA Methylation; Female; Forkhead Transcription Factors; Gene Expression Profiling; Humans; Lymphocyte Activation; Male; Melanoma; Skin Neoplasms; Smad2 Protein; T-Lymphocytes, Helper-Inducer; T-Lymphocytes, Regulatory; Transforming Growth Factor beta | 2009 |
Id2 suppression of p15 counters TGF-beta-mediated growth inhibition of melanoma cells.
Proliferative resistance to transforming growth factor beta (TGF-beta) is regarded as a critical turning point in the malignant progression of many cancer types. In melanoma this resistance is associated with more aggressive metastatic behaviour. A recent study by our group identified proliferative and invasive subtypes of melanoma cultures and found that these are, respectively, susceptible and resistant to TGF-beta suppression of proliferation. Here, using previously characterised proliferative and invasive phenotype melanoma cultures, we explored molecular responses involved in modulating susceptibility to TGF-beta-mediated inhibition of proliferation. The Id2 gene was identified as being expressed more strongly in invasive phenotype cells less susceptible to TGF-beta repression than in proliferative phenotype cells. We correlated TGF-beta repression of Id2 gene expression in proliferative phenotype cells with p15(Ink4b) induction and cell cycle arrest. Furthermore, ectopic Id2 expression in proliferative phenotype cells counteracted p15(Ink4b) induction and consequently protected them from TGF-beta-mediated inhibition of proliferation. We conclude that transition to increased aggressiveness in melanoma cells requires Id2 upregulation to suppress TGF-beta induction of p15(Ink4b) and thus help to circumvent TGF-beta-mediated inhibition of proliferation. Topics: Cell Line, Tumor; Cell Proliferation; Cyclin-Dependent Kinase Inhibitor p15; G1 Phase; Gene Expression Regulation, Neoplastic; Humans; Inhibitor of Differentiation Protein 2; Melanoma; Neoplasm Invasiveness; Transforming Growth Factor beta | 2009 |
Beneficial regulation of matrixmetalloproteinases and their inhibitors, fibrillar collagens and transforming growth factor-beta by Polypodium leucotomos, directly or in dermal fibroblasts, ultraviolet radiated fibroblasts, and melanoma cells.
The extracellular matrix (ECM) that gives tissue its structural integrity is remodeled in skin aging/photoaging and cancer via the increased expression/activities of matrixmetalloproteinases (MMP), inhibition of the tissue inhibitors of matrix metalloproteinases (TIMP), or inhibition of collagen synthesis. Transforming growth factor-beta (TGF-beta), a predominant regulator of the ECM, is inhibited in aging/photoaging and stimulated in carcinogenesis. P. leucotomos (fern) extract has potential to counteract these alterations via its antioxidant, anti-inflammatory and photoprotective properties. The goal of this research was to determine the efficacy of P. leucotomos to (a) directly inhibit MMP-1, 2, 3, and 9 activities, (b) inhibit MMP-2, and stimulate TIMPs, fibrillar collagens and TGF-beta in non-irradiated or ultraviolet (UV) radiated fibroblasts, and (c) inhibit MMPs and TGF-beta, and stimulate TIMPs in melanoma cells. To this purpose, we examined the direct effect of P. leucotomos (0-1%) on MMPs' activities, and its effects on the expression (protein and/or transcription levels) of (1) MMPs and TIMPs in dermal fibroblasts, and melanoma cells, (2) TGF-beta in non-irradiated, UVA (2.5 J/cm2) or UVB (2.5 mJ/cm2) irradiated fibroblasts, and melanoma cells, and (3) types I, III, and V collagen in non-irradiated or UV irradiated fibroblasts. P. leucotomos directly inhibited the activities of MMPs as well as the expression of MMPs in fibroblasts, and melanoma cells while stimulating the expression of TIMPs in these cells. P. leucotomos stimulated types I, III, and V collagen in non-irradiated fibroblasts, and types I and V collagen in UV radiated fibroblasts. P. leucotomos had predominant stimulatory effects on TGF-beta expression in non-irradiated or UV radiated fibroblasts, and inhibited TGF-beta expression in melanoma cells. The effects of P. leucotomos were largely similar to that of ascorbic acid. P. leucotomos demonstrated dual protective effects on the ECM via its inhibition of the ECM proteolytic enzymes and the stimulation of the structural ECM collagens. The effects of P. leucotomos on fibroblasts and melanoma cells may be partly via its cell-specific regulation of TGF-beta expression and partly via its antioxidant property. The intake or topical application of P. leucotomos may be beneficial to skin health, in aging and cancer prevention or treatment. Topics: Antioxidants; Ascorbic Acid; Cells, Cultured; Cytoprotection; Extracellular Matrix; Fibrillar Collagens; Fibroblasts; Gene Expression Regulation; Humans; Infant, Newborn; Matrix Metalloproteinases; Melanoma; Plant Extracts; Polypodium; Skin Aging; Tissue Inhibitor of Metalloproteinases; Transforming Growth Factor beta; Ultraviolet Rays | 2009 |
Maspin expression, angiogenesis, prognostic parameters, and outcome in malignant melanoma.
Maspin is a serine protease inhibitor that is thought of as a tumor suppressor because of observations that loss of maspin expression in breast, prostate, and oral cancer is associated with poor prognosis. In addition, maspin may function as an inhibitor of angiogenesis. However, it has been correlated with malignant behavior in pancreatic and ovarian cancer. The role of maspin in malignant melanoma (MM) has not yet been systematically examined.. We aimed to examine the immunohistochemical expression of maspin and several proangiogenic factors (vascular endothelial growth factor, transforming growth factor-beta, alphaVbeta3 integrin, cyclooxygenase-2, and CD44) in MM and correlate each to angiogenesis, tumor thickness, and outcome.. In all, 77 formalin-fixed, paraffin-embedded MM samples were immunostained for maspin and other proangiogenic factors (vascular endothelial growth factor, transforming growth factor-beta, alphaVbeta3 integrin, cyclooxygenase-2, and CD44) and were correlated with angiogenesis as mean microvessel density. Three normal-appearing skin samples and 10 nevi were also immunostained for maspin. Breslow thickness, Clark level, clinical stage, and follow-up information were obtained for outcome analysis.. Immunohistochemical analysis revealed strong nuclear melanocytic maspin expression in all 10 nevi (half of which were dysplastic) but none in melanocytes from 3 normal-appearing skin samples. Strong nuclear maspin staining was demonstrated in 78% of radial phase melanoma and 46% of vertical growth phase melanoma. In addition, there was a significant inverse relationship between maspin and microvessel density (P = .018) and tumor thickness greater than 0.76 mm (P = .007), indicating that maspin is expressed in thinner tumors with less angiogenesis. Conversely, vascular endothelial growth factor expression, Clark level, and Breslow thickness all significantly correlated with microvessel density (P = .047, P = .027, and P = .011, respectively). Cyclooxygenase-2 expression significantly correlated with thicker tumors (P = .006) but not with angiogenesis (P = .714). In addition, Clark level, Breslow thickness, and stage were all significant predictors of overall survival (P < .001, P = .005, and P < .001, respectively).. This study represents a single institution.. These results demonstrate maspin expression in nevi and radial growth phase melanoma, but this expression seems to be lost in the transition from radial growth phase to vertical growth phase melanoma. In addition, maspin is correlated with decreased angiogenesis and tumor thickness less than 0.76 mm in MM. These results indicate maspin may function as a tumor suppressor in MM. Topics: Cyclooxygenase 2; Female; Follow-Up Studies; Humans; Hyaluronan Receptors; Immunohistochemistry; Integrin alphaVbeta3; Male; Melanoma; Neovascularization, Pathologic; Prognosis; Serpins; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A | 2009 |
Cancer cell growth and extracellular matrix remodeling mechanism of ascorbate; beneficial modulation by P. leucotomos.
Ascorbate has dose-dependent inverse effects on cancer cells growth and expression of matrixmetalloproteinases (MMP) and transforming growth factor-beta (TGF-beta), which regulate extracellular matrix (ECM) remodeling. We examined melanoma cell viability and ECM remodeling mechanisms of ascorbate and its modulation by an extract from Polypodium leucotomos (PL) (a fern) via the regulation of apoptosis, heat-shock proteins (HSPs), MMP-1 or tissue inhibitors of matrix metalloproteinase-1 (TIMP-1) that inhibits MMP-1. The dose-dependent regulation of cell viability/proliferation by ascorbate was associated with inverse regulation of apoptosis and stimulation of HSPs at growth-inhibitory concentrations. PL antagonized the stimulation of MMP-1, TGF-beta and HSPs by a growth-inhibitory ascorbate dose and stimulated the expression of TIMP-1, while maintaining growth inhibition. We infer that a combination of ascorbate with PL is beneficial to cancer management via the simultaneous inhibition of cell growth and expression of MMP-1, TGF-beta and HSPs, and furthermore, the stimulation of TIMP-1. Topics: Antineoplastic Combined Chemotherapy Protocols; Antioxidants; Ascorbic Acid; Caspase 3; Cell Proliferation; Cells, Cultured; Drug Synergism; Enzyme-Linked Immunosorbent Assay; Extracellular Matrix; Heat-Shock Proteins; Humans; Matrix Metalloproteinase 1; Matrix Metalloproteinase Inhibitors; Melanoma; Plant Extracts; Polypodium; Tissue Inhibitor of Metalloproteinase-1; Transforming Growth Factor beta; Tumor Cells, Cultured | 2009 |
SKI knockdown inhibits human melanoma tumor growth in vivo.
The SKI protein represses the TGF-beta tumor suppressor pathway by associating with the Smad transcription factors. SKI is upregulated in human malignant melanoma tumors in a disease-progression manner and its overexpression promotes proliferation and migration of melanoma cells in vitro. The mechanisms by which SKI antagonizes TGF-beta signaling in vivo have not been fully elucidated. Here we show that human melanoma cells in which endogenous SKI expression was knocked down by RNAi produced minimal orthotopic tumor xenograft nodules that displayed low mitotic rate and prominent apoptosis. These minute tumors exhibited critical signatures of active TGF-beta signaling including high levels of nuclear Smad3 and p21(Waf-1), which are not found in the parental melanomas. To understand how SKI promotes tumor growth we used gain- and loss-of-function approaches and found that simultaneously to blocking the TGF-beta-growth inhibitory pathway, SKI promotes the switch of Smad3 from tumor suppression to oncogenesis by favoring phosphorylations of the Smad3 linker region in melanoma cells but not in normal human melanocytes. In this context, SKI is required for preventing TGF-beta-mediated downregulation of the oncogenic protein c-MYC, and for inducing the plasminogen activator inhibitor-1, a mediator of tumor growth and angiogenesis. Together, the results indicate that SKI exploits multiple regulatory levels of the TGF-beta pathway and its deficiency restores TGF-beta tumor suppressor and apoptotic activities in spite of the likely presence of oncogenic mutations in melanoma tumors. Topics: Animals; DNA-Binding Proteins; Gene Knockdown Techniques; Humans; Male; Melanoma; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Invasiveness; Phosphorylation; Plasminogen Activator Inhibitor 1; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-myc; RNA Interference; Signal Transduction; Smad3 Protein; Transforming Growth Factor beta; Transplantation, Heterologous; Tumor Cells, Cultured | 2009 |
Clinical markers and driving mechanisms in melanoma progression: VEGF-C, RhoC, c-Ski/SnoN and EGFR.
This research project aimed at evaluating the clinical and prognostic value of different molecules involved in signalling transduction pathways involved in melanoma progression. Vascular endothelial growth factor-C or VEGF-C induces lymphangiogenesis. This study showed high VEGF-C expression to be associated with the presence of a positive sentinel lymph node. The presence of VEGF-C expression in melanoma cells was associated with reduced disease free and overall survival. RhoC is important in the organization of the actin filamental system. We observed RhoC mRNA and protein expression to be upregulated in a highly metastatic melanoma cell line (DX3aza), whereas only low expression levels were found in a melanoma cell line with low proliferative and invasive capacity (MeWo). RhoC immunoreactivity in melanoma tissue was associated with high Breslow tumour thickness and the presence of ulceration. C-Ski and SnoN have been identified as negative regulators in the TGF-beta pathway. We found a significant association between the presence of nuclear c-Ski and thicker, ulcerated melanomas. SnoN expression was associated with the presence of ulceration and a positive sentinel lymph node. Epidermal Growth Factor Receptor (EGFR) expression has been associated with tumour progression and poor outcome in a variety of solid tumours, EGFR immunoreactivity was more frequently present in patients with a positive sentinel lymph node. EGFR gene amplification was not observed; however, the presence of polysomy was associated with higher Breslow tumour thickness. Treating BLM melanoma cells with different concentrations of cetuximab reduced the invasive capacity of the melanoma cells, without impact on cell viability and growth. Topics: Adult; Cell Line, Tumor; Disease Progression; ErbB Receptors; Female; Gene Expression Regulation, Neoplastic; Humans; Intracellular Signaling Peptides and Proteins; Lymphatic Metastasis; Male; Melanoma; Middle Aged; Neoplasm Invasiveness; Proto-Oncogene Proteins; rho GTP-Binding Proteins; rhoC GTP-Binding Protein; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta; Vascular Endothelial Growth Factor C | 2009 |
SKI is critical for repressing the growth inhibitory function of TGF-beta in human melanoma.
Topics: Cell Proliferation; Disease Progression; DNA-Binding Proteins; Down-Regulation; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Proto-Oncogene Proteins; Signal Transduction; Transforming Growth Factor beta | 2008 |
Transforming growth factor-beta activation promotes genetic context-dependent invasion of immortalized melanocytes.
Accumulation of distinct sets of genetic/epigenetic alterations is thought to contribute to stepwise progression of human cutaneous melanomas. We found evidence of frequent tumor cell autonomous transforming growth factor-beta (TGF-beta) signal activation in both premalignant and malignant stages of human cutaneous melanoma histogenesis and investigated its potential causative roles using human organotypic skin cultures. PTEN deficiency and Braf activation, two common coincident genetic alterations found in primary cutaneous melanomas, were first introduced into human melanocytes previously immortalized by the SV40 large T antigen and telomerase. These changes individually supported anchorage-independent growth and conferred benign, hyperplastic growth in a skin-like environment. In addition, PTEN deficiency combined with Braf activation together induced a melanoma in situ-like phenotype without dermal invasion. Further addition of cell autonomous TGF-beta activation in the context of PTEN deficiency and Braf activation promoted dermal invasion in skin cultures without significantly promoting proliferation in vitro and in vivo. This proinvasive phenotype of cell autonomous TGF-beta activation is genetic context-dependent, as hyperactivating the TGF-beta type I receptor without PTEN deficiency and Braf activation failed to induce an invasive behavior. Evidence of genetic interactions among PTEN deficiency, Braf activation, and cell autonomous TGF-beta activation shows that distinct stages of human melanoma are genetically tractable in the proper tissue architecture. Topics: Apoptosis; Cell Cycle; Cell Line, Transformed; Cell Proliferation; Humans; Melanocytes; Melanoma; Neoplasm Invasiveness; PTEN Phosphohydrolase; Transforming Growth Factor beta | 2008 |
Aggressive melanoma cells escape from BMP7-mediated autocrine growth inhibition through coordinated Noggin upregulation.
Bone morphogenetic proteins (BMPs) are members of the TGF-beta superfamily responsible for mediating a diverse array of cellular functions both during embryogenesis and in adult life. Previously, we reported that upregulation of BMP7 in human melanoma correlates with tumor progression. However, melanoma cells are either inhibited by or become resistant to BMP7 as a function of tumor progression, with normal melanocytes being most susceptible. Herein, real-time quantitative reverse transcriptase-polymerase chain reactions and western blotting revealed that the expression of BMP antagonist, Noggin, correlates with resistance to BMP7 in advanced melanoma cells. To test the hypothesis that coordinated upregulation of Noggin protects advanced melanoma cells from autocrine inhibition by BMP7, functional expression of Noggin in susceptible melanoma cells was achieved by adenoviral gene transfer. The Noggin-overexpressing cells exhibited a growth advantage in response to subsequent BMP7 transduction in vitro under anchorage-dependent and -independent conditions, in three-dimensional skin reconstructs, as well as in vivo in severe combined immunodeficient mice. In concordance, Noggin knockdown by lentiviral shRNA confers sensitivity to BMP7-induced growth inhibition in advanced melanoma cells. Our findings suggest that, like TGF-beta, BMP7 acts as an autocrine growth inhibitor in melanocytic cells, and that advanced melanoma cells may escape from BMP7-induced inhibition through concomitant aberrant expression of Noggin. Topics: Adenoviridae; Autocrine Communication; Bone Morphogenetic Protein 7; Bone Morphogenetic Proteins; Carrier Proteins; Cell Culture Techniques; Cell Line, Tumor; Cell Movement; Cell Proliferation; Growth Inhibitors; Humans; Melanocytes; Melanoma; Neoplasm Invasiveness; Transduction, Genetic; Transforming Growth Factor beta; Up-Regulation | 2008 |
Potential for cripto-1 in defining stem cell-like characteristics in human malignant melanoma.
The diagnosis of melanoma is becoming ever more frequent. Although surgical excision of early lesions is associated with relatively significant high cure rates, treatment modalities are largely unsuccessful for advanced disease. Characteristics such as cellular heterogeneity and plasticity, expression of certain molecules such as the multidrug resistance protein-1 (MDR1) or the aberrant expression of embryonic signaling molecules and morphogens like Nodal, important for self renewal and pluripotency, suggest that a stem cell-like population may reside in aggressive melanomas. This perspective focuses on preliminary findings obtained in our laboratory which indicate that the expression of the Nodal coreceptor, Cripto-1, in a subset of malignant melanoma cells may be exploited to identify possible melanoma stem cells (MSC). In fact, the use of anti-Cripto-1 antibodies to cell sort Cripto-1-positive cells in the metastatic melanoma cell line C8161 has identified a slow growing, sphere forming subpopulation that expresses increased levels of Oct4, Nanog and MDR1. If current in vivo studies confirm the self renewal and tumorigenic characteristics of these cells, the expression of Cripto-1 may represent a useful marker to identify cancer stem cells in melanoma, and possibly other aggressive tumors as well. Topics: ATP Binding Cassette Transporter, Subfamily B; ATP Binding Cassette Transporter, Subfamily B, Member 1; Biomarkers, Tumor; Cell Line, Tumor; Epidermal Growth Factor; Epigenesis, Genetic; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; Melanoma; Membrane Glycoproteins; Neoplasm Proteins; Neoplastic Stem Cells; Nodal Protein; Octamer Transcription Factor-3; Protein Kinases; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta | 2008 |
TGF-beta targets PAX3 to control melanocyte differentiation.
In the absence of UV radiation, keratinocytes secrete TGF-beta which blocks melanocyte differentiation. Yang et al. (2008), in a recent issue of Molecular Cell, show that TGF-beta induces Smad signaling in melanocytes to repress PAX3, which encodes a transcription factor crucial for melanocyte differentiation. In the presence of UV radiation, a Jnk/AP-1 pathway represses TGF-beta, which together with a UV-induced p53 pathway promotes melanocyte differentiation. Topics: Animals; Cell Differentiation; Humans; Keratinocytes; Melanocytes; Melanoma; Models, Biological; Neoplasm Invasiveness; Paired Box Transcription Factors; PAX3 Transcription Factor; Signal Transduction; Stem Cells; Transforming Growth Factor beta; Tumor Suppressor Protein p53; Ultraviolet Rays | 2008 |
Use of FoxP3 expression to identify regulatory T cells in healthy dogs and dogs with cancer.
Regulatory T cells (Treg) are a distinct group of T lymphocytes with immunosuppressive properties that serve normally to prevent harmful autoimmune responses. However, Tregs can also interfere with beneficial immune responses such as anti-tumor and anti-viral immunity in humans and rodents. Given the overall importance of Tregs, it is likely that they play an important role in diseases of dogs as well. However, at present reagents required for identification of Tregs in dogs are not available. Therefore, we investigated whether expression of FoxP3, a transcription factor that is highly expressed in Tregs in humans and rodents could also be used to identify Tregs in dogs. We found that a cross-reactive FoxP3 antibody identified a subset of CD4(+) T cells in blood and lymph nodes of dogs. By flow cytometry the mean percentage of FoxP3(+)CD4(+) T cells in normal dogs was 4.3% in blood and 9.8% in the lymph nodes. In dogs with cancer, there was a significant increase in numbers of Treg in blood (7.5%) and tumor-draining lymph nodes (17.1%) compared to age-matched healthy control dogs. We also found that FoxP3(+)CD4(+) T cells in dogs could be significantly expanded in vitro by TCR activation together with addition of TGF-beta and IL-2. Treated cells also significantly increased expression of TGF-beta and IL-10mRNA. We conclude from these studies that a cross-reactive FoxP3 antibody can be used to identify Tregs in dogs and that this reagent may serve as a useful tool for investigating the role of Treg in a variety of diseases of dogs. Topics: Animals; Biomarkers, Tumor; Dog Diseases; Dogs; Flow Cytometry; Forkhead Transcription Factors; Interleukin-10; Lymph Nodes; Lymphocyte Activation; Melanoma; Osteosarcoma; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Statistics, Nonparametric; T-Lymphocytes, Regulatory; Transforming Growth Factor beta | 2007 |
Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis.
Melanoma has a propensity to metastasize to bone, where it is exposed to high concentrations of transforming growth factor-beta (TGF-beta). Because TGF-beta promotes bone metastases from other solid tumors, such as breast cancer, we tested the role of TGF-beta in melanoma metastases to bone. 1205Lu melanoma cells, stably transfected to overexpress the natural TGF-beta/Smad signaling inhibitor Smad7, were studied in an experimental model of bone metastasis whereby tumor cells are inoculated into the left cardiac ventricle of nude mice. All mice bearing parental and mock-transfected 1205Lu cells developed osteolytic bone metastases 5 weeks post-tumor inoculation. Mice bearing 1205Lu-Smad7 tumors had significantly less osteolysis on radiographs and longer survival compared with parental and mock-transfected 1205Lu mice. To determine if the reduced bone metastases observed in mice bearing 1205Lu-Smad7 clones was due to reduced expression of TGF-beta target genes known to enhance metastases to bone from breast cancer cells, we analyzed gene expression of osteolytic factors, parathyroid hormone-related protein (PTHrP) and interleukin-11 (IL-11), the chemotactic receptor CXCR4, and osteopontin in 1205Lu cells. Quantitative reverse transcription-PCR analysis indicated that PTHrP, IL-11, CXCR4, and osteopontin mRNA steady-state levels were robustly increased in response to TGF-beta and that Smad7 and the TbetaRI small-molecule inhibitor, SB431542, prevented such induction. In addition, 1205Lu-Smad7 bone metastases expressed significantly lower levels of IL-11, connective tissue growth factor, and PTHrP. These data suggest that TGF-beta promotes osteolytic bone metastases due to melanoma by stimulating the expression of prometastatic factors via the Smad pathway. Blockade of TGF-beta signaling may be an effective treatment for melanoma metastasis to bone. Topics: Animals; Bone Neoplasms; Female; Humans; Melanoma; Mice; Mice, Nude; Neoplasm Metastasis; Smad7 Protein; Survival Analysis; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured | 2007 |
Bone morphogenetic protein 7 (BMP7) expression is a potential novel prognostic marker for recurrence in patients with primary melanoma.
The purpose of this study was to investigate whether protein expression of bone morphogenetic protein 7 (BMP7) is associated with clinico-pathologic characteristics in benign and malignant melanocytic skin tumors. Tissue microarrays (TMAs) were used to analyze BMP7 expression and the Ki-67 labeling index immunohistochemically. Expression was scored semi quantitatively (0-2+). BMP7 protein expression of any intensity (1+-2+) was detected in 50.2% (153/305) of informative cases. In general, BMP7 expression was significantly induced in malignant melanomas (P< 0.001) and melanoma metastases (P< 0.001), compared to benign nevi. Additionally, expression of BMP7 in primary melanomas was associated with Ki-67 labeling index > 5% suggesting that induction of BMP7 expression is associated with proliferation (P=0.028). None of the other clinical and histological factors analyzed was significantly related to BMP7 expression. Interestingly, lymph node metastases demonstrated a significantly higher BMP7 expression compared to skin metastases (P<0.01). Strong BMP7 expression (score 2+) was significantly associated with shorter tumor recurrence (P< 0.05). In summary, induction of BMP7 expression is frequent in melanomas and may serve as a novel prognostic marker of progression in melanoma patients. Topics: Biomarkers, Tumor; Bone Morphogenetic Protein 7; Bone Morphogenetic Proteins; Female; Humans; Immunohistochemistry; Kaplan-Meier Estimate; Male; Melanoma; Middle Aged; Neoplasm Recurrence, Local; Prognosis; Skin Neoplasms; Tissue Array Analysis; Transforming Growth Factor beta | 2007 |
Melanoma induces immunosuppression by up-regulating FOXP3(+) regulatory T cells.
The immune response to melanoma is rarely curative, suggesting the emergence of immunosuppression. FOXP3-expressing regulatory T cells (T(reg) cells) function to suppress immune responses. The objective of this study was to determine if melanoma evades immune surveillance, in part, by inducing T(reg) cells.. Peripheral blood mononuclear cells (PBMCs) were isolated and exposed to melanoma-conditioned media (MCM) or control media for 1 week. The induction of T(reg) cells in these PBMCs was determined by measuring the proportion of CD25(+)FOXP3(+) T cells in all CD4(+) T cells by flow cytometry. FOXP3 expression was determined by mean fluorescence intensity (MFI) and Western blot. Supernatant cytokines were determined by ELISA.. Normal PBMCs exposed to MCM revealed higher proportions of T(reg) cells than those exposed to control media after 6 days (3.4% versus 1.3%, respectively, P < 0.02). The expression of FOXP3 in T(reg) cells from PBMCs exposed to MCM increased over time by MFI and Western blot but was not significantly different than those exposed to control media. The level of IL-10 and TGF-beta in supernatants after 6 days growth was higher in MCM than control media, but this did not reach statistical significance.. Exposure of PBMCs to melanoma results in induction of FOXP3(+) T(reg) cells. Topics: CD4-Positive T-Lymphocytes; Culture Media, Conditioned; Forkhead Transcription Factors; Humans; Immunity, Cellular; Immunosuppression Therapy; Interleukin-10; Melanoma; Skin Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Up-Regulation | 2007 |
Immunosuppressive dendritic and regulatory T cells are upregulated in melanoma patients.
Immunologic therapies for melanoma rarely succeed, suggesting a persistent counter-regulatory immune modulation. Regulatory T cells (T(regs)) and plasmacytoid subpopulations of dendritic cells (pDCs) inhibit the immune response. We hypothesize that melanoma upregulates T(regs )and subpopulations of immunosuppressive dendritic cells (DCs).. Peripheral blood mononuclear cells (PBMCs) were obtained from healthy controls, stage I and stage IV melanoma patients. T(regs )were identified as CD4+ and CD25(hi). Dendritic cells were identified using a DC cocktail of antibodies including CD11c+ myeloid dendritic cells (mDCs) and CD123+ pDCs. Serum transforming growth factor-beta (TGF-beta), interleukin-4 (IL-4) and interleukin-10 (IL-10) levels were determined by enzyme-linked immunosorbent assay (ELISA). Statistical analysis was performed using analysis of variance (ANOVA).. Stage IV melanoma patients had a doubling of regulatory T cells compared to both normal subjects and stage I melanoma patients. There was a significantly higher number of DCs in all melanoma patients compared to normal subjects. Stage I melanoma patients had a significantly higher number of pDCs than normal subjects, and all melanoma patients had a higher concentration of mDCs than controls. Serum IL-4 and IL-10 were not detectable but serum TGF-beta levels were significantly higher in stage I and stage IV melanoma patients compared to normal controls.. Advanced melanoma is associated with increased numbers of circulating dendritic cells and regulatory T cells. These data suggest that melanoma induces immunosuppressive DCs and regulatory T cells in the systemic circulation. Topics: Aged; CD4 Antigens; Dendritic Cells; Enzyme-Linked Immunosorbent Assay; Female; Humans; Immune Tolerance; Interleukin-10; Interleukin-2 Receptor alpha Subunit; Interleukin-3 Receptor alpha Subunit; Interleukin-4; Lymphocyte Count; Male; Melanoma; Middle Aged; Neoplasm Staging; Skin Neoplasms; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Tumor Escape; Up-Regulation | 2007 |
Expression of collagenase-1 (MMP-1) promotes melanoma growth through the generation of active transforming growth factor-beta.
Tumor cell invasion through basement membranes and into stromal tissue are key steps for promoting growth and metastasis. Tumor cells express various extracellular-matrix-degrading enzymes such as matrix metalloproteinases (MMPs) to degrade extracellular matrix components to facilitate tumor migration and invasion. Histological and clinical studies suggest a role for MMP-1 (collagenase-1) in malignant melanoma invasion. In this study, we evaluated MMP-1 in regulating malignant phenotypes of human melanoma cells by generating human melanoma cells stably transfected with pro-MMP-1 cDNA. The transfectants expressed the active form of MMP-1 associated with cells and showed enhanced invasive and growth abilities in type I collagen gel. Furthermore, MMP-1 expression promoted anchorage-independent growth, which was inhibited in the presence of type II transforming growth factor (TGF)-beta receptor:Fc fusion protein that scavenges TGF-beta receptors. Finally, we demonstrated that MMP-1 directly generated active TGF-beta from its latent form. Thus, these results suggest that MMP-1 produced from melanoma cells would play a role in tumor progression by both degrading matrix proteins and generating active growth factors such as TGF-beta in vivo. Topics: Animals; Cell Line, Tumor; Collagen Type I; Epithelial Cells; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Humans; Lung; Matrix Metalloproteinase 1; Melanoma; Mink; Models, Biological; Neoplasm Metastasis; Transfection; Transforming Growth Factor beta | 2007 |
Transforming growth factor-beta signaling and regulatory T cells.
Topics: Adult; Animals; Cancer Vaccines; CD4-Positive T-Lymphocytes; HLA-DR Antigens; Humans; Interleukin-2 Receptor alpha Subunit; Lipopolysaccharide Receptors; Melanoma; Mice; Myeloid Cells; Transforming Growth Factor beta; Treatment Outcome | 2007 |
Influence of melanoma inhibitory activity on transforming growth factor-beta signaling in malignant melanoma.
Melanoma cells escape transforming growth factor-beta (TGFbeta)-mediated growth inhibition by expressing the Smad (mothers against decapentaplegic homolog, Drosophila) inhibitors Ski and Sno. Here, we demonstrate that melanoma inhibitory activity (MIA) influences the expression of these inhibitors. A Smad responsive reporter construct was activated after TGFbeta1 treatment in the MIA-deficient cell clones but not in the parental cell line. According to this finding, the TGFbeta target genes JunB and Id-1 showed a strong induction of expression. Additional analyses revealed that Ski and Sno, repressors of TGFbeta/SMAD signaling, are not expressed in the MIA-deficient cells but in the parental cell line HMB2 and the mock control. Further investigation showed that Ski and Sno expression might be regulated via the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling cascade. Treatment of HMB2 cells with a MEK inhibitor revealed a reduction of Ski and Sno expression, which leads to the conclusion that, in our melanoma cell model, Ski and Sno expression is regulated via MAPK/ERK signaling. Topics: Blotting, Western; Cell Proliferation; DNA-Binding Proteins; Extracellular Matrix Proteins; Gene Expression Regulation, Neoplastic; Humans; Intracellular Signaling Peptides and Proteins; Melanoma; Mitogen-Activated Protein Kinases; Neoplasm Proteins; Proto-Oncogene Proteins; RNA, Messenger; Signal Transduction; Skin Neoplasms; Smad Proteins; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation | 2006 |
A mathematical model for the regulation of tumor dormancy based on enzyme kinetics.
In this paper we present a two-compartment model for tumor dormancy based on an idea of Zetter [1998, Ann. Rev. Med. 49, 407-422] to wit: The vascularization of a secondary (daughter) tumor can be suppressed by an inhibitor originating from a larger primary (mother) tumor. We apply this idea at the avascular level to develop a model for the remote suppression of secondary avascular tumors via the secretion of primary avascular tumor inhibitors. The model gives good agreement with the observations of [De Giorgi et al., 2003, Derm. Surgery 29, 664-667]. These authors reported on the emergence of a polypoid melanoma at a site remote from a primary polypoid melanoma after excision of the latter. The authors observed no recurrence of the melanoma at the primary site, but did observe secondary tumors at secondary sites 5-7 cm from the primary site within a period of 1 month after the excision of the primary site. We attempt to provide a reasonable biochemical/cell biological model for this phenomenon. We show that when the tumors are sufficiently remote, the primary tumor will not influence the secondary tumor while, if they are too close together, the primary tumor can effectively prevent the growth of the secondary tumor, even after it is removed. It should be possible to use the model as the basis for a testable hypothesis. Topics: Algorithms; Cell Movement; Cell Proliferation; Computer Simulation; Enzymes; Fibrinolysin; Fibroblast Growth Factor 2; Humans; Kinetics; Melanoma; Models, Biological; Neoplasm Metastasis; Neoplasms; Plasminogen; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator | 2006 |
Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-beta-mediated suppressive activity on T lymphocytes.
Human tumors constitutively release endosome-derived microvesicles, transporting a broad array of biologically active molecules with potential modulatory effects on different immune cells. Here, we report the first evidence that tumor-released microvesicles alter myeloid cell function by impairing monocyte differentiation into dendritic cells and promoting the generation of a myeloid immunosuppressive cell subset. CD14+ monocytes isolated from healthy donors and differentiated with interleukin (IL)-4 and granulocyte macrophage colony-stimulating factor in the presence of tumor-derived microvesicles turned into HLA-DR(-/low) cells, retaining CD14 expression and failing to up-regulate costimulatory molecules, such as CD80 and CD86. These phenotypic changes were paralleled by a significant release of different cytokines, including IL-6, tumor necrosis factor-alpha, and transforming growth factor-beta (TGF-beta), and a dose-dependent suppressive activity on activated T-cell-proliferation and cytolytic functions, which could be reversed by anti-TGF-beta-neutralizing antibodies. Microvesicles isolated from plasma of advanced melanoma patients, but not from healthy donors, mediated comparable effects on CD14+ monocytes, skewing their differentiation toward CD14+HLA-DR-/low cells with TGF-beta-mediated suppressive activity on T-cell-functions. Interestingly, a subset of TGF-beta-secreting CD14+HLA-DR- cells mediating suppressive activity on T lymphocytes was found to be significantly expanded in peripheral blood of melanoma patients compared with healthy donors. These data suggest the development in cancer patients of an immunosuppressive circuit by which tumors promote the generation of suppressive myeloid cells through the release of circulating microvesicles and without the need for cell-to-cell contact. Therapeutic interventions on the crucial steps of this pathway may contribute to restore tumor/immune system interactions favoring T-cell-mediated control of tumor growth in cancer patients. Topics: Apoptosis; Cell Differentiation; Cell Line, Tumor; Colorectal Neoplasms; Dendritic Cells; Endosomes; HLA-DR Antigens; Humans; Leukocytes, Mononuclear; Lipopolysaccharide Receptors; Melanoma; Myeloid Cells; Secretory Vesicles; T-Lymphocytes; Transforming Growth Factor beta | 2006 |
Interferon-alpha and transforming growth factor-beta co-induce growth inhibition of human tumor cells.
A hallmark of resistance to type I interferons (IFNs) is the lack of antiproliferative responses. We show here that costimulation with IFN-alpha and transforming growth factor beta-1 (TGF-beta) potentiates antiproliferative activity in a sensitive (ME15) and resistant (D10) human melanoma cell line. A DNA microarray-based search for proliferation control genes involved that are cooperatively activated by IFN-alpha and TGF-beta, yielded 28 genes. Among these are the insulin-like growth factor-binding protein 3 (IGFBP3) and the calcium-binding protein S100A2; we demonstrate, that recombinant IGFBP3 protein is a potent growth inhibitor requiring TGF-beta activity. The antiproliferative activity of S100A2 is significantly enhanced by IFN-alpha in stably transfected ME15 or D10 cell lines. We show for the first time that IFN-alpha is a potent inducer of intracellular calcium release required for activation of S100A2. Our study provides a functional link between IFN-alpha and TGF-beta signaling and extends the function of IFN signaling to calcium-sensitive processes. Topics: Antineoplastic Agents; Cell Line, Tumor; Cell Proliferation; Chemotactic Factors; Drug Resistance, Neoplasm; Gene Expression Profiling; Gene Expression Regulation; Genomics; Humans; Insulin-Like Growth Factor Binding Protein 3; Interferon-alpha; Melanoma; Oligonucleotide Array Sequence Analysis; S100 Proteins; Signal Transduction; Transforming Growth Factor beta | 2006 |
Circulating serum levels of angiogenic factors and vascular endothelial growth factor receptors 1 and 2 in melanoma patients.
Angiogenesis is essential for tumor progression and metastasis; however, the angiogenesis regulators that are biologically relevant for melanoma are still unknown. In this study, we analyzed the circulating serum levels of potent angiogenic factors, including vascular endothelial growth factor (VEGF), angiogenin, transforming growth factor-beta1 and VEGF receptors, VEGFR1 and VEGFR2, in human melanoma patients. One hundred and fourteen patients with histopathologically verified cutaneous melanoma at different stages and 30 healthy controls were investigated. Serum levels of angiogenic factors and VEGF receptors were quantitatively analyzed by solid-phase enzyme-linked immunosorbent assay. The age of the patients (61 men and 53 women) ranged from 18 to 80 years; median age was 51 years. Serum transforming growth factor-beta1 (P < 0.001), VEGF (P = 0.006) and VEGFR1 (P = 0.007) levels were significantly higher in patients with melanoma than in the control group. No significant differences, however, exist in the serum angiogenin and VEGFR2 levels between melanoma patients and the controls. The positive correlations of elevated serum levels of transforming growth factor-beta1, VEGF and VEGFR1 with advanced stages of disease were found. Significant relationship was found only between serum levels of VEGF and VEGFR2. Elevated serum transforming growth factor-beta1 (P < 0.001) and VEGF levels (P = 0.0012) were found to be poor prognostic factors. Serum level of angiogenin and VEGF receptors, however, had no effect on survival. Our data suggest that the angiogenic serum factors, including VEGF, transforming growth factor-beta1 and VEGFR1, but not angiogenin and VEGFR2 were increased in melanoma patients, especially associated with advanced disease stages. The mechanism of VEGF regulation of angiogenesis may in part be due to enhanced proliferation of VEGFRs, especially VEGFR1. Topics: Adolescent; Adult; Aged; Aged, 80 and over; Angiogenesis Inducing Agents; Cell Proliferation; Female; Humans; Male; Melanoma; Middle Aged; Ribonuclease, Pancreatic; Transforming Growth Factor beta; Treatment Outcome; Vascular Endothelial Growth Factor Receptor-1; Vascular Endothelial Growth Factor Receptor-2 | 2006 |
Melanoma pathogenesis and Nodal: a partial picture?
Topics: Humans; Melanoma; Nodal Protein; Transforming Growth Factor beta | 2006 |
FGF-2 blocks TGF-beta1-mediated suppression of Bcl-2 in normal melanocytes.
Normal melanocytes require growth support provided by the adjacent basement membrane. In contrast, nevus cells and melanoma cells survive in the dermis, and in vitro on a soft collagen gel. Transforming growth factor-beta1 (TGF-beta1) produced by melanocytes themselves induces apoptosis in normal melanocytes cultured on collagen gel, an effect that can be counteracted by fibroblast growth factor-2 (FGF-2). The purpose of this study was to investigate the mechanisms by which FGF-2 counteracts the apoptotic signals from TGF-beta1 in melanocytes cultured on collagen gel. We report that FGF-2 did not interfere with the signal transduction from the TGF-beta1 receptors to SMAD2/3 proteins. Instead, TGF-beta1 decreased the level of Bcl-2 in normal melanocytes cultured on collagen gel, and FGF-2 reversed the TGF-beta1-mediated reduction in the level of Bcl-2. In nevus and melanoma cells, TGF-beta1 was unable to induce a decrease in the level of Bcl-2, and treatment with FGF-2 did not cause an increase in the level of Bcl-2 in nevus or melanoma cells. In conclusion, our results suggest that a reduction in the level of the anti-apoptotic Bcl-2 is involved in the execution of apoptosis induced by TGF-beta1 in normal melanocytes cultured on collagen gel and that FGF-2 can prevent TGF-beta1 from causing this reduction. Topics: Apoptosis; Cells, Cultured; Collagen; Fibroblast Growth Factor 2; Gels; Humans; Melanocytes; Melanoma; Nevus; Proto-Oncogene Proteins c-bcl-2; Receptors, Transforming Growth Factor beta; Recombinant Proteins; Signal Transduction; Skin Neoplasms; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2005 |
Characterization of Sno expression in malignant melanoma.
Normal cells are controlled by several exocrine factors, whereas tumor cells often lose control by antiproliferative stimuli. It is known that melanoma cells produce transforming growth factor (TGF) beta1, 2 and 3, but do not respond with growth inhibition. Recently, the Smad inhibitor Ski was found to play a role in this process. We originally analyzed Ski expression in nine melanoma cell lines, however, only one cell line (SK Mel 28) was positive. As all nine cell lines were unresponsive to TGF-beta, we continued to search for the responsible mechanism. Sequencing of the TGF-beta-receptor II, known to be mutated in other kinds of cancer, did not reveal any mutation. A family member of Ski, the proto-oncogene Sno was strongly expressed in all melanoma cell lines on RNA and protein level, but not in melanocytes. To confirm functional relevance of this observation, we used stable antisense Sno transfection for the generation of cell clones with reduced Sno expression. These cell clones displayed reduced cell proliferation, indicating participation of Sno in the escape of melanoma cells from TGF-beta dependent growth control. Searching for TGF-beta target genes that are under control of Sno interference, Id1 but not antiproliferative genes p21, p15, p57 and p27 was identified in the cell clones after antisense Sno expression. In summary, constitutive Sno expression was identified as an important mechanism to shut off antiproliferative TGF-beta signaling in malignant melanoma. Topics: Humans; Intracellular Signaling Peptides and Proteins; Melanoma; Proto-Oncogene Mas; Proto-Oncogene Proteins; Signal Transduction; Skin Neoplasms; Transcription Factors; Transforming Growth Factor beta; Tumor Cells, Cultured | 2005 |
TGF-beta 1 attenuates the acquisition and expression of effector function by tumor antigen-specific human memory CD8 T cells.
TGF-beta1 is a potent immunoregulatory cytokine. However, its impact on the generation and effector function of Ag-specific human effector memory CD8 T cells had not been evaluated. Using Ag-specific CD8 T cells derived from melanoma patients immunized with the gp100 melanoma Ag, we demonstrate that the addition of TGF-beta1 to the initial Ag activation cultures attenuated the gain of effector function by Ag-specific memory CD8 T cells while the phenotypic changes associated with activation and differentiation into effector memory were comparable to control cultures. These activated memory CD8 T cells consistently expressed lower mRNA levels for T-bet, suggesting a mechanism for TGF-beta1-mediated suppression of gain of effector function in memory T cells. Moreover, TGF-beta1 induced a modest expression of CCR7 on Ag-activated memory CD8 T cells. TGF-beta1 also suppressed cytokine secretion by Ag-specific effector memory CD8 T cells, as well as melanoma-reactive tumor-infiltrating lymphocytes and CD8 T cell clones. These results demonstrate that TGF-beta1 suppresses not only the acquisition but also expression of effector function on human memory CD8 T cells and tumor-infiltrating lymphocytes reactive against melanoma, suggesting that TGF-beta1-mediated suppression can hinder the therapeutic benefits of vaccination, as well as immunotherapy in cancer patients. Topics: Cell Line, Tumor; Clone Cells; Coculture Techniques; Cytokines; Down-Regulation; Epitopes, T-Lymphocyte; gp100 Melanoma Antigen; Humans; Immunologic Memory; Immunophenotyping; Immunosuppressive Agents; Lymphocyte Activation; Lymphocytes, Tumor-Infiltrating; Melanoma; Membrane Glycoproteins; Neoplasm Proteins; Recombinant Proteins; T-Lymphocyte Subsets; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2005 |
Selective induction of interleukin-8 expression in metastatic melanoma cells by transforming growth factor-beta 1.
Interleukin (IL)-8 and transforming growth factor (TGF)-beta1 are proangiogenic factors overexpressed in advanced human melanoma. We investigated the effects of TGF-beta1 on IL-8 expression in the well-characterized A375 human melanoma system. We demonstrated by enzyme-linked immunoassay and Northern blot analysis that TGF-beta1 selectively induced IL-8 expression, at both protein and mRNA levels, in highly metastatic A375SM cells but not cells of their poorly metastatic parental line A375P. Transient transfection with luciferase reporter gene constructs revealed that TGF-beta1 activated IL-8 promoter activity in A375SM cells but not A375P cells. Studies with progressive 5' deletion constructs and site-specific mutations demonstrated that a construct containing -133 to +44 of the 5'-flanking sequence was necessary and sufficient for maximal TGF-beta1-induced transcription response and that TGF-beta1-induced activation of IL-8 promoter depended on AP-1 (-126 to -120 bp), NF-kappaB (-94 to -71 bp), and C/EBP-like factor NF-IL6 (-94 to -81 bp) in this region. Interestingly, both A375P and A375SM cells expressed type I and type II TGF-beta receptors and TGF-beta1 induced the nuclear translocation of Smad3 protein in both A375P and A375SM cells. Moreover, both A375P and A375SM cells were susceptible to TGF-beta1-induced growth inhibition. Our data thus demonstrated that TGF-beta1 selectively induced IL-8 expression in highly metastatic A375SM melanoma cells. This TGF-beta1-induced IL-8 expression could be an amplification cascade responsible for overexpression of IL-8 in human melanoma and one of potential mechanisms by which TGF-beta1 promotes angiogenesis, growth, and metastasis of human melanoma. Topics: Cell Line, Tumor; Fluorescent Antibody Technique; Humans; Interleukin-8; Melanoma; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2005 |
Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo.
We previously identified constitutive Smad signaling in human melanoma cells despite resistance to transforming growth factor-beta (TGF-beta) control of cell proliferation. This led us to investigate the effect of inhibitory Smad7 overexpression on melanoma cell behavior. Using the highly metastatic cell line, 1205-Lu, we thus generated melanoma cell clones constitutively expressing Smad7, and their mock-transfected counterparts. Stable expression of Smad7 resulted in an inhibition of constitutive Smad2/3 phosphorylation, and in a reduced TGF-beta response of Smad3/Smad4-driven gene transactivation, as measured using transfected Smad3/4-specific reporter gene constructs. Smad7 overexpression, however, did not alter their proliferative capacity and resistance to TGF-beta-driven growth inhibition. On the other hand, expression of Smad7 efficiently reduced the capacity of human melanoma cells to invade Matrigel in Boyden migration chambers, while not affecting their motility and adhesion to collagen and laminin. Gelatin zymography identified reduced MMP-2 and MMP-9 secretion by Smad7-expressing melanoma cells as compared with their control counterparts. Smad7-expressing melanoma cells exhibited a dramatically reduced capacity to form colonies under anchorage-independent culture conditions, and, when injected subcutaneously into nude mice, were largely delayed in their ability to form tumors. These results suggest that TGF-beta production by melanoma cells not only affects the tumor environment but also directly contributes to tumor cell aggressiveness through autocrine activation of Smad signaling. Topics: Animals; Cell Adhesion; Cell Movement; Gene Expression Profiling; Humans; Matrix Metalloproteinase 9; Melanoma; Mice; Mice, Nude; Neoplasm Invasiveness; Neoplasm Metastasis; Signal Transduction; Skin Neoplasms; Smad7 Protein; Transforming Growth Factor beta; Tumor Cells, Cultured | 2005 |
Myeloma cells suppress bone formation by secreting a soluble Wnt inhibitor, sFRP-2.
Multiple myeloma (MM) develops devastating bone destruction with enhanced bone resorption and suppressed bone formation. In contrast to enhanced osteoclastogenesis, little is known about the mechanism of impaired bone formation in MM. Because a canonical Wingless-type (Wnt) signaling pathway has recently been shown to play an important role in osteoblast differentiation, we examined whether MM cells affect a canonical Wnt pathway to suppress bone formation. Conditioned media from RPMI8226 and U266 MM cell lines and primary MM cells suppressed in vitro mineralization as well as alkaline phosphatase activity in osteoblasts induced by bone morphogenetic protein 2 (BMP-2). These cell lines constitutively produced a soluble Wnt inhibitor, secreted Frizzled-related protein 2 (sFRP-2), but not other Wnt inhibitors including sFRP-1, sFRP-3, and dickkopf 1 (DKK-1) at the protein level. Most MM cells from patients with advanced bone destructive lesions also expressed sFRP-2. Furthermore, exogenous sFRP-2 suppressed osteoblast differentiation induced by BMP-2, and immunodepletion of sFRP-2 significantly restored mineralized nodule formation in vitro, suggesting a predominant role for MM cell-derived sFRP-2 in the impairment of bone formation by MM. Thus, in addition to enhanced osteolysis, MM cells also suppress bone formation at least in part through an inhibition of the canonical Wnt pathway by secreting sFRP-2. Topics: Animals; Antibodies; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Calcification, Physiologic; Cell Differentiation; Genes, Reporter; Humans; Melanoma; Membrane Proteins; Mice; Osteoblasts; Osteogenesis; Solubility; Transforming Growth Factor beta; Tumor Cells, Cultured; Wnt Proteins | 2005 |
A potential role for TGFbeta in the regulation of uveal melanoma adhesive interactions with the hepatic endothelium.
TGFbeta has been shown to have a regulatory effect on uveal melanoma invasion, but it is not known which processes are specifically influenced. The purpose of this study was to analyze the effect of TGFbeta stimulation on the adhesive interactions of uveal melanomas with the extracellular matrix (ECM) and endothelium and, in addition, its effect on the secretion of collagenases.. Invasive and a noninvasive uveal melanoma cell lines, supported by short-term primary uveal melanoma cultures, were used to assess the effect of TGFbeta on ECM and endothelial adhesion and degradation of the ECM. Changes in cell adhesion molecule expression were assessed by flow cytometry, and conditioned media were analyzed by gelatin zymography. Assays of adhesion to ECM substrates and endothelial cells were also performed.. Treatment with TGFbeta increased low basal levels of adhesion molecule and latent MMP-2 expression, as well as adhesion to hepatic endothelial cells by the noninvasive cell line. Conversely, TGFbeta reduced adhesion to laminin and a laminin-binding integrin by invasive cells but had no effect on their adhesion to the endothelium.. In this preliminary study, TGFbeta was found to upregulate levels of MMP-2, reduce adhesion to laminin, and downregulate expression of laminin-binding integrins. Specifically, TGFbeta was found to increase adhesion of noninvasive uveal melanoma cells to the hepatic, but not the dermal, endothelium and may therefore contribute to the preferential targeting of the liver by uveal melanomas. Topics: Activin Receptors, Type I; Adult; Cell Adhesion; Cell Adhesion Molecules; Endothelium, Vascular; Extracellular Matrix; Flow Cytometry; Humans; Immunohistochemistry; Laminin; Liver; Matrix Metalloproteinase 2; Melanoma; Protein Serine-Threonine Kinases; Proteins; Receptor, Transforming Growth Factor-beta Type I; Receptors, Transforming Growth Factor beta; Skin; Transforming Growth Factor beta; Transforming Growth Factor beta2; Tumor Cells, Cultured; Up-Regulation; Uveal Neoplasms | 2005 |
Normal human fibroblasts enable melanoma cells to induce angiogenesis in type I collagen.
We previously reported that fibroblasts induce human microvascular endothelial cells (HMVECs) to differentiate from monolayer to capillarylike morphology. We now test the hypothesis that fibroblasts modulate angiogenesis in melanoma cells.. We tested 12 human melanoma lines (2 radial growth phase (RGP), 3 vertical growth phase (VGP), and 7 metastatic (MM)) for ability to induce HMVECs to invade/migrate into collagen and form capillarylike networks. HMVEC monolayers were overlaid with 3-dimensional collagen gels embedded with melanoma cells alone (M), fibroblasts alone (F), or a 1:1 mixture of the 2 cells (M+F). After 5 days, gels were removed, fixed, and HMVEC networks were quantified by von Willebrand's factor (vWF) immunofluorescence. The influence of soluble factors on HMVEC invasion/migration into collagen was assessed with the use of acellular 3-D collagen gels overlaid on HMVEC monolayers, cultured with conditioned media (CM) derived from monolayers of M, F, or M+F. Angiogenic growth factors involved in the observed invasion/migration were identified with the use of a RayBio Cytokine Antibody Array (RayBiotech, Norcross, Ga).. Cell line-specific variability in melanoma-supported angiogenesis was observed only when in combination with fibroblasts (analysis of variance [ANOVA], P < .01). Melanoma plus fibroblasts uniformly resulted in a significantly higher angiogenic response than melanoma alone (P < .05). One vertical growth phase and one metastatic melanoma line, while weakly angiogenic alone, induced significantly higher angiogenesis than either fibroblast or melanoma alone (P < .05) when combined with fibroblasts. CM from M or M+F induced significantly less HMVEC invasion/migration into collagen than CM from fibroblasts alone. Interleukin 8, monocyte chemotactic protein-1, and tissue inhibitor of metalloproteinase-2 were identified as significantly elevated in the media derived from M+F cultures, compared with either cell type alone.. To our knowledge, this is the first report demonstrating that melanoma-supported angiogenesis in collagen is more significantly influenced by normal skin-derived fibroblasts than by the intrinsic biology of the melanoma cell type. Interleukin 8, monocyte chemotactic protein-1, and tissue inhibitor of metalloproteinase-2 are implicated as potential paracrine factors regulating this observed effect. Topics: Angiogenic Proteins; Becaplermin; Cell Line, Tumor; Cells, Cultured; Collagen; Culture Media, Conditioned; Endothelium, Vascular; Epidermal Growth Factor; Extracellular Matrix; Fibroblasts; Humans; Melanoma; Neovascularization, Pathologic; Neovascularization, Physiologic; Platelet-Derived Growth Factor; Proto-Oncogene Proteins c-sis; Transforming Growth Factor beta; Umbilical Veins | 2005 |
Stromal cell-derived factor-1alpha promotes melanoma cell invasion across basement membranes involving stimulation of membrane-type 1 matrix metalloproteinase and Rho GTPase activities.
Tissue invasion by tumor cells involves their migration across basement membranes through activation of extracellular matrix degradation and cell motility mechanisms. Chemokines binding to their receptors provide chemotactic cues guiding cells to specific tissues and organs; they therefore could potentially participate in tumor cell dissemination. Melanoma cells express CXCR4, the receptor for the chemokine stromal cell-derived factor-1alpha (SDF-1alpha). Using Matrigel as a model, we show that SDF-1alpha promotes invasion of melanoma cells across basement membranes. Stimulation of membrane-type 1 matrix metalloproteinase (MT1-MMP) activity by SDF-1alpha was necessary for invasion, involving at least up-regulation in the expression of this metalloproteinase, as detected in the highly metastatic BLM melanoma cell line. Moreover, SDF-1alpha triggered the activation of the GTPases RhoA, Rac1, and Cdc42 on BLM cells, and expression of dominant-negative forms of RhoA and Rac1, but not Cdc42, substantially impaired the invasion of transfectants in response to SDF-1alpha, as well as the increase in MT1-MMP expression. Furthermore, CXCR4 expression on melanoma cells was notably augmented by transforming growth factor-beta1, a Matrigel component, whereas anti-transforming growth factor-beta antibodies inhibited increases in CXCR4 expression and melanoma cell invasion toward SDF-1alpha. The identification of SDF-1alpha as a potential stimulatory molecule for MT1-MMP as well as for RhoA and Rac1 activities during melanoma cell invasion, associated with an up-regulation in CXCR4 expression by interaction with basement membrane factors, could contribute to better knowledge of mechanisms stimulating melanoma cell dissemination. Topics: Basement Membrane; cdc42 GTP-Binding Protein; Cell Line, Tumor; Chemokine CXCL12; Chemokines, CXC; Enzyme Activation; Humans; Lymphatic Metastasis; Matrix Metalloproteinases, Membrane-Associated; Melanoma; Metalloendopeptidases; Neoplasm Invasiveness; rac1 GTP-Binding Protein; Receptors, CXCR4; rho GTP-Binding Proteins; rhoA GTP-Binding Protein; Transfection; Transforming Growth Factor beta; Transforming Growth Factor beta1; Up-Regulation | 2004 |
Foxp3 expressing CD4+CD25(high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells.
Dominant tolerance is mediated by regulatory T cells (T(reg)) that control harmful autoimmune T cells in the periphery. In this study, we investigate the implication of T(reg) in modulating infiltrating T lymphocytes in human metastatic melanoma. We found that CD4(+)CD25(high) T cells are overrepresented in metastatic lymph nodes (LNs) with a 2-fold increased frequency compared with both tumor-free LNs and autologous PBMCs. These cells express the Foxp3 transcription factor, display an activated phenotype, and display a polyclonal TCR Vbeta chain repertoire. They inhibit in vitro the proliferation and cytokine production of infiltrating CD4(+)CD25(-) and CD8(+) T cells (IL-2, IFN-gamma) through a cell-contact-dependent mechanism, thus behaving as T(reg). In some cases, the presence of T(reg) type 1/Th3-like lymphocytes could also be demonstrated. Thus, T(reg) are a major component of the immunosuppressive microenvironment of metastatic melanoma LNs. This could explain the poor clinical response of cancer patients under immunotherapeutic protocols, and provides a new basis for future immunotherapeutic strategies counteracting in vivo T(reg) to reinforce local antitumor immune responses. Topics: CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cell Division; Cytokines; DNA-Binding Proteins; Forkhead Transcription Factors; Humans; Interleukin-10; Lymphatic Metastasis; Melanoma; Transforming Growth Factor beta | 2004 |
Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals.
Mesenchymal stem cells (MSCs) are largely studied for their potential clinical use. Recently, they have gained further interest after demonstration of an immunosuppressive role. In this study, we investigated whether in vivo injection of MSCs could display side effects related to systemic immunosuppression favoring tumor growth. We first showed in vitro that the murine C3H10T1/2 (C3) MSC line and primary MSCs exhibit immunosuppressive properties in mixed lymphocyte reaction. We demonstrated that this effect is mediated by soluble factors, secreted only on "activation" of MSCs in the presence of splenocytes. Moreover, the immunosuppression is mediated by CD8+ regulatory cells responsible for the inhibition of allogeneic lymphocyte proliferation. We then demonstrated that the C3 MSCs expressing the human bone morphogenetic protein 2 (hBMP-2) differentiation factor were not rejected when implanted in various allogeneic immunocompetent mice and were still able to differentiate into bone. Importantly, using a murine melanoma tumor model, we showed that the subcutaneous injection of B16 melanoma cells led to tumor growth in allogeneic recipients only when MSCs were coinjected. Although the potential side effects of immunosuppression induced by MSCs have to be considered in further clinical studies, the usefulness of MSCs for various therapeutic applications still remains of great interest. Topics: Animals; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; CD8-Positive T-Lymphocytes; Cell Differentiation; Cell Division; Cell Line, Tumor; Humans; Immunosuppression Therapy; Lymphocyte Activation; Lymphocyte Culture Test, Mixed; Melanoma; Mesenchymal Stem Cell Transplantation; Mesenchymal Stem Cells; Mice; Mice, Inbred Strains; Neoplasm Transplantation; Spleen; Transforming Growth Factor beta | 2003 |
Immunologic escape and angiogenesis in human malignant melanoma.
Melanoma escape mechanisms include immunosuppressive and angiogenic cytokine production.. We sought to determine vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) expression by immunohistochemistry, and soluble circulating plasma levels of VEGF, bFGF, IL-10, and transforming growth factor-beta2 in patients with different stages of melanoma.. Biopsy specimens from 42 patients with primary melanoma and 9 with cutaneous metastases were studied by immunohistochemistry. In another 46 patients with melanoma (8 stage I and II; 18, III; and 20, IV) and in 10 healthy control participants, bFGF, VEGF, IL-10, and transforming growth factor-beta2 circulating levels were analyzed.. bFGF was positive in 85% and VEGF in 47.5% of 42 primary melanomas. Of 10 patients with primary melanoma (Breslow depth 1.5-3 mm) 6 were VEGF positive and had metastases develop, whereas 4 were VEGF negative and had no metastases at 5 years of follow up. VEGF, bFGF, and IL-10 plasma levels in patients with stages III and IV melanoma were higher than the control group (P <.05 and P <.01, respectively). An inverse relationship was found between VEGF and IL-10. Specifically, in 7 patients with IL-10 levels higher than 10 pg/mL, VEGF levels were less than 49 pg/mL (P <.05); in 9 patients with VEGF levels higher than 100 pg/mL, IL-10 levels were less than 6.7 pg/mL (P <.01).. VEGF expression in 1.5- to 3.0-mm Breslow depth melanomas may be considered as an unfavorable prognostic factor. Immunosuppressive (IL-10, transforming growth factor-beta2) and proangiogenic (bFGF, VEGF) cytokines are increased in metastatic melanoma. Inverse plasma levels between IL-10 and VEGF in patients with metastatic melanoma are shown in vivo for the first time, the significance of which must be further investigated. Topics: Adult; Aged; Aged, 80 and over; Angiogenesis Inducing Agents; Enzyme-Linked Immunosorbent Assay; Female; Fibroblast Growth Factor 2; Humans; Immunohistochemistry; Interleukin-10; Male; Melanoma; Middle Aged; Skin Neoplasms; Transforming Growth Factor beta; Transforming Growth Factor beta2; Vascular Endothelial Growth Factor A | 2003 |
Transforming growth factor-beta1 inhibits tumor growth in a mouse melanoma model by down-regulating the plasminogen activation system.
The degradation of basement membranes by tumor cells involves secretion and activation of proteinases, such as matrix metalloproteinases (MMPs) and the plasminogen activation system (uPA, tPA, PAI-1), and results from an imbalance between their inhibitors and activators, controlled by various growth factors or cytokines. Among them, the TGF-beta family is one of the most intriguing because it has been reported either to decrease or promote cancer progression. In the present paper, we studied the effect of TGF-beta1 in a mouse melanoma model. In vivo, TGF-beta1 inhibited tumor growth after subcutaneous injection of B16F1 cells in syngenic mice. In vitro, TGF-beta1 did not alter B16F1 cell proliferation, but strongly decreased their migration through Matrigel-coated membranes. The protease production was analyzed by zymography, Western blot, or RT-PCR. MMP-2 and TIMP-2 expression were not altered by TGF-beta1. In contrast, TGF-beta1 triggered a large decrease of uPA and tPA, as well as a decrease of uPA and uPAR mRNAs. By Western blot and RT-PCR analyses, TGF-beta1 was shown to induce a strong increase of PAI-1 synthesis. Collectively, these results suggest that TGF-beta1 may inhibit melanoma tumor growth by specifically decreasing plasmin activity of tumor cells and play a protective role during the earliest stages of tumor progression. Topics: Animals; Cell Division; Cell Line, Tumor; Cell Movement; Disease Models, Animal; Down-Regulation; Female; Fibrinolysin; Matrix Metalloproteinases; Melanoma; Mice; Mice, Inbred C57BL; Neoplasm Metastasis; Plasminogen Activator Inhibitor 1; Plasminogen Activators; Receptors, Cell Surface; Receptors, Urokinase Plasminogen Activator; RNA, Messenger; Skin Neoplasms; Tissue Plasminogen Activator; Transforming Growth Factor beta; Transforming Growth Factor beta1; Urokinase-Type Plasminogen Activator | 2003 |
Cytokine gene single nucleotide polymorphisms and susceptibility to and prognosis in cutaneous malignant melanoma.
Cutaneous malignant melanoma (CMM) is a potentially fatal malignancy in which exposure to UV light is the most important risk factor. Several lines of evidence suggest that CMM patients develop an immune response to their tumours, although, in most cases, anti-tumour immune responses are insufficient to abrogate tumour development. Polymorphism in genes regulating the immune response and cell growth may result in increased susceptibility to and/or poorer prognosis in certain individuals. In this study, we addressed whether single nucleotide polymorphisms (SNPs) associated with differential expression of selected pro- and anti-inflammatory cytokines and growth factors [interleukin (IL)-1beta-35 and -511, IL-2 -330, IL-4 -590, IL-6 -174, IL-8 -251, interferon (IFN)-gamma+874 and transforming growth factor (TGF)beta1 +915] or as markers of candidate cytokine genes (IL-12 +1188) are associated with susceptibility to or known prognostic indicators (e.g. initial tumour growth phase, Breslow thickness, mitotic count in vertical growth phase tumours, tumour regression) in CMM. One hundred and sixty-nine British caucasian CMM patients and 261 controls were included in the study and all SNPs were genotyped by ARMS-PCR. No SNP genotypes or alleles showed significant associations with CMM susceptibility and only the IL-1beta-511 TT genotype was associated with thinner invasive tumours at presentation, as assessed by Breslow thickness at the clinically significant cut-off point of 1.5 mm [occurring in 2/51 (3.9%) thicker vs. 14/78 (17.9%) thinner tumours (P = 0.03; relative risk = 0.29 (95% confidence interval 0.05-0.95)]. These findings suggest that - with the possible exception of IL-1beta- genetic variation associated with differential expression of the selected pro- and anti-inflammatory cytokines is unlikely to play a major role in susceptibility to and prognosis in CMM. Topics: Case-Control Studies; Cytokines; Gene Frequency; Genetic Predisposition to Disease; Humans; Interferon-gamma; Interleukin-1; Interleukin-2; Interleukin-6; Melanoma; Polymorphism, Single Nucleotide; Prognosis; Skin Neoplasms; Transforming Growth Factor beta | 2003 |
Effect of transforming growth factor-beta1, insulin-like growth factor-I, and hepatocyte growth factor on proteoglycan production and regulation in canine melanoma cell lines.
To identify extracellular proteoglycans produced by canine melanoma cell lines and analyze the effect of transforming growth factor-beta1 (TGF-beta1), insulin-like growth factor-I (IGF-I), and hepatocyte growth factor (HGF) on these proteoglycans.. 3 canine melanoma cell lines (ie, CML-1, CML-6M, and CML-10c2).. Extracellular proteoglycans were analyzed by use of metabolic labeling and western immunoblot analysis. The effect of TGF-beta1 on cell proliferation was determined by incorporation of 5-bromo-2'-deoxyuridine.. The CML-1 and CML-6M melanoma cell lines produced 2 main extracellular proteoglycans. One of them was identified as versican, a proteoglycan found in undifferentiated human melanoma cell lines. The CML-10c2 cells produced a small amount of extracellular proteoglycans. Addition of TGF-beta1 (1.25 to 6.25 ng/ml) increased the release of sulfated proteoglycans into the medium. The TGF-beta1 had mainly a posttranslational effect, because it increased the molecular mass of the sulfated bands. Addition of IGF-I (50 ng/ml) slightly increased production of proteoglycans in the CML-6M cell line, whereas HGF (50 ng/ml) did not have any effect on proteoglycan production.. The proteoglycan content and response toTGF-beta1 treatment for CML-1 and CML-6M canine melanoma cell lines are similar to that for undifferentiated human melanoma cell lines. In contrast, CML-10c2 cells produced a low amount of proteoglycans with high molecular weight. Because these extracellular proteoglycans are involved in the control of cell adhesion, proliferation, and migration, they may play an important role in the progression of melanomas in dogs. Topics: Animals; Blotting, Western; Cell Division; Chondroitin Sulfate Proteoglycans; Dog Diseases; Dogs; Hepatocyte Growth Factor; Insulin-Like Growth Factor I; Lectins, C-Type; Melanoma; Proteoglycans; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Versicans | 2002 |
Mutational analysis of selected genes in the TGFbeta, Wnt, pRb, and p53 pathways in primary uveal melanoma.
It is known that the pRb pathway cell-cycle inhibitor p16(INK4A) plays a significant role in cutaneous melanoma and that alteration of p16(INK4A), which resides within the 9p21-22 locus that also contains p15(INK4B) and p14(ARF), may occur in up to one third of uveal melanomas. The absence of TGFbeta responsiveness noted in cultured uveal melanoma cells also suggests that the TGFbeta pathway plays a role in the formation of this tumor. Therefore, mutational screening was performed in several key genes in tumor-suppressor pathways that are known to be altered in some uveal melanomas.. Using denaturing high-performance liquid chromatography (DHPLC) analysis and DNA sequencing, a series of 67 uveal melanomas were screened for inactivating mutations in the TGFbeta pathway members Smad4 and TGFbeta receptor type 2 (TGFbetaR2), the downstream cell-cycle inhibitor p15(INK4B), and the cell-cycle inhibitors p14(ARF) and p16(INK4A). p16(INK4A) was also investigated for promoter hypermethylation. Mutational analysis was also performed on the Wnt pathway gene beta-catenin, known to be mutated in approximately one quarter of cutaneous melanoma cell lines.. Polymorphisms in p16(INK4A) were detected in 3 of 50 samples, but no inactivating mutations were detected in any of the genes screened. Promoter hypermethylation of p16(INK4A) was detected in 5 of 55 tumors, and loss of heterozygosity of the p16(INK4A) locus was detected in 5 of 16 tumors.. Most primary uveal melanomas do not appear to contain somatic mutations in Smad4, TGFbetaR2, p14(ARF), p15(INK4B), p16(INK4A), or beta-catenin. However, methylation of the p16(INK4A) promoter and loss of heterozygosity of the p14(ARF)-p16(INK4A) locus occurs in some tumors. Topics: Cell Cycle Proteins; Chromatography, High Pressure Liquid; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p16; DNA Methylation; DNA Mutational Analysis; DNA, Neoplasm; Female; Humans; Loss of Heterozygosity; Male; Melanoma; Middle Aged; Mutation; Neoplasm Proteins; Polymerase Chain Reaction; Polymorphism, Genetic; Proto-Oncogene Proteins; Signal Transduction; Transforming Growth Factor beta; Tumor Suppressor Protein p14ARF; Tumor Suppressor Protein p53; Tumor Suppressor Proteins; Uveal Neoplasms; Wnt Proteins; Zebrafish Proteins | 2002 |
Stimulation and inhibition of uveal melanoma invasion by HGF, GRO, IL-1alpha and TGF-beta.
To investigate potential factors involved in uveal melanoma migration and invasion in vitro.. Using a microchemotaxis chamber, the effects were studied of a range of stimulators and inhibitors on a series of 10 primary uveal melanomas and 2 uveal melanoma cell lines, by assessing invasion through an 8- micro m pore membrane, precoated with an extracellular matrix solution. In addition, invasion in response to the effect of cells and conditioned media derived from the liver and other tissues was studied for one uveal melanoma culture, by using double-chambered wells, and invasion was assessed through an 8- micro m pore membrane, precoated with synthetic extracellular matrix. In all instances, invading cells were counted under x400 magnification on the lower surface of the membrane. Levels of invasion were correlated with histopathologic markers of prognosis.. Conditioned media and cells derived from other tissues, including the liver, increased cellular invasion of the uveal melanoma cell line studied. For specific regulators, maximum stimulation of invasion was induced by hepatic growth factor (HGF), growth-related oncogene (GRO), and macrophage inflammatory protein (MIP)-1beta, whereas significant inhibition was induced by IL-1alpha, TGF-beta1, and TGF-beta2.. The primary site of metastasis in patients with uveal melanoma is the liver. For the degree of site specificity commonly seen, regulators involved in the process may be expressed at the secondary sites, promoting adhesion, migration, invasion, and proliferation of tumor cells. HGF, GRO, MIP-1beta, IL-1alpha, TGF-beta1, and TGF-beta2 may play a significant role in regulating invasion of uveal melanoma cells. Topics: Aged; Aged, 80 and over; Chemokine CCL4; Chemokine CXCL1; Chemokines; Chemokines, CXC; Chemotactic Factors; Female; Hepatocyte Growth Factor; Humans; Intercellular Signaling Peptides and Proteins; Interleukin-1; Macrophage Inflammatory Proteins; Male; Melanoma; Middle Aged; Neoplasm Invasiveness; Transforming Growth Factor beta; Tumor Cells, Cultured; Uveal Neoplasms | 2002 |
Human melanomas of fibroblast and epithelial morphology differ widely in their ability to synthesize retinyl esters.
Reduced retinyl ester synthesis has been associated with several forms of cancer; we therefore proposed studying melanoma development from the perspective of this biochemical pathway. Cultures of human melanoma cells with fibroblastoid morphology showed negligible retinyl ester synthesis; in sharp contrast, those with epithelioid morphology were capable of retinol esterification. Further, isolated proliferating epidermal melanocytes (HFSC/2) esterified retinol, whereas proliferating normal skin fibroblasts (F:CCD-1121.Sk) did not. A primary site cutaneous melanoma and its metastatic match (both of epithelioid morphology) were capable of retinol esterification, while a matched fibroblastoid tumor pair did not synthesize retinyl esters; nevertheless, LRAT (lecithin:retinol acyltransferase) protein was found in microsomal fractions from all four tumors. A mutation screen in the LRAT coding region and adjacent intronic sequences revealed several novel mutations in these melanomas as well as in HFSC/2 and F:CCD-1121.Sk cells: a single nucleotide polymorphism in exon 1(37A-->G), a silent mutation in exon 2a (188 A-->G/186 G-->A), and an insertion in the 5'UTR (9-10insC). CRBP-1 basal expression was present in the HFSC/2, and in both sets of matched tumor pairs; however, steady-state levels in the fibroblastoid melanoma pair were one-third that found in the epithelioid matched tumor pair. Co-culture of human primary site epithelioid melanoma with proliferating normal human skin fibroblasts abrogated retinol esterification within 96 h and increased the expression of the active form of TGFbeta-1 by 2.4-fold. A concomitant 3.2-fold downregulation of CRBP-1 expression took place. This is the first study to (1) demonstrate an association between retinyl ester synthesis and cutaneous melanoma morphological phenotypes; (2) suggest the existence of a soluble, diffusible inhibitor of the retinol esterification pathway; (3) report the ability of the isolated, proliferating human epidermal melanocyte to esterify retinol; and (4) provide evidence of DNA variants in the coding region of LRAT. Topics: Acyltransferases; Biological Factors; Cells, Cultured; Diffusion; DNA Mutational Analysis; Epithelial Cells; Esterification; Esters; Fibroblasts; Gene Expression Regulation, Neoplastic; Genes; Humans; Introns; Melanoma; Microsomes; Mutagenesis, Insertional; Neoplasm Metastasis; Neoplasm Proteins; Neoplastic Stem Cells; Polymorphism, Single Nucleotide; Retinol-Binding Proteins; Retinol-Binding Proteins, Cellular; Skin Neoplasms; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Vitamin A | 2002 |
TGF-beta in uveal melanoma.
Uveal melanoma is the most common primary ocular cancer among adults and patients with distant metastases seldom survive longer than a year. Melanomas of the eye have the advantage of growing in the special environment of an immune privileged site and it has long been shown, that the special immunosuppressive properties of the intraocular microenvironment are strongly mediated by cytokines, especially transforming growth factor-beta (TGF-beta). Here, we sought to investigate the presence of TGF-beta in surgically removed uveal melanoma specimens using immunohistochemical methods to verify possible autocrine mechanisms. Immunocytochemistry for pan-TGF-beta and TGF-beta(2) was performed on 13 melanoma specimens using an alkaline phosphatase labeling procedure. Melanocytic origin of the tumors was confirmed by HMB-45 staining. All tissue samples exhibited positive staining using either pan-TGF-beta or TGF-beta(2) antibody regardless of cell-type, size of the tumor, or tumor location. The intensity of staining did not vary significantly within a given tumor. All tumors stained positive against the HMB-45 antibody. Many cytokines have been found to act on melanoma tumors. The presence of the TGF-beta(2) isoform in all specimens points to progressive tumor-growth as has been shown for melanomas of the skin. Based on our immunohistochemical findings and the immunosuppressive properties of TGF-beta, we suppose that ocular melanomas should be able to create their own immunosuppressive environment even in the uvea, which might be a non-privileged site. Topics: Aged; Aged, 80 and over; Female; Humans; Immune Tolerance; Immunohistochemistry; Male; Melanoma; Middle Aged; Transforming Growth Factor beta; Uveal Neoplasms | 2001 |
Comparative analysis of immunocritical melanoma markers in the mouse melanoma cell lines B16, K1735 and S91-M3.
The mouse melanoma cell lines B16, K1735 and Cloudman S91-M3 (and various sublines) are frequently used as melanoma models. Extensive comparative data of their immunological features are not available. In order to define the immunological profiles of these cell lines, relevant tumour markers were studied. S91-M3 melanoma cells constitutively expressed high levels of major histocompatibility complex (MHC) I, in contrast to K1735-M2 and B16-F1 cells. MHC II expression was restricted to B16-F1 cells following interferon-gamma treatment. Tyrosinase, tyrosinase-related protein-2 and gp100 were detected in B16-F1 and S91-M3 cells, but not in K1735-M2 cells. Constitutive surface expression and secretion of intercellular adhesion molecule-1 was found on S91-M3 cells. No substantial secretion of interleukin-10 could be detected. In contrast, low levels of latent transforming growth factor-beta were found in the cell supernatants of B16-F1 and K1735-M2 cells. The expression pattern of Fas, FasL and FLICE inhibitory protein was comparable in all three cell lines. Thus our findings indicate that each cell line presents a characteristic immunological profile, confirming that B16-F1 is an appropriate murine tumour model for tumours with low levels of MHC I but expressing melanoma-associated antigens. S91-M3 represents a complementary, more immunogenic model. In contrast, K1735-M2 does not seem to be an appropriate model for melanoma. Topics: Animals; Bacterial Proteins; Biological Assay; Biomarkers, Tumor; Blotting, Northern; Blotting, Western; Carrier Proteins; CASP8 and FADD-Like Apoptosis Regulating Protein; Enzyme-Linked Immunosorbent Assay; Fas Ligand Protein; fas Receptor; Flow Cytometry; Genes, MHC Class I; Genes, MHC Class II; gp100 Melanoma Antigen; Intercellular Adhesion Molecule-1; Interferon-gamma; Interleukin-10; Intracellular Signaling Peptides and Proteins; Intramolecular Oxidoreductases; Melanoma; Melanoma, Experimental; Membrane Glycoproteins; Mice; Monophenol Monooxygenase; Neoplasm Proteins; Transforming Growth Factor beta; Tumor Cells, Cultured | 2001 |
Is there a relationship between tissue inhibitor of metalloproteinase-1 and transforming growth factor-beta 1 with respect to malignant melanoma progression?
Recent articles about the roles and relationships of tissue inhibitor of metalloproteinase-1 and transforming growth factor-beta 1 in various types of normal tissues and malignancies give rise to the question: 'Is there a relationship between them with regard to malignant melanoma progression?' In the light of many references, it seems to be highly probable that the tissue inhibitor of metalloproteinase-1, -- being a multifunctional protein -- functions as a growth factor with possible stimulation by transforming growth factor-beta 1 in progression of malignant melanoma, rather than its other existing functions in many different normal and cancer tissues (e.g. inhibition of the matrix metalloproteinases or functioning as an insignificant inhibitor of angiogenesis). Topics: Cell Division; Disease Progression; Humans; Melanoma; Neoplasm Invasiveness; Skin Neoplasms; Tissue Inhibitor of Metalloproteinase-1; Transforming Growth Factor beta | 2001 |
Cytoplasmic localization of the oncogenic protein Ski in human cutaneous melanomas in vivo: functional implications for transforming growth factor beta signaling.
The oncogenic protein Ski associates with Smad proteins and counteracts their activation of gene expression and growth inhibition in response to transforming growth factor beta (TGF-beta). Here we show that Ski protein levels are increased in all 44 human melanoma tumor tissues analyzed in vivo. In addition, Ski subcellular localization changes from nuclear, in preinvasive melanomas (melanomas in situ), to nuclear and cytoplasmic in primary invasive and metastatic melanomas. Furthermore, Ski/Smad association in the cytoplasm seems to prevent Smad3 nuclear translocation in response to TGF-beta. The biological significance of Ski overexpression in melanomas was established by showing that down-regulation of Ski levels, by antisense Ski vectors, restored TGF-beta-mediated growth inhibition. Such inhibition is apparently mediated by up-regulation of the cyclin-dependent kinase-I p21(Waf-1) and inhibition of cyclin-dependent kinase 2 activity. Our results suggest that high levels of Ski in human melanomas produce a disruption of TGF-beta signaling phenotypically similar to that in cells harboring mutations in TGF-beta receptors or Smad proteins, and this may represent a significant event in the progression of melanomas in vivo. Topics: Cell Division; Cytoplasm; DNA-Binding Proteins; Humans; Melanoma; Proto-Oncogene Proteins; Signal Transduction; Skin Neoplasms; Smad3 Protein; Trans-Activators; Transforming Growth Factor beta; Tumor Cells, Cultured | 2001 |
Transforming growth factor-beta1 increases survival of human melanoma through stroma remodeling.
Transforming growth factor (TGF)-beta is growth inhibitory for normal epithelial cells and melanocytes but can stimulate mesenchymal cells. Resistance to its inhibitory effects is characteristic of human melanoma, the growth of which may instead be promoted by TGF-beta, because its production is increased with melanoma progression. Whether TGF-beta has an autocrine function for melanoma cells or is important for paracrine stimulation of the tumor stroma is not known. In this study, TGF-beta1 was expressed in melanoma cells via adenoviral gene transfer, and tumor growth was analyzed in vitro, in human skin grafts, and in mixtures with fibroblasts that were injected s.c. into immunodeficient mice. The TGF-beta1 produced by the melanoma cells activated the fibroblasts to produce matrix within and around the tumor mass, whereas control tumors showed less stroma and more cell death. High expression of collagen, fibronectin, tenascin, and alpha2 integrin was detected in the TGF-beta1-expressing tumors by immunohistochemistry. Number and size of lung metastases were significantly increased. cDNA expression array analysis of TGF-beta1-transduced fibroblasts embedded in type I collagen and of TGF-beta1-transduced melanoma cells demonstrated induction of types XV, XVIII, and VI collagens, tenascin, plasminogen activator inhibitor-I, vascular endothelial growth factor, cysteine-rich fibroblast growth factor receptor-1, and platelet-derived growth factor receptor-beta, which could be linked to promotion of growth and survival in melanoma. These data suggest that remodeling of the neighboring stroma, which provides a supporting scaffolding and a positive feedback stimulation of tumor growth, is an important function of TGF-beta1 in melanoma. Topics: Animals; Cell Communication; Cell Death; Cell Survival; Extracellular Matrix Proteins; Female; Fibroblasts; Gene Expression; Gene Expression Profiling; Humans; Immunohistochemistry; Male; Melanoma; Mice; Mice, SCID; Stromal Cells; Transduction, Genetic; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2001 |
Human melanoma cells inhibit the earliest differentiation steps of human Langerhans cell precursors but failed to affect the functional maturation of epidermal Langerhans cells.
Tumour-derived factors suppress differentiation and function of in vitro generated DC. Here, we investigate the effect of two melanoma clones differing in their invasive and metastatic properties on the generation and/or functional maturation of human epidermal LC. LC were generated from CD34(+) cord blood progenitors under GM-CSF/TNF-alpha/TGF-beta 1. CD34(+) cells were co-cultured with or without melanoma cells using Transwell dishes. After 11 days of co-culture, CD34(+)-derived cells display a non-adherent undifferentiated morphology, a high level of monocytic CD14 marker, a down-regulated expression of LC markers (CD1a, E-cadherin) and DC markers (CD40, CD80, CD54, CD58, CD83, CD86, HLA-DR, HLA-class I). These cells were less potent than control LC in inducing allogeneic T cell proliferation. The generation of the CD14(+) population was correlated with a decrease in the CD1a(+) population, without any statistical differences between the two clones. Melanoma cells diverted the differentiation of CD34(+) cells towards a dominant CD14(+) population only if the progenitors were in an early growth phase. IL-10, TGF-beta 1 and VEGF were not responsible for these effects, as assessed by using blocking antibodies. By contrast, co-culture of fresh epidermal LC with melanoma cells did not affect their phenotype and function. Our data demonstrate that melanoma cells inhibit the earliest steps of LC differentiation, but failed to affect the functional maturation of epidermal LC. This suggests that melanoma cells participate in their own escape from immunosurveillance by preventing LC generation in the local cutaneous microenvironment. Topics: Antigens, CD34; Cell Communication; Cell Differentiation; Cell Division; Clone Cells; Coculture Techniques; Endothelial Growth Factors; Epidermal Cells; Epidermis; Fetal Blood; Humans; Immunologic Surveillance; Immunophenotyping; Interleukin-10; Langerhans Cells; Lipopolysaccharide Receptors; Lymphokines; Melanoma; Neoplasm Proteins; Neoplastic Stem Cells; Skin Neoplasms; T-Lymphocytes; Transforming Growth Factor beta; Tumor Cells, Cultured; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors | 2001 |
Expression of a decorin-like moleculein human melanoma.
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 |
Ski acts as a co-repressor with Smad2 and Smad3 to regulate the response to type beta transforming growth factor.
The c-ski protooncogene encodes a transcription factor that binds DNA only in association with other proteins. To identify co-binding proteins, we performed a yeast two-hybrid screen. The results of the screen and subsequent co-immunoprecipitation studies identified Smad2 and Smad3, two transcriptional activators that mediate the type beta transforming growth factor (TGF-beta) response, as Ski-interacting proteins. In Ski-transformed cells, all of the Ski protein was found in Smad3-containing complexes that accumulated in the nucleus in the absence of added TGF-beta. DNA binding assays showed that Ski, Smad2, Smad3, and Smad4 form a complex with the Smad/Ski binding element GTCTAGAC (SBE). Ski repressed TGF-beta-induced expression of 3TP-Lux, the natural plasminogen activator inhibitor 1 promoter and of reporter genes driven by the SBE and the related CAGA element. In addition, Ski repressed a TGF-beta-inducible promoter containing AP-1 (TRE) elements activated by a combination of Smads, Fos, and/or Jun proteins. Ski also repressed synergistic activation of promoters by combinations of Smad proteins but failed to repress in the absence of Smad4. Thus, Ski acts in opposition to TGF-beta-induced transcriptional activation by functioning as a Smad-dependent co-repressor. The biological relevance of this transcriptional repression was established by showing that overexpression of Ski abolished TGF-beta-mediated growth inhibition in a prostate-derived epithelial cell line. Topics: Cell Nucleus; DNA-Binding Proteins; Female; Gene Expression Regulation; Gene Expression Regulation, Neoplastic; Genes, Reporter; Humans; Luciferases; Macromolecular Substances; Melanoma; Neoplasm Proteins; Promoter Regions, Genetic; Proto-Oncogene Proteins; Recombinant Fusion Proteins; Repressor Proteins; Smad2 Protein; Smad3 Protein; Trans-Activators; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Two-Hybrid System Techniques | 2000 |
Regulation of the murine silver locus product (gp87) by the hypopigmenting cytokines TGF-beta1 and TNF-alpha.
The melanosomal proteins encoded by the silver (si, SILV, or PMEL17) locus play important roles in melanogenesis and are actively investigated as targets for melanoma immunotherapy. The human silver locus yields two proteins, gp100 and PMEL17, by alternative splicing of a common mRNA precursor. Mouse melanocytes exclusively express the gp100 orthologue, here termed gp87, thus providing a simpler model with which to study the silver locus products. We have analyzed the effects of [Nle4, D-Phe7]-alpha melanocyte-stimulating hormone (alphaMSH) and two hypopigmenting cytokines, tumor necrosis factor (TNF)-alpha and transforming growth factor (TGF)-beta1, on the expression of gp87 in B16 mouse melanoma cells. TNF-alpha and TGF-beta1 (at saturating doses for 48 hr) decreased gp87 mRNA by 50%. The gp87 protein was almost undetectable by Western immunoblotting after TNF-alpha treatment, but was not affected by TGF-beta1. alphaMSH increased the mRNA and the gp87 protein approximately 2-fold. Moreover, the amount of gp87 was not reduced by TNF-alpha in the presence of the hormone, in spite of a 50%, decrease in its mRNA. Therefore, the levels of mRNA and gp87 protein did not correlate after treatment with the cytokines. Overall, our data suggest that the silver locus product is not regulated exclusively at the transcriptional level, and highlight the importance of still-uncharacterized regulatory translational and/or post-translational events. Topics: alpha-MSH; Animals; Blotting, Northern; Carboxypeptidases; Gene Expression; Melanocytes; Melanoma; Membrane Glycoproteins; Mice; Pigments, Biological; Protein Biosynthesis; Proteins; RNA, Messenger; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha | 2000 |
Expression of activated N-ras in a primary melanoma cell line counteracts growth inhibition by transforming growth factor-beta.
One critical factor in melanoma progression is the change from radial growth phase to vertical growth phase. We previously showed a high incidence of ras mutations in progressing but not early human melanomas. We also found that stable expression of activated Ras in a primary human melanoma cell line (WM35) led to enhanced proliferation, anchorage-independent survival, migration and invasion in vitro and enhanced subcutaneous tumor formation in vivo, transforming the melanoma phenotype from the radial growth phase to the vertical growth phase. Inhibitory cytokines, especially transforming growth factor-beta, are important in homeostasis of normal human melanocytes. Proliferation of early melanoma cells can be inhibited by transforming growth factor-beta, whereas more aggressive stages lose this response. Using a transforming growth factor-beta activated luciferase reporter transiently transfected into WM35, WM35N-ras, and WM35H-ras (WM35 transfected with mutant N-ras or H-ras genes), we demonstrated significant decreases (p < 0. 04) in transforming growth factor-beta induced reporter expression in both ras transfected cell lines. Transforming growth factor-beta also induced significant decreases (p < 0.002) in the proportion of WM35 cells in S-phase of the cell cycle; this effect was not observed in WM35N-ras cells. Furthermore, we demonstrated that an important controlling factor in transforming growth factor-beta inhibition of cell cycle progression, the phosphorylation of the Rb protein, was altered in WM35N-ras; transforming growth factor-beta caused a marked relative increase in hypophosphorylated pRb in WM35 cells, but not in WM35N-ras. These data suggest that activated Ras plays an important part in melanoma progression from the radial growth phase to the vertical growth phase by counteracting inhibition by cytokines such as transforming growth factor-beta, thus providing a growth advantage. Topics: Cell Cycle Proteins; Cell Division; Gene Expression; Gene Expression Regulation; Genes, ras; Humans; Melanoma; Mutation; Phosphorylation; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured | 2000 |
Cloning of an isoform of integrin-linked kinase (ILK) that is upregulated in HT-144 melanoma cells following TGF-beta1 stimulation.
We have shown previously that integrin-linked kinase (ILK) is upregulated in human HT-144 melanoma cells following TGF-beta1 stimulation. Using mRNA from TGF-beta1 stimulated HT-144 cells and reverse transcriptase polymerase chain reaction, we have isolated a cDNA encoding a protein highly homologous to ILK. Sequencing of the full-length 1359 base pair cDNA and polypeptide translation revealed that this protein, designated ILK-2, differs from the known ILK (hereafter called ILK-1) by only four amino acids, while the cDNA sequence diverges by 102 nucleotides, thus excluding that ILK-2 is an allelic variant of ILK-1. Expression of ILK-2 mRNA was observed in metastatic human HT-144 melanoma and HT-1080 fibrosarcoma cell lines, but not in normal human tissues. Moreover, stimulation of HT-144 cells with TGF-beta1, but not with EGF, PDGF-AB or insulin, induced a selective overexpression of ILK-2 mRNA as compared to ILK-1 mRNA. Bacterially-expressed GST/ILK-2 autophosphorylated and labeled myelin basic protein as well as a recombinant GST/beta3 integrin cytoplasmic tail peptide. Transfection of either ILK-2 or ILK-1 cDNA into the non-metastatic melanoma cell line SK-Mel-2, expressing exclusively ILK-1, induced anchorage independent cell growth and cell proliferation, as demonstrated by growth in soft agar. Our data provide evidence that ILK-2 is a new isoform of ILK-1 that is expressed in some highly invasive tumor cell lines but not in normal adult human tissues and whose expression is regulated by TGF-beta1. Topics: Amino Acid Sequence; Base Sequence; Blotting, Western; Cell Division; DNA, Complementary; Isoenzymes; Melanoma; Molecular Sequence Data; Neoplasm Proteins; Polymorphism, Restriction Fragment Length; Protein Serine-Threonine Kinases; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation | 2000 |
The alpha 3(IV)185-206 peptide from noncollagenous domain 1 of type IV collagen interacts with a novel binding site on the beta 3 subunit of integrin alpha Vbeta 3 and stimulates focal adhesion kinase and phosphatidylinositol 3-kinase phosphorylation.
We have recently identified integrin alpha(v)beta(3) and the associated CD47/integrin-associated protein (IAP) together with three other proteins as the potential tumor cell receptors for the alpha(3) chain of basement membrane type IV collagen (Shahan, T.A., Ziaie, Z., Pasco, S., Fawzi, A., Bellon, G., Monboisse, J. C., and Kefalides, N. A. (1999) Cancer Res. 59, 4584-4590). Using different cell lines expressing alpha(v)beta(3), alpha(IIb)beta(3), and/or CD47 and a liquid phase receptor capture assay, we now provide direct evidence that the synthetic and biologically active alpha3(IV)185-206 peptide, derived from the alpha3(IV) chain, interacts with the beta(3) subunit of integrin alpha(v)beta(3), independently of CD47. Increased alpha3(IV) peptide binding was observed on transforming growth factor-beta(1)-stimulated HT-144 cells shown to up-regulate alpha(v)beta(3) independently of CD47. Also, incubation of HT-144 melanoma cells in suspension induced de novo exposure of ligand-induced binding site epitopes on the beta(3) subunit similar to those observed following Arg-Gly-Asp-Ser (RGDS) stimulation. However, RGDS did not prevent HT-144 cell attachment and spreading on the alpha3(IV) peptide, suggesting that the alpha3(IV) binding domain on the beta(3) subunit is distinct from the RGD recognition site. alpha3(IV) peptide binding to HT-144 cells in suspension stimulated time-dependent tyrosine phosphorylation, while the RGDS peptide did not. Two major phosphotyrosine proteins of 120-130 and 85 kDa were immunologically identified as focal adhesion kinase and phosphatidylinositol 3-kinase (PI3-kinase). A direct involvement of PI3-kinase in alpha3(IV)-dependent beta(3) integrin signaling could be documented, since pretreatment of HT-144 cells with wortmannin, a PI3-kinase inhibitor, reverted the known inhibitory effect of alpha3(IV) on HT-144 cell proliferation as well as membrane type 1-matrix metalloproteinase gene expression. These results provide evidence that the alpha3(IV)185-206 peptide, by directly interacting with the beta(3) subunit of alpha(v)beta(3), activates a signaling cascade involving focal adhesion kinase and PI3-kinase. Topics: Amino Acid Sequence; Animals; Antigens, CD; Binding Sites; Carrier Proteins; CD47 Antigen; Cell Adhesion; CHO Cells; Collagen; Collagen Type IV; Cricetinae; Enzyme Activation; Focal Adhesion Kinase 1; Focal Adhesion Protein-Tyrosine Kinases; Humans; Integrin beta3; Melanoma; Molecular Sequence Data; Peptide Fragments; Phosphatidylinositol 3-Kinases; Phosphorylation; Platelet Membrane Glycoproteins; Protein-Tyrosine Kinases; Receptors, Vitronectin; Recombinant Proteins; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation | 2000 |
Abnormalities of the transforming growth factor-beta pathway in ocular melanoma.
The majority of ocular melanomas occur in the uveal tract. Chemotherapy is generally ineffective and large tumours requiring enucleation have a greater than 50% mortality at 5 years. Monosomy for chromosome 3 is common in uveal melanoma and it is known that there is loss of responsiveness to transforming growth factor beta (TGFbeta) in melanoma cell lines. Since the gene for TGFbeta receptor II (TGFbetaR2) is located on chromosome 3p22, this study investigates the possibility that the TGFbeta pathway, and TGFbetaR2 in particular, might be involved in the pathogenesis of this rare eye tumour. To this end, the expression of molecules in the pathway has been examined by immunocytochemistry (TGFbeta, TGFbetaR2, SMAD2, SMAD3, SMAD4, and p27), backed up by a cell culture assay of TGFbeta-mediated growth suppression, RT-PCR for SMAD4, and loss of heterozygosity (LOH) on 3p22. There was LOH at 3p22 in 6/19 tumours and loss of TGFbetaR2 expression in 10/27 tumours. Immunohistochemistry for SMADs 2, 3, and 4 showed potential loss of signal transduction in 14/27 tumours. The results indicate abnormality of the TGFbeta pathway in 61% of tumours for which unequivocal results were obtained and suggest that abrogation of control of melanocyte growth by the TGFbeta pathway may be important in the formation of uveal melanoma. Topics: Adult; Aged; Aged, 80 and over; DNA, Neoplasm; Female; Humans; Immunoenzyme Techniques; Loss of Heterozygosity; Male; Melanoma; Microsatellite Repeats; Middle Aged; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured; Uveal Neoplasms | 2000 |
Overexpression of nPKCdelta in BL6 murine melanoma cells enhances TGFbeta1 release into the plasma of metastasized animals.
Transforming growth factor-beta (TGFbeta) contributes to the promotion of invasion, metastasis, angiogenesis and even immunosuppression. Since overexpression of the delta isoform of protein kinase C (nPKCdelta) in BL6 murine melanoma cells (BL6T cells) increases their metastatic capacity, we investigated the possible involvement of TGFbeta1 in this process. Immunohistochemical analysis demonstrated lower levels of TGFbeta1 in BL6T lung metastases compared with BL6 lung metastases. On the other hand, higher levels of this cytokine, in particular in its active form, occur in the plasma of BL6T metastasized animals, suggesting a nPKCdelta-dependent TGFbeta1 release. Therefore, nPKCdelta-dependent TGFbeta1 release and activation may be involved in the greater angiogenic and metastatic capacity of murine melanoma BL6T cells. Topics: Animals; Blotting, Western; Densitometry; Immunohistochemistry; Isoenzymes; Lung Neoplasms; Melanoma; Mice; Mice, Inbred C57BL; Neoplasm Metastasis; Neoplasm Transplantation; Neovascularization, Pathologic; Protein Isoforms; Protein Kinase C; Protein Kinase C-delta; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured | 2000 |
Different levels of TGFbeta, IL-10, IFNgamma and gelatinase A occur in experimental white and black metastases induced by bryostatin 1 or by phorbol ester-treated BL6T murine melanoma cells.
Bryostatin 1 and phorbol esters reduce the intracellular melanin level in high metastatic overexpressing nPKCdelta BL6 (BL6T) cells, thereby inducing white experimental metastasis in syngeneic mice. We evaluate here the possible differences between white and black metastases induced by both treatments on the proliferative and metastatic potential as well as on the expression of some cytokines involved in the metastatic process such as TGFbeta, IL-10 and IFNgamma. The level of expression of gelatinase A is also considered. White and black metastases induced after the injection of bryostatin 1- or phorbol ester-treated cells into the tail vein of syngenic mice were isolated and analysed for the levels of LDH usually used as markers of cytotoxicity, for the levels of cytokines and gelatinase A or dissociated and cultured in vitro for a few passages. The cultured cells were analysed in vitro for the proliferative capacity and the melanin synthesis. The same cells were also re-injected into syngeneic mice and the number of experimental metastases were counted after 17 days or injected with matrigel in order to quantify the proliferative capacity in vivo. The results show only one significant difference between bryostatin I and phorbol ester, namely the cells obtained from white bryostatin 1-treated cells return to a black phenotype after a few passages in culture. This suggests that PKC mediates many of the biological effects of bryostatin 1 but that its effect is lost in vitro. On the other hand, white and black metastases (at least for metastases induced by BL6T cells treated with phorbol ester) do appear significantly different. In vivo white metastases show lower levels of LDH, lower levels of proliferative capacity into matrigel, higher levels of TGFbeta and IFNgamma and, when re-injected into syngeneic mice, give big black metastases. Therefore, in murine melanoma cells, the treatment with bryostatin I induces the appearance of a white population expressing different levels of TGFbeta, IFNgamma, IL-10 and gelatinase A. Such a white population is more difficult to diagnose and is capable of turning into a more aggressive phenotype under suitable environmental conditions. Topics: Animals; Antineoplastic Agents; Bryostatins; Interferon-gamma; Interleukin-10; L-Lactate Dehydrogenase; Lactones; Lung Neoplasms; Macrolides; Matrix Metalloproteinase 2; Melanoma; Mice; Mice, Inbred C57BL; Protein Kinase C; Reverse Transcriptase Polymerase Chain Reaction; Tetradecanoylphorbol Acetate; Transforming Growth Factor beta | 2000 |
Independent regulation of growth and SMAD-mediated transcription by transforming growth factor beta in human melanoma cells.
Increased production of transforming growth factor beta (TGF-beta) coupled with resistance to the growth-inhibitory effects of TGF-beta is characteristic of several types of neoplasia including human melanoma. In select epithelial malignancies, lack of TGF-beta-induced growth inhibition is associated with disruptions of TGF-beta-dependent SMAD signaling and transcription. In contrast, the results of the present study indicate intact SMAD-dependent transcription in human melanoma cells, regardless of their proliferative response to exogenous TGF-beta. Furthermore, in some melanoma cell lines constitutive SMAD-dependent transcription was observed, which was due in part to endogenous TGF-beta. These results establish that resistance of melanoma cells to TGF-beta-induced growth inhibition occurs independently of intact TGF-beta receptor/SMAD-mediated transcriptional regulation. They also suggest that melanoma-derived TGF-beta may exert autocrine effects on SMAD-sensitive target genes. Topics: Cell Division; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Humans; Melanocytes; Melanoma; Signal Transduction; Trans-Activators; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured | 1999 |
Desensitization of melanoma cells to autocrine TGF-beta isoforms.
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 |
Differential expression of transforming growth factor beta 1 and interleukin 10 in progressing and regressing areas of primary melanoma.
The coexistence of tumor specific immunity with a progressing tumor remains a major paradox of tumor immunology. This enigma is most evident in partially regressing melanomas, where efficient eradication of tumor cells is closely linked to uncontrolled tumor growth. Mechanisms involved in this differential susceptibility of tumor cells to the host immune response may include altered production of immunosuppressive cytokines, i.e., transforming growth factor (TGF) beta or interleukin (IL) 10. Since only limited amounts of tissue samples are available from primary tumors, a semi-quantitative reverse transcriptase polymerase chain reaction (RT-PCR) was established which allowed to estimate the amount of cytokine mRNA expressed in a small number of melanoma cells segregated by indirect immunomagnetic isolation. Thereby, we determined the expression of TGF-beta 1 and IL-10 mRNA in melanoma cells obtained from regressing and progressing areas of 9 primary tumors. TGF-beta 1 mRNA could be detected in all undiluted samples from progressing areas and in 7 samples from regression zones. Titration of the sample revealed that in 6 cases TGF-beta 1 mRNA could be detected at a significant higher titer in progressing than in regressing areas. IL-10 mRNA was present in 8 samples obtained from progressing and in 7 samples from regressing tumor areas. In 6 tumors IL-10 mRNA was detectable at a higher titer in the progression zones. Specificity of the PCR amplification was confirmed with a series of restriction enzyme digestions of the resulting PCR product. Based on these findings the hypothesis that immunosuppressive cytokines, such as TGF-beta 1 or IL-10, represent important factors for the melanoma cells to escape immune surveillance is supported. Topics: Disease Progression; Humans; Immune Tolerance; Immunologic Surveillance; Interleukin-10; Melanoma; Neoplasm Regression, Spontaneous; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Skin Neoplasms; Transforming Growth Factor beta | 1999 |
Autocrine TGF-beta-regulated expression of adhesion receptors and integrin-linked kinase in HT-144 melanoma cells correlates with their metastatic phenotype.
We have previously shown that 2 human melanoma cell lines, the metastatic HT-144 and the non-metastatic SK-Mel-2 cells, exhibit marked in vitro heterogeneity with respect to integrin expression, migration and invasion potential. Here, we provide evidence that HT-144 melanoma cells, but not SK-Mel-2 cells, undergo a reversible transition to a fibroblastoid morphology following treatment with either their own serum-free acidified conditioned medium or biologically active exogenous TGF-beta1, thus identifying TGF-beta as an autocrine regulator of the spindle shape morphology of HT-144 melanoma cells. The fibroblastoid phenotype correlated with up-regulated beta1 and beta3 integrin and down-regulated E-cadherin expression, as shown by flow cytometry, Western blot and RT-PCR, as well as up-regulated expression of the matrix metalloproteinase MMP-9, as demonstrated by zymography. Our data further illustrate the TGF-beta1-dependent up-regulation of integrin-linked kinase and the nuclear translocation of beta-catenin, 2 intracellular proteins involved in integrin and cadherin signaling. Topics: Antigens, CD; Blotting, Western; Cadherins; Cell Movement; Cell Size; Collagenases; Down-Regulation; Flow Cytometry; Humans; Integrin beta1; Integrin beta3; Matrix Metalloproteinase 9; Melanoma; Phenotype; Platelet Membrane Glycoproteins; Protein Serine-Threonine Kinases; Reverse Transcriptase Polymerase Chain Reaction; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation | 1999 |
Differential expression of G1 cyclins and cyclin-dependent kinase inhibitors in normal and transformed melanocytes.
To investigate the levels of the different regulatory proteins involved in the G1 progression and G1/S transition in normal and transformed human choroidal melanocytes (CM).. Three choroidal melanoma cell lines and three CM cultures were used. The purity of the CM cultures was assessed by different approaches, including morphologic study, specific immunostaining, cell proliferation behavior, and transforming growth factor-beta1 responsiveness. The cell cycle protein levels were evaluated by specific immunoblotting of total extracts obtained from the different cell lines.. Alterations were observed in the expression of cylins D1 and E in the transformed cells, whereas the amounts of the cyclin-dependent kinases (CDKs) CDK2 and CDK4 were almost identical in both cell types. Although the expression of cyclin H was slightly increased in transformed cells, neither the CDK7 level nor the CDK7 and cyclin H localizations were altered when compared with those in normal CM. The results suggest the absence of the CDK inhibitor (CKI) p21 in two of the three melanoma cell lines and, as a main feature, a striking underexpression of p27 in the three transformed cell lines. Finally, although the p16 level was almost the same in normal and transformed cells, a loss of p16-CDK4 interaction was observed in two of the three melanoma cell lines.. Deregulated expression of G1 cyclins and CKIs and alteration in the interaction of CKIs with CDKs may be implicated in the neoplastic transformation of human ocular melanocytes to malignant melanoma cells. Topics: Cell Division; Cell Line, Transformed; Choroid Neoplasms; Cyclin-Dependent Kinases; Cyclins; Electrophoresis, Polyacrylamide Gel; Fibroblasts; Fluorescent Antibody Technique, Indirect; G1 Phase; Humans; Immunoblotting; Immunoenzyme Techniques; Melanocytes; Melanoma; Pigment Epithelium of Eye; Transforming Growth Factor beta; Tumor Cells, Cultured | 1998 |
Elevated plasma levels of transforming growth factor (TGF)-beta1 and TGF-beta2 in patients with disseminated malignant melanoma.
Overexpression of transforming growth factor-beta isoforms (TGF-beta1, -beta2, -beta3) has been previously reported in human melanoma cell lines and tumours. The aim of the present study was to evaluate the plasma levels of TGF-beta isoforms in melanoma patients. Significantly elevated levels of TGF-beta1 (4.2 x the controls, P = 0.0094) and of TGF-beta2 (1.5 x the controls, P = 0.012) but not of TGF-beta3 were measured in patients with disseminated but not locoregional melanoma. These results indicate systemic circulation of potentially immunosuppressive peptides of the TGF-beta family in end-stage melanoma patients. Topics: Humans; Lymphatic Metastasis; Melanoma; Neoplasm Proteins; Neoplasm Staging; Transforming Growth Factor beta | 1998 |
Cytokine sensitivity of metastatic human melanoma cell lines-- simultaneous inhibition of proliferation and enhancement of gelatinase activity.
The effect of a panel of cytokines on the proliferation and type IV collagenase production was studied in four melanoma cell lines of different origin, tumorigenicity and metastatic capacity. TGF-b, TNF-a and to a lesser extent, IL-1a exhibited antiproliferative effect on the cell lines, with some lines showing varying degree of resistance. The sensitivity did not correlate directly with the origin or the biological behavior of the tumor lines, suggesting that cytokine resistance of advanced stage melanoma cells may be relative. IL-2, IL-10 and IL-12 displayed little or no effect on proliferation. The effect of cytokines on metalloproteinase production showed a cell line dependent pattern. Interestingly, those cytokines that exhibited the most pronounced antiproliferative activity, also proved most effective in stimulating collagenase secretion, often simultaneously, in the same line. The results indicate that pleiotropic cytokines can have positive and negative effects simultaneously on various steps of tumor progression. Topics: Animals; Cell Division; Cytokines; Gelatinases; Humans; Interleukin-1; Interleukin-10; Interleukin-12; Interleukin-2; Melanoma; Mice; Mice, Nude; Neoplasm Metastasis; Recombinant Proteins; Skin Neoplasms; Transforming Growth Factor beta; Transplantation, Heterologous; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha | 1998 |
In vitro modulation of human melanoma cell invasion and proliferation by all-trans-retinoic acid.
Invasive growth and formation of metastases involve complex interactions between tumour cells, host cells and components of the extracellular matrix. Retinoids, a group of vitamin A derivatives, modulate cell growth and differentiation and have been found to suppress tumour cell invasion in vitro and formation of metastases in vivo. The aim of our study was to investigate changes in proliferation and invasion through membrane barriers in vitro of seven human melanoma cell lines, established from human primary melanomas or metastases, in response to treatment with retinoic acid (RA). These changes were compared with the expression regulation of molecules that have been identified as targets of RA-mediated signal pathways. Invasiveness in vitro was correlated with the origin of the cell lines and was significantly higher in the lines derived from metastases. In all the cell lines proliferation and chemotaxis were inhibited by 10(-5) M RA, but the cell lines established from metastases were significantly more sensitive with respect to inhibition of invasion by RA. The specific expression patterns of MMP-1 and TIMP-2 were detected and regulated by RA in almost all cell lines, whereas expression of MMP-2 and TIMP-1 was not influenced by RA treatment. The most striking difference between the cell lines was a strong downregulation of transforming growth factor-beta (TGF-beta) expression in cell lines derived from metastases when treated with RA in contrast to cell lines from primary melanomas. These data provide evidence that RA modulates growth, chemotaxis and invasion in a broad panel of melanoma cell lines derived both from primary non-metastasized melanomas and metastases. However, distinct molecular mechanisms are involved in mediating these effects. Topics: Antineoplastic Agents; Blotting, Northern; Cell Division; Chemotaxis; Collagen; Collagenases; Dose-Response Relationship, Drug; Drug Combinations; Enzyme-Linked Immunosorbent Assay; Gelatinases; Gene Expression Regulation; Humans; Laminin; Matrix Metalloproteinase 1; Matrix Metalloproteinase 2; Matrix Metalloproteinase 9; Melanoma; Metalloendopeptidases; Neoplasm Invasiveness; Neoplasm Proteins; Protease Inhibitors; Proteoglycans; Tissue Inhibitor of Metalloproteinase-1; Tissue Inhibitor of Metalloproteinase-2; Transforming Growth Factor beta; Tretinoin; Tumor Cells, Cultured | 1998 |
Effects of oncostatin M and tamoxifen on human melanoma cells.
New agents are required in the treatment of malignant melanoma, to be used alone or in combination with established therapies. Oncostatin M (OSM), a member of the gp130 family of cytokines, has previously been shown to inhibit the growth of melanoma cell lines. Tamoxifen (TAM) is widely used in the treatment of melanoma, typically in combination with chemo- and/or immunotherapy. A component of the antitumour activity of TAM is via modulation of transforming growth factor-beta (TGF beta) which has previously been shown to be synergistic with OSM in vitro. To further investigate the clinical potential of OSM, alone and in combination with TAM, set concentrations of each were added to nine fresh and two well-established melanoma cell lines. The proliferation of seven of the 11 cell lines was inhibited by OSM, while two were unresponsive at the dose range tested. Of particular interest was the finding that the growth of two of the cell lines was significantly stimulated at low doses of OSM. The combination of OSM with TAM produced widely divergent results, most frequently resembling the effects of OSM alone. No synergism between the two was evident in any of the cell lines tested. Our results indicate that a combination of OSM and TAM in clinical trials needs further evaluation before it can be recommended. Furthermore, while OSM alone may be useful in the treatment of melanoma, this may be complicated by the possibility of stimulating tumour growth in some instances. Topics: Antineoplastic Agents; Antineoplastic Combined Chemotherapy Protocols; Cell Division; Humans; Melanoma; Oncostatin M; Peptides; Tamoxifen; Transforming Growth Factor beta; Tumor Cells, Cultured | 1998 |
Adenoviral transduction of melanoma cells with B7-1: antitumor immunity and immunosuppressive factors.
Previous studies in experimental models have demonstrated that the transduction of human or murine melanoma cells with the co-stimulatory B7-1 molecule induces effective antitumor immune responses. In order to develop B7-1 gene transfer as a therapeutic tool in the clinical management of melanoma, efficient means of in vivo gene transfer must be used. To this end we evaluated in vitro and in vivo immune responses associated with adenoviral transduction of murine and human melanoma cells with B7-1. Adenovirus-mediated transduction of human and murine melanoma cells with B7-1 leads to high-level transgene expression in vitro and in vivo and does not affect MHC class I and II expression. Adenovirus-delivered B7-1 induced antitumor immune responses, on the basis of observations that human melanoma cells transduced to express human B7-1 were able to co-stimulate allogeneic and autologous T cells to proliferate and that murine melanoma K1735 cells transduced to express murine B7-1 were rejected by syngeneic, immunocompetent mice. By contrast, intratumoral injection of an adenovirus encoding murine B7-1 failed to eliminate established murine melanoma (K1735) despite high-level transgene expression in tumor cells. Potent T cell inhibitory factor(s) secreted by both K1735 cells and select human melanoma cells may contribute to the failure to achieve protection in this setting. Thus, immune inhibitory melanoma-derived factors need to be taken into account when considering the clinical use of B7-1 immunotherapy. Topics: Adenoviridae; Animals; B7-1 Antigen; CHO Cells; Cricetinae; Gene Expression; Histocompatibility Antigens Class I; Histocompatibility Antigens Class II; Humans; Immunosuppressive Agents; Lymphocyte Activation; Melanoma; Melanoma, Experimental; Mice; T-Lymphocytes; Transduction, Genetic; Transforming Growth Factor beta; Transgenes; Tumor Cells, Cultured | 1998 |
Expression of fibrogenic cytokines in desmoplastic malignant melanoma.
Desmoplastic malignant melanoma (DMM) consists of amelanotic spindle-shaped melanoma cells and is accompanied by desmoplasia with fibrous stromata. It has a strong tendency for local infiltrative growth and recurrence and a propensity for neurotropism. It is not yet known which cytokine is responsible for the desmoplasia in DMM. In the present study, we investigated the roles of several fibrogenic cytokines and cytokine receptors in DMM: basic fibroblast growth factor (bFGF), connective tissue growth factor (CTGF), transforming growth factor-beta, platelet-derived growth factor (PDGF) and PDGF receptors. Immunostaining and in situ hybridization were conducted in four cases of DMM and four cases of amelanotic malignant melanoma (AMM) as negative controls for desmoplasia. PDGF-beta receptor, bFGF and CTGF were intensely expressed in the DMM specimens in comparison with the AMM specimens. The reaction of PDGF-B ligand and CTGF to PDGF-beta receptor, in addition to the expression of bFGF, may contribute to the desmoplasia in DMM. Topics: Adult; Aged; Aged, 80 and over; Connective Tissue Growth Factor; Female; Fibroblasts; Growth Substances; Humans; Immediate-Early Proteins; Immunoenzyme Techniques; In Situ Hybridization; Intercellular Signaling Peptides and Proteins; Male; Melanoma; Middle Aged; Neoplasm Proteins; Platelet-Derived Growth Factor; Receptors, Platelet-Derived Growth Factor; RNA, Messenger; Skin Neoplasms; Transforming Growth Factor beta | 1998 |
Human ex vivo carcinoma cells produce transforming growth factor beta and thereby can inhibit lymphocyte functions in vitro.
We tested 20 human carcinoma samples for the production of transforming growth factor beta (TGFbeta) in vitro. Tumour cell suspensions without obvious contamination with non-malignant cells were kept in culture conditions for 16 h and their supernatants were added to CCL-64 cells. The proliferation of these cells is inhibited by TGFbeta. According to this assay, the supernatants contained both active and latent TGFbeta. In addition, the supernatants were found to suppress the spontaneous cytotoxic function and activation of T-cell-enriched lymphocyte populations. A specific monoclonal antibody (mAb) counteracted these effects and therefore we concluded that they were mediated to a large extent by TGFbeta. In line with the results obtained with the supernatants, activation of lymphocytes could also be inhibited by tumour cells and their inhibitory effect was weaker in the presence of the TGFbeta-specific mAb. It is important to note that, when TGFbeta-specific mAb was added to autologous mixed lymphocyte/tumour cell cultures, lymphocyte activation occurred more often. These results thus substantiate the assumption that production of TGFbeta may help the survival of potentially immunogenic tumour cells in immunocompetent patients. Topics: Carcinoma; Female; Humans; Lung Neoplasms; Lymphocyte Activation; Male; Melanoma; Ovarian Neoplasms; T-Lymphocytes, Cytotoxic; Transforming Growth Factor beta; Tumor Cells, Cultured | 1997 |
Stimulation of interleukin-11 production from osteoblast-like cells by transforming growth factor-beta and tumor cell factors.
Bone is one of the most common sites of metastasis in melanoma and breast cancer cells. Human melanoma (A375M) and human breast cancer (MDA-MB-231) cells form osteolytic bone metastasis in vivo when these tumor cells are injected into the left ventricles of BALB/c nude mice. These tumor cells promote bone resorption in the in vitro neonatal murine calvaria organ culture system by indirectly stimulating the production of a bone resorption-inducing factor (or factors) from human osteoblast-like cells. This secreted factor was identified as interleukin-11 (IL-11). Although many cytokines and hormones were associated with IL-11 production from osteoblasts, transforming growth factor-beta (TGF-beta) was found to be involved in the promotion of IL-11 production from osteoblasts, because the addition of a neutralizing anti-TGF-beta antibody decreased the production of IL-11. However, these tumor cells did not produce TGF-beta by themselves. We found that they enhanced IL-11 production by activating latent TGF-beta produced from osteoblast-like cells. Our results indicate that metastatic tumor cells induce osteolysis by activating TGF-beta, which leads IL-11 production from osteoblasts to promote bone resorption. Topics: Animals; Antibodies; Bone Neoplasms; Bone Resorption; Breast Neoplasms; Cell Line; DNA Primers; Enzyme-Linked Immunosorbent Assay; Female; Humans; Interleukin-11; Interleukin-6; Melanoma; Mice; Mice, Nude; Osteoblasts; Polymerase Chain Reaction; RNA, Messenger; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured | 1997 |
Sequence and expression of a novel member (LTBP-4) of the family of latent transforming growth factor-beta binding proteins.
Overlapping cDNA clones from human heart and melanoma libraries were used to establish the 1587-residue sequence of a novel protein (LTBP-4) belonging to the family of extracellular microfibrillar proteins which also bind transforming growth factor-beta. LTBP-4 consists of 20 EG modules, 17 of them with a consensus sequence for calcium binding, 4 TB modules with 8 cysteines and several proline-rich regions. Northern blots demonstrated a single 5 kb mRNA which is highly expressed in heart but also present in skeletal muscle, pancreas, placenta and lung. The modular structure predicts that LTBP-4 should be a microfibrillar protein which probably also binds TGF-beta. Topics: Adaptor Proteins, Signal Transducing; Amino Acid Sequence; Base Sequence; Blotting, Northern; Calcium; Carrier Proteins; Cloning, Molecular; Extracellular Matrix; Gene Expression; Humans; Intracellular Signaling Peptides and Proteins; Latent TGF-beta Binding Proteins; Melanoma; Molecular Sequence Data; Muscle Proteins; Myocardium; Protein Binding; RNA, Messenger; Sequence Alignment; Sequence Analysis, DNA; Transforming Growth Factor beta | 1997 |
In situ expression of transforming growth factor beta is associated with melanoma progression and correlates with Ki67, HLA-DR and beta 3 integrin expression.
Transforming growth factor-beta (TGF beta), which is secreted by cultured melanoma cells constitutively, inhibits the proliferation of normal melanocytes and of most melanoma cells in vitro, but some melanoma cells from advanced stages of the disease develop resistance to TGF beta-dependent growth inhibition, without developing any change in TGF beta cell surface binding. In vitro TGF beta also downregulates the expression of HLA-DR molecules on melanoma cells, and upregulates the expression of the beta 3 integrin subunit on some cell lines. Immunohistochemical analysis of 53 melanocytic lesions (12 naevi, 30 primary melanomas and 11 metastases) revealed a trend of increasing expression of TGF beta and TGF beta receptor type III with tumour progression, and a significantly higher expression of both TGF beta (P < 0.0001) and the receptor (P < 0.05) in metastatic and thick (> 1 mm) primary melanomas compared with thin (< 1 mm) primary melanomas. The expression of TGF beta correlated with expression of a marker of proliferation, Ki67, and with HLA-DR and beta 3 integrin subunit expression. Coexpression of the four molecules was observed in all metastases and in most thick primary melanomas. These findings argue against an inhibitory effect of TGF beta on cell proliferation or HLA-DR antigen expression in melanoma, and suggest the upregulation of the beta 3 subunit. TGF beta protein appears to be a biological marker of melanoma progression in situ. Topics: Adult; Antigens, CD; Cell Division; Disease Progression; Down-Regulation; Female; HLA-DR Antigens; Humans; Integrin beta3; Ki-67 Antigen; Male; Melanoma; Nevus; Phenotype; Platelet Membrane Glycoproteins; Skin Neoplasms; Transforming Growth Factor beta; Up-Regulation | 1997 |
Transforming growth factor-beta enhances adhesion of melanoma cells to the endothelium in vitro.
Melanoma invasion requires migration through the vascular barrier. An early event in this process is the adhesion of metastatic cells to the endothelium. To elucidate the role of TGF-beta in the regulation of this process, human melanoma SK-MEL24 cells were labelled with [5'-(3)H]-thymidine and co-cultured with bovine pulmonary artery endothelial-cell monolayers. Radioactivity was assumed to be proportional to the number of SK-MEL24 cells bound to the endothelium. A low number of melanoma cells adhered to endothelial cells in a time-related manner. Pretreatment for 24 hr with 0.001 to 10 ng/ml TGF-beta1 or TGF-beta2 of both cell types enhanced melanoma-endothelium adhesion in a dose-dependent manner. Both melanoma and endothelial cells expressed RI- and RII-type TGF-beta receptors. The effect of TGF-beta was abolished by co-incubation with the proteoglycan decorin. Conditioned media from melanoma-endothelium co-cultures contained latent TGF-beta and failed to affect cell-cell adhesion. However, activation of TGF-beta by heating the medium or reducing the pH, increased melanoma-endothelium adhesion to an extent similar to that of the TGF-beta administered to the cultures. Zimography demonstrated that both cell types expressed urokinase-type plasminogen activator (uPA). Addition of plasminogen to the co-cultures, which was likely to be activated to plasmin by uPA, resulted in activation of TGF-beta and parallel stimulation of melanoma-endothelium adhesion. In conclusion, TGF-beta may enhance adhesion of melanoma cells to the endothelium, playing a relevant autocrine/paracrine role in the progression of invasive melanoma. Topics: Activin Receptors, Type I; Animals; Cattle; Cell Adhesion; Cells, Cultured; Coculture Techniques; Decorin; Endothelium, Vascular; Extracellular Matrix Proteins; Fibrinolysin; Humans; Kinetics; Melanoma; Plasminogen; Protein Serine-Threonine Kinases; Proteoglycans; Pulmonary Artery; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Recombinant Proteins; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured; Urokinase-Type Plasminogen Activator | 1997 |
Melanoma-associated expression of transforming growth factor-beta isoforms.
Melanocytic neoplasia is characterized by the aberrant overproduction of multiple cytokines in vitro. However, it is largely unknown how cytokine expression relates to melanoma progression in vivo. Transforming growth factor beta (TGF-beta) is a multifunctional cytokine commonly produced by cultured melanoma cells. The in situ expression of all three TGF-beta isoforms (TGF-beta1, -2, and -3) was determined immunohistochemically in melanocytes and in 51 melanocytic lesions using isoform-specific antibodies. Significant linear trends of expression were observed from melanocytes through nevi and primary and metastatic melanomas for all three isoforms. TGF-beta1 was expressed by some melanocytes and almost uniformly by nevi and melanomas. TGF-beta2 and -3 were not expressed in normal melanocytes but were expressed in nevi and early and advanced primary (radial and vertical growth phase) and metastatic melanomas in a tumor-progression-related manner. TGF-beta2 was heterogeneously expressed in advanced primary and metastatic melanomas, whereas TGF-beta3 was uniformly and highly expressed in these lesions. Thus, expression of TGF-beta isoforms in melanocytes and melanocytic lesions is heterogeneous and related to tumor progression, and expression of TGF-beta2 and TGF-beta3 commences at critical junctures during progression of nevi to primary melanomas. Topics: Biomarkers, Tumor; Blotting, Western; Disease Progression; Humans; Immunohistochemistry; Melanocytes; Melanoma; Nevus, Intradermal; Retrospective Studies; Skin Neoplasms; Transforming Growth Factor beta | 1996 |
Effect of transforming growth factor-beta on plasminogen activator production of cultured human uveal melanoma cells.
Human uveal melanoma cells have been shown to produce plasminogen activator (PA), an enzyme which can enhance tumor metastasis by promoting degradation of extracellular matrix. This study used cultured human uveal melanoma cells to determine whether the PA production of uveal melanoma cells could be modulated by transforming growth factor-beta2 (TGF-beta2), a mitogen present in the uvea.. Five different cell lines of human uveal melanoma of differing cellular morphology (2 spindle, 2 epithelioid, 1 mixed) derived from tumors from different locations in the eye (3 choroidal, 1 ciliochoroidal, 1 orbital) were grown in serum-free media, in the presence or absence of TGF-beta2 (1ng/ml to 100ng/ml). After 24 hrs, the conditioned media were collected and quantitated for PA activity by measuring the radial diffusion in fibrin-agarose clot and for total PA concentration using an enzyme-linked immunoassay.. Among the cell lines studied, all produced PA. Cell lines derived from intraocular tumors secreted tissue-type PA (tPA), and TGF-beta2 stimulated tPA activity and secretion of cell lines containing epithelioid cells but had no effect on spindle cells. In contrast, tumor cells isolated from an orbital tumor secreted urokinase (uPA), activity and secretion of which was inhibited by TGF-beta2.. We conclude that cultured human uveal melanoma cells produce either tPA or uPA, and TGF-beta2 can have a variable effect on PA production of these cells. Topics: Culture Media, Conditioned; Fibroblast Growth Factor 2; Humans; Melanoma; Plasminogen Activator Inhibitor 1; Tissue Plasminogen Activator; Transforming Growth Factor beta; Tumor Cells, Cultured; Urokinase-Type Plasminogen Activator; Uveal Neoplasms | 1996 |
Transforming growth factor beta 1 increases the synthesis and shedding of the melanoma-specific proteoglycan in human melanoma cells.
Melanoma cells produce a cell-specific proteoglycan, mel-PG, which is an integral chondroitin sulfate proteoglycan that can be released into the medium as a result of the proteolytic cleavage of the trans-membrane form. The effect of transforming growth factor beta 1 (TGF-beta 1) on proteoglycan production was studied in three melanoma cell lines at various stages of differentiation: SK-mel-1.36-1-5 (early), SK-mel-3.44 (intermediate), and SK-mel-23 (late). The main effect of TGF-beta 1 was to increase the synthesis and shedding of mel-PG into the medium without affecting the amount present in the cell membranes nor the balance between the proteoglycan and the glycoprotein forms of mel-PG. After TGF-beta 1 treatment, there was an increase in the amount of mel-PG present in the medium as observed in metabolic labeling, immunoprecipitation, and pulse-chase experiments. This effect was more pronounced in the SK-mel-1.36-1-5 than in the SK-mel-3.44 cell line, whereas the SK-mel-23 cells did not contain mel-PG either in the presence or in the absence of TGF-beta 1. Characterization of mel-PG purified from the medium from control and TGF-beta 1-treated cells showed that the factor increased slightly the GAG chain length in SK-mel-1.36-1-5 but not in SK-mel-3.44 cells, without modifying the degree of sulfation. Topics: Cell Differentiation; Cell Membrane; Extracellular Space; Humans; Melanoma; Molecular Structure; Proteoglycans; Transforming Growth Factor beta; Tumor Cells, Cultured | 1996 |
Active transforming growth factor-beta in human melanoma cell lines: no evidence for plasmin-related activation of latent TGF-beta.
Cultured human melanoma cells were found to secrete TGF-beta mostly in latent biologically inactive form but in addition five of six melanoma cell lines studied produced in conditioned culture medium active TGF-beta in the range from 370 to 610 pg per 10(6) cells per 24 h. A distinct characteristic of these melanoma cell lines is that they form active surface-bound plasmin by the activation of plasminogen with surface-bound tissue-type plasminogen activator. The present study was performed to assess the role of plasmin in the process of latent TGF-beta activation in the melanoma cell lines. No direct correlation was found between cell-associated plasmin activity and the amount of active TGF-beta present in the conditioned medium of individual cell lines. The melanoma cell lines exhibited diverse responses to exogenous active TGF-beta 1; three cell lines were growth-stimulated, two were growth-inhibited, and one had a very low sensitivity to the growth factor. The active TGF-beta produced by the melanoma cells was found to inhibit the natural killer cell function of peripheral blood lymphocytes, suggesting that it may have an immunosuppressive effect and a role in the development of melanomas. Topics: Culture Media, Conditioned; DNA Replication; Fibrinolysin; Hot Temperature; Humans; Melanoma; Plasminogen; Transforming Growth Factor beta; Tumor Cells, Cultured | 1996 |
Selective growth of human melanoma cells in the brain parenchyma of nude mice.
The purpose of this study was to determine whether the growth of human melanoma cells in the brain parenchyma is selective and represents the growth of unique cells. Six human melanoma cell lines derived from cutaneous lymph node or brain metastases (from six different patients) and melanoma cells isolated from fresh surgical specimens of two primary cutaneous melanomas, two lymph node metastases and two brain metastases (each from a different patient) were injected into the subarachnoid space of nude mice. All melanomas produced growths in the leptomeninges, but only melanoma cells isolated from brain metastases infiltrated into and grew in the brain parenchyma of nude mice. The results from in vitro assays for cell motility or production of gelatinase activity did not correlate with in vivo growth pattern. However, the in vitro growth of human melanoma cells in the presence of TGF-beta 2 inversely correlated with potential for brain parenchyma metastasis, i.e. the growth of cells from brain metastases was least inhibited by TGF-beta 2. These data suggest that melanoma brain parenchyma metastases are produced by unique cells that may be resistant to the antiproliferative effects of TGF-beta 2. Topics: Animals; Brain Neoplasms; Cell Division; Cell Movement; Gelatinases; Humans; Injections, Intraventricular; Lymphatic Metastasis; Male; Melanoma; Melanoma, Experimental; Meningeal Neoplasms; Mice; Mice, Nude; Neoplasm Transplantation; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured | 1996 |
Expression and structural features of endoglin (CD105), a transforming growth factor beta1 and beta3 binding protein, in human melanoma.
Human endoglin (CD105) is a member of the transforming growth factor beta (TGF-beta) receptor family that binds TGF-beta1 and -beta3, but not TGF-beta2, on human endothelial cells. Immunohistochemical analyses demonstrated that CD105 is expressed on normal and neoplastic cells of the melanocytic lineage. The anti-CD105 MAb, MAEND3, stained 50, 25 and 34% of intradermal naevi, primary and metastatic melanomas investigated, respectively, and nine out of 12 melanoma cell lines. Sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis revealed that CD105 expressed by melanoma cells consists of a homodimeric protein with an apparent molecular weight of 180 and 95 kDa under non-reducing and reducing conditions. Cross-linking of 125I-labelled TGF-beta1 to melanoma cells, Mel 97, by disuccinimidyl suberate (DSS) demonstrated that CD105 expressed on pigmented cells binds TGF-beta1; the pattern of binding of TGF-beta1 to melanoma cells was found to be similar to that of human umbilical vein endothelial cells. The addition of exogenous, bioactive TGF-beta1 significantly (P<0.05) inhibited the growth of CD105-positive melanoma cells, Mel 97, but did not affect that of CD105-negative melanoma cells, F0-1. These data, altogether, demonstrate that CD105 is expressed on pigmented cells and might play a functionally relevant role in the biology of human melanoma cells by regulating their sensitivity to TGF-betas. Topics: Animals; Antigens, CD; Cell Division; Electrophoresis, Polyacrylamide Gel; Endoglin; Fluorescent Antibody Technique, Indirect; Humans; Immunohistochemistry; Iodine Radioisotopes; Melanoma; Mice; Mice, Inbred BALB C; Neoplasm Proteins; Nevus; Receptors, Cell Surface; Skin Diseases; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured; Vascular Cell Adhesion Molecule-1 | 1996 |
TGF-beta mediated G1 arrest in a human melanoma cell line lacking p15INK4B: evidence for cooperation between p21Cip1/WAF1 and p27Kip1.
We have studied TGF-beta mediated G1 arrest in WM35, an early stage human melanoma cell line. These cells have lost p15INK4B expression through loss of one chromosome 9 and rearrangement of the other. In asynchronously growing WM35, TGF-beta caused reductions in cyclin D1, cyclin A and cdk4 proteins and their associated kinase activities and an increase in both p21Cip1/WAF1 and p27Kip1. These findings were confirmed in cells released from quiescence in the presence of TGF-beta, in which TGF-beta inhibited or delayed the reduction in the cdk inhibitors that normally occurs in late G1. In contrast to observations in other cell types, there was an increased association of both p21Cip1/WAF1 and p27Kip1 with cyclin D1/cdk4 and with cyclin E/cdk2 during TGF-beta mediated arrest of asynchronously growing cells. Upregulation of p21Cip1/WAF1 preceded that of p27Kip1. Furthermore, p21Cip1/WAF1 and p27Kip1 were not present in the same cdk complexes but bound distinct populations of target cdk molecules. Both p21Cip1/WAF1 and p27Kip1 immunoprecipitates from asynchronously growing cells contained active kinase complexes. These KIP-associated kinase activities were reduced in TGF-beta arrested cells. It has been proposed that in TGF-beta arrested epithelial cells, up-regulation of p15INK4B and of p15INK4B binding to cdk4 serves to destabilize the association of p27Kip1 with cyclin D1/cdk4, promoting p27Kip1 binding and inhibition of cyclin E/cdk2. Our findings demonstrate that this is not a universal mechanism of G1 arrest by TGF-beta. In TGF-beta arrested WM35, which lack p15INK4B, the increased p21Cip1/WAF1 may serve a similar function to that of p15INK4B: initiating kinase inhibition and providing an additional mechanism to supplement the effect of p27Kip1 on G1 cyclin/cdks. Topics: Carrier Proteins; Cell Cycle Proteins; Chromosomes, Human, Pair 9; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p16; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinase Inhibitor p27; Cyclins; G1 Phase; Gene Rearrangement; Humans; Melanoma; Microtubule-Associated Proteins; Neoplasm Proteins; Phosphorylation; Retinoblastoma Protein; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Proteins; Up-Regulation | 1996 |
The effect of TGF-beta 1 on cell proliferation and proteoglycan production in human melanoma cells depends on the degree of cell differentiation.
The effect of transforming growth factor beta 1 (TGF-beta 1) on cell proliferation, colony formation in soft agar and synthesis and structure of proteoglycans was studied in three human melanoma cell lines at different stages of differentiation: SK-mel-1.36-1-5 (early), SK-mel-3.44 (intermediate) and SK-mel-23 (late). TGF-beta 1 potently inhibited cell growth in monolayer as well as in soft agar. TGF-beta 1 increased the release of sulfated proteoglycans into the medium, including the cell-specific melanoma proteoglycan, mel-PG, and induced changes in disaccharide composition and sulfation of the glycosaminoglycan chains. In all the cases, the effect of TGF-beta 1 was more pronounced in the most undifferentiated cell line SK-mel-1.36-1-5 than in the SK-mel-3.44, whereas it had no effect on the most differentiated SK-mel-23 cells. Topics: Cell Differentiation; Cell Division; Humans; Melanoma; Neoplasm Proteins; Proteoglycans; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Stem Cell Assay | 1996 |
Transforming growth factor-beta down-regulates major histocompatibility complex class I antigen expression and increases the susceptibility of uveal melanoma cells to natural killer cell-mediated cytolysis.
Intraocular melanomas, especially those of the anterior segment, reside within an immunologically privileged milieu. Aqueous humour contains a variety of immunomodulatory factors that are believed to contribute to ocular immune privilege. Among these is transforming growth factor-beta (TGF-beta), which has been shown to down-regulate major histocompatibility complex (MHC) class I antigens on normal cells. Since the susceptibility of tumour cells to natural killer (NK) cell-mediated lysis is inversely correlated with the expression of MHC class I antigens, tumour cells exposed to TGF-beta might be expected to experience enhanced susceptibility to NK-mediated killing. This was examined by incubating two human uveal melanoma cell lines in the presence of TGF-beta and evaluating the expression of MHC class I antigen and susceptibility to NK cell-mediated lysis. OCM1 and OCM8 melanoma cells constitutively express high levels of class I antigen (85-90% positive) and low susceptibility to NK-mediated lysis in vitro (3-8%). Incubation with TGF-beta produced a significant reduction in class I antigen expression (52-62%) and a proportional increased susceptibility to NK cell-mediated cytolysis (17%). Analogous effects were found using a human uveal melanoma cell line (OCM3) that constitutively expresses low amounts of class I (< 5% positive) and high NK susceptibility (35% lysis). Stimulation of class I antigen expression by incubation with interferon-gamma resulted in a sharp increase in class I expression (80% positive) and a comparable diminution in susceptibility to NK cell-mediated lysis (< 10%). The results indicate that TGF-beta, at concentrations found in the aqueous humour, can significantly alter MHC class I antigen expression and the susceptibility of ocular melanoma cells to NK cell-mediated cytolysis. Topics: Animals; Cytotoxicity, Immunologic; Down-Regulation; Histocompatibility Antigens Class I; Humans; Interferon-gamma; Killer Cells, Natural; Melanoma; Mice; Mice, Inbred BALB C; Mice, Nude; Recombinant Proteins; Transforming Growth Factor beta; Tumor Cells, Cultured; Up-Regulation; Uveal Neoplasms | 1995 |
Effects of various synthetic retinoids on proliferation and immunophenotype of human melanoma cells in vitro.
Since response rates in human melanoma are low with currently available therapeutic modalities, we have reevaluated the potential usefulness of retinoids as new alternatives for therapy of metastatic melanoma. Nine synthetic retinoids with high affinity and/or selectivity for the retinoic acid receptors (RAR) alpha, beta, and gamma were studied in comparison to all-trans retinoic acid (RA) for their in vitro effects on melanoma cell proliferation and for their immunomodulating capacities using four human melanoma cell lines. Eight out of ten retinoids tested had no effect on melanoma cell growth, whereas the remaining two compounds with high RAR-gamma selectivity (CD437 and CD2325) showed a dose-dependent antiproliferative effect on all melanoma cell lines with IC50 (concentration inhibiting response by 50%) values between 10(-6) and 10(-7)M. Further analyses showed that paracrine-mediated tumor cell growth inhibition such as induction of transforming growth factor (TGF)-beta described as one mechanism of retinoid action and enzyme systems such as tyrosinase and monoamine oxidase were not involved in mediating the antiproliferative effects exerted by the two retinoids. Four of nine retinoids modulated HLA-DR expression on human melanoma cells, and expression levels of intercellular adhesion molecule 1 (ICAM-1) was increased by another subset of compounds. These effects were, however, not correlated to the receptor selectivity of the retinoids. The potent growth inhibitory effect of the RAR-gamma-selective retinoids and the immunomodulating capacities of the retinoids open an interesting alternative for new antiproliferative and immunomodulatory strategies in the treatment of metastatic melanoma. Topics: Antigens, CD; Antigens, Neoplasm; Antigens, Surface; Cell Division; Gene Expression Regulation, Neoplastic; HLA-DR Antigens; Immunophenotyping; Intercellular Adhesion Molecule-1; Melanoma; Neoplasm Proteins; Receptors, Retinoic Acid; Retinoids; Skin Neoplasms; Transforming Growth Factor beta; Tumor Cells, Cultured | 1995 |
In situ analysis of transforming growth factor-beta s (TGF-beta 1, TGF-beta 2, TGF-beta 3), and TGF-beta type II receptor expression in malignant melanoma.
We have analysed, by in situ hybridization, mRNA expression of TGF-beta 1, TGF-beta 2, TGF-beta 3, and of TGF-beta type II receptor in benign melanocytic naevi, primary melanomas, and in skin metastases of malignant melanomas. Our results show that melanoma progression correlates with overexpression of TGF-beta. All skin metastases and most primary melanomas invasive to Clark's level IV-V revealed specific TGF-beta 2 mRNA and protein expression. However, expression of this cytokine was not observed in benign melanocytic lesions and was detected only in one of five early primary melanomas investigated. Some primary melanomas and skin metastases also revealed specific TGF-beta 1 mRNA signals although expression of this isoform was not found in benign naevi. TGF-beta 3 expression, which was only barely detectable in benign melanocytic lesions, was enhanced in some skin metastases. Interestingly, the epidermis overlaying melanomas revealed lower levels of TGF-beta 3 mRNA expression than epidermis of healthy skin or epidermis adjacent to benign naevi, thereby suggesting that paracrine mechanisms between tumour cells and keratinocytes may influence melanoma development. In primary melanomas TGF-beta type II receptor mRNA signals were much more heterogeneously distributed when compared to benign melanocytic naevi, suggesting variable degrees of TGF-beta resistance among melanoma cells within individual lesions. However, melanoma progression appeared not to be correlated with a complete loss of TGF-beta type II receptor gene expression, since all skin metastases revealed clearly detectable although heterogeneous levels of TGF-beta type II receptor mRNA expression. Topics: Disease Progression; Humans; Immunohistochemistry; In Situ Hybridization; Isomerism; Melanoma; Nevus, Pigmented; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Messenger; Sensitivity and Specificity; Skin Neoplasms; Tissue Distribution; Transforming Growth Factor beta | 1995 |
Regulation of interleukin-8 expression in human melanoma cells by the organ environment.
The in vitro expression level of interleukin-8 (IL-8) correlates with the metastatic potential of human melanoma cells. The purpose of this study was to determine whether the expression level of IL-8 in human melanoma cells is influenced by the organ microenvironment. A375P cells, a low metastatic human melanoma, and A375SM cells, a highly metastatic variant, were injected into the subcutis (s.c.), spleen (to produce liver metastases), and lateral tail vein (to produce lung metastases) of athymic nude mice. Northern blot and immunohistochemical analyses determined that s.c. tumors, lung lesions, and liver lesions expressed high, intermediate, and low IL-8, mRNA, and protein, respectively. This differential regulation of IL-8 was not due to the size or density of the lesions or to selection of subpopulations of cells. We based this conclusion on the results of three experiments: (a) melanoma cell lines established in culture from in vivo-growing tumors exhibited similar levels of IL-8 mRNA transcripts; (b) in a crossover experiment, the level of IL-8 mRNA was always high in A375 tumors reestablished in the skin and low in the tumors reestablished in the liver, regardless of whether the melanoma cells had been first harvested from s.c. or liver tumors; and (c) A375 melanoma cells cocultured with human keratinocytes produced high levels of IL-8 protein, whereas A375 cells cocultured with highly differentiated human hepatoma cells produced decreased levels. When A375P cells were then incubated with cytokines associated with keratinocytes (IL-1 and interferon beta) or hepatocytes (transforming growth factor alpha or beta), IL-1 enhanced the production of IL-8 protein, whereas TGF-beta decreased its production. These data show that IL-8 expression in melanoma cells is modulated by local host factors. Topics: Adaptation, Psychological; Animals; Cell Communication; Cell Division; Female; Humans; Interleukin-1; Interleukin-8; Keratinocytes; Liver Neoplasms, Experimental; Lung; Lung Neoplasms; Male; Melanoma; Mice; Mice, Inbred BALB C; Mice, Nude; Middle Aged; Neoplasm Transplantation; Organ Specificity; RNA, Messenger; Skin Neoplasms; Skin Physiological Phenomena; Transforming Growth Factor beta | 1995 |
Expression of transforming growth factor-beta 2 in malignant melanoma correlates with the depth of tumor invasion. Implications for tumor progression.
Transforming growth factor-beta 2 (TGF-beta 2) is a potent regulatory of proliferation and differentiation of both normal and malignant cells. In addition, TGF-beta 2 can exert a variety of immunosuppressive effects, suggesting that the production of this molecule contributes to impaired immunological surveillance of tumor development. In vitro, TGF-beta 2 expression has been demonstrated in cell lines derived from metastatic malignant melanocytes. but not in those derived from normal melanocytes. We sought to evaluate a potential role of TGF-beta 2 in the initiation or progression of malignant melanoma in vivo. We examined by nucleic acid in situ hybridization the expression of TGF-beta 2 mRNA transcripts in 124 melanocytic lesions including metastatic and primary invasive melanomas, melanomas in situ, nevi with architectural disorder and cytologic atypia, ordinary benign melanocytic nevi, and Spitz nevi. All metastatic melanomas and a majority (94%) of primary melanomas invasive to Clark's level III, IV, or V expressed TGF-beta 2 mRNA. A minority (41%) of Clark's level II primary invasive melanomas expressed this factor. All definitive melanomas in situ and nevi were negative. The results suggest that TGF-beta 2 expression in malignant melanoma may be a critical event in the development of deep invasion and metastases in malignant melanoma. Topics: Base Sequence; Humans; In Situ Hybridization; Melanoma; Molecular Sequence Data; Neoplasm Invasiveness; RNA, Messenger; Transforming Growth Factor beta | 1994 |
Transforming growth factor beta production and responsiveness in normal human melanocytes and melanoma cells.
Previous studies have shown that some human melanoma cells express transforming growth factor beta (TGF-beta) mRNA and are growth inhibited by exogenous TGF-beta, suggesting a possible negative autocrine role for this melanoma-derived growth factor. To better understand the role of endogenous TGF-beta in the development of melanoma, we investigated patterns of TGF-beta protein production and responsiveness of human melanoma cells as compared to normal melanocytes. Both cultured melanoma cells and normal melanocytes secreted biologically inactive, latent TGF-beta protein which, upon acid treatment, became biologically active. In melanoma cells, TGF-beta production occurred constitutively, i.e., in the absence of exogenous polypeptide growth factors. By contrast, in melanocytes, TGF-beta production depended on stimulation by exogenous growth factors such as insulin-like growth factor I. Exogenous, bioactive TGF-beta 1 at picomolar concentrations inhibited tritiated thymidine uptake of normal melanocytes, whereas melanoma cells demonstrated various degrees of resistance to TGF-beta-induced inhibition of DNA synthesis. Five of six cell lines were less sensitive than any of the melanocyte lines tested, and one cell line was completely resistant to inhibitory effects of TGF-beta on DNA synthesis. In vivo selection of melanoma cells for metastatic ability in athymic mice produced a variant cell line that was resistant to TGF-beta 1-induced inhibition of DNA synthesis and proliferation. Development of TGF-beta resistance in the variant cell line was not associated with changes in TGF-beta cell surface binding. Stable transfection of melanocytes with a plasmid expressing the Simian Virus 40 large T-antigen rendered these cells resistant to growth inhibition by TGF-beta, suggesting that TGF-beta inhibits melanoma/melanocyte growth via interaction with Simian Virus 40 large T-antigen-responsive transcription elements. Topics: Animals; Cell Division; Cell Transformation, Viral; Culture Media; Humans; Melanocytes; Melanoma; Mice; Mice, Nude; Receptors, Transforming Growth Factor beta; Time Factors; Transforming Growth Factor beta; Tumor Cells, Cultured | 1994 |
Production of multiple cytokines by cultured human melanomas.
We screened a panel of 8 primary and 21 metastatic melanoma cell lines for constitutive secretion of cytokines. Melanomas expressed bioactivity for TGF-beta (8/25 lines) and IFN (7/12), but not IL-2. Immunoassays detected IL-1 alpha (4/25), IL-1 beta (12/25), IL-6 (13/29), IL-8 (29/29), TGF-beta 2 (5/12) and GM-CSF (11/29), but not IL-3, IL-4, TNF-alpha, or IFN-gamma. There was no preferential association of cytokine production with cells cultured from primary versus metastatic disease, and only IL-8 was produced by all lines tested. These data demonstrate that cultured melanomas produce a variety of cytokines which are potentially capable of influencing tumor growth in vivo. Topics: Biological Assay; Culture Media, Conditioned; Cytokines; Enzyme-Linked Immunosorbent Assay; Humans; Interferons; Interleukin-8; Interleukins; Melanoma; Neoplasm Proteins; Transforming Growth Factor beta; Tumor Cells, Cultured | 1993 |
Transduction of human melanoma cell lines with the human interleukin-7 gene using retroviral-mediated gene transfer: comparison of immunologic properties with interleukin-2.
Two human melanoma cell lines were transduced with the human interleukin (IL)-7 and IL-2 genes using retroviral-mediated gene transfer. Stable, high-level cytokine expression was achieved. The in vitro growth of transduced tumors was unaltered. Neither of the IL-2-transduced melanoma cell lines grew in athymic mice, whereas one IL-7-transduced melanoma line showed retarded in vivo growth. This is consistent with animal studies suggesting a predominantly T-cell response to IL-7-transduced tumors and a more nonspecific response to IL-2-transduced tumors. Both IL-7- and IL-2-transduced melanoma cell lines could induce cytotoxic lymphocytes in mixed lymphocyte-tumor cultures. The expression of putative melanoma antigens (MAGE)-1 and MAGE-3 was unaltered by cytokine transduction. In one cell line, IL-7 transduction resulted in a marked inhibition of the immunosuppressive peptide transforming growth factor (TGF)beta 1. The results allow a comparison of immunobiologic properties of IL-7- and IL-2-transduced human melanoma cell lines in consideration of their use in genetically engineered tumor vaccines. IL-7 transduction results in stable cytokine expression and phenotypic alterations that appear to be favorable for enhanced immunogenicity and it deserves clinical testing. Topics: Animals; Antigens, Neoplasm; Base Sequence; Cell Division; Cell Line; Cytotoxicity, Immunologic; DNA Primers; DNA, Viral; Gene Transfer Techniques; Humans; Interleukin-2; Interleukin-7; Melanoma; Melanoma-Specific Antigens; Mice; Mice, Nude; Molecular Sequence Data; Neoplasm Proteins; Neoplasm Transplantation; Polymerase Chain Reaction; Retroviridae; RNA, Messenger; Transforming Growth Factor beta; Transplantation, Heterologous; Tumor Cells, Cultured; Virus Integration | 1993 |
Human melanoma cells express a functional interleukin-2 receptor.
Flow cytometric analysis reveals that 5 human melanoma cell lines (M14, IGR3, ME1477, JUSO, GLL19) express both alpha and beta chain of the interleukin 2 receptor (IL-2R alpha and IL-2R beta). These chains are able to specifically bind IL-2 and to form high-affinity heterodimers (IL-2R alpha beta). Analysis of poly A+ RNAs by Northern blot reveals the presence of typical transcripts for both the IL-2R alpha gene (3.6 kb) and the IL-2R beta gene (4 kb). Reverse transcription/polymerase chain reaction analysis allowed transcripts for the IL2R gamma (p64) gene to be detected in 3 of these melanoma cell lines (M14, IGR3, ME 1477). Incubation with human recombinant IL-2 modifies in IL-2R alpha+beta+gamma+ (M14) the expression of several surface molecules: down-regulation of ICAM-1, HLA class I and HLA-DR and up-regulation of CD44. IL-2 is also active on IL-2 alpha+beta+gamma- cell lines since it decreases ICAM-1 and HLA class-II expression at the surface of JUSO cells. Down-regulation of ICAM-1, whose expression in melanoma cells is a marker of tumor progression, is detectable within 3 hr in M14 cells and is maximal after 48 hr incubation, at IL-2 concentrations corresponding to the high-affinity heterodimers. This feature is specific since it is partially inhibited by MAbs directed against the IL-2 binding site of the IL-2R alpha (MAR93, 10T14) and IL-2R beta (MiK beta 1, TU27) chains. Our data support the notion of a direct effect of IL-2 on human melanoma cells. Modulation of the expression of surface molecules which is important for the interaction with immunocompetent cells or for tumor progression, could have a role to play during in vivo IL-2 treatment of human melanomas. Topics: Antigens, Neoplasm; Antigens, Surface; Base Sequence; Blotting, Northern; Flow Cytometry; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Molecular Sequence Data; Polymerase Chain Reaction; Receptors, Interleukin-2; RNA, Messenger; RNA, Neoplasm; Transforming Growth Factor beta; Tumor Cells, Cultured | 1993 |
Human pulmonary macrophages utilize prostaglandins and transforming growth factor beta 1 to suppress lymphocyte activation.
The ability to activate peripheral blood lymphocytes (PBLs) in vitro with interleukin-2 (IL-2) is suppressed by the presence of autologous human pulmonary alveolar macrophages (AMs). AMs suppress both IL-2-induced proliferation and the induction of lymphokine-activated killer cell (LAK) activity in a dose-dependent manner (79 +/- 6% suppression of LAK activity at a 0.25:1 AM/PBL ratio). Increasing the IL-2 concentration increased baseline LAK activity but did not prevent AM-mediated suppression. At least two different mechanisms of suppression were observed, one diffusible in nature and the other contact dependent. Indomethacin prevented the component of inhibition that diffused across porous polycarbonate membranes, indicating prostaglandins as the diffusible inhibitor. In contrast, indomethacin had no effect when added alone into conventional AM-PBL cocultures, but a combination of indomethacin and anti-transforming growth factor beta 1 (TGF-beta 1) antibody did prevent inhibition. This result suggests that TGF-beta 1 acts as an additional contact-dependent inhibitor. PBLs that were rendered unresponsive to IL-2 completely recovered their responsiveness within 4 days after removing AMs from the coculture. These features suggest that pulmonary macrophages have multiple mechanisms for locally suppressing IL-2 responses and lymphocyte activation. Topics: Adult; Antibodies; Bronchoalveolar Lavage Fluid; Cell Communication; Cells, Cultured; Cytotoxicity, Immunologic; Humans; Indomethacin; Interferon-gamma; Interleukin-2; Killer Cells, Lymphokine-Activated; Lymphocyte Activation; Macrophages, Alveolar; Melanoma; Middle Aged; Prostaglandins; Receptors, Interleukin-2; Transforming Growth Factor beta; Tumor Cells, Cultured | 1993 |
Human melanoma cell-derived factor(s) stimulate fibroblast glycosaminoglycan synthesis.
Conditioned media from cultures of human metastatic Hs294T melanoma cells contain a factor/factors that promote(s) fibroblast-mediated contraction of collagen lattices, and stimulate(s) fibroblast glycosaminoglycan (GAG) synthesis. Complete medium from melanoma cell cultures stimulated fibroblast hyaluronate synthesis 9.3-fold, and sulphated GAG synthesis 2.6-fold, as measured by 3H-glucosamine incorporation. 35SO4 incorporation into sulphated GAGS was essentially unaltered, the net result being a decrease in the degree of sulphation. Fibroblasts synthesized hyaluronate with an increased molecular weight when grown in the presence of the melanoma-cell culture medium, while the molecular weights of heparan and chondroitin sulphates remained essentially unaltered. Our results indicate that the tumour-cell-derived factor(s) stimulate(s) changes in fibroblast glycosaminoglycan synthesis, and that these changes may facilitate tumour cell invasion in vivo. Topics: Fibroblasts; Glycosaminoglycans; Humans; Melanoma; Sulfates; Transforming Growth Factor beta; Tumor Cells, Cultured | 1992 |
Degenerative changes in the A375 melanoma line induced by transforming growth factor beta 1.
A375 human melanoma cell cultures grown in the presence of TGF beta contained greatly reduced cell numbers and exhibited drastic alterations in cell morphology compared to the control cultures. Preincubation of the cells with the cytokine for only 18 h was sufficient to induce these changes irreversibly. Examination of TGF beta-treated cells in the electron microscope revealed large numbers of lipid-filled vacuoles in the cytoplasm, greatly contracted nuclei and some loss of the otherwise abundant microvilli. Thus TGF beta may have a direct toxic effect on the A375 melanoma cells. Topics: Cell Count; Cell Division; Dose-Response Relationship, Drug; Humans; Melanoma; Transforming Growth Factor beta; Tumor Cells, Cultured | 1992 |
Characterization of tenascin secreted by human melanoma cells.
Tenascin is a large glycoprotein of the extracellular matrix. It shows a site-restricted expression during embryogenesis and can be found in adult tissues during wound healing and tumorigenesis. Because of the potential involvement of tenascin in adhesion and invasion during metastasis, the study of the interactions of tumor cells with tenascin is of considerable interest. Using five anti-melanoma monoclonal antibodies to four different epitopes of human tenascin, we found that most melanoma cells secrete tenascin in vitro constitutively. Transforming growth factor beta 1 in the medium increased secretion in tenascin-producing cells. Tenascin was present in sera of melanoma patients, with significantly elevated levels in patients with advanced melanomas as compared to patients with low tumor burden or to normal donors. Normal and malignant melanocytes did not attach to tenascin as substrate within 1 to 2 h and tenascin could also inhibit fibronectin-dependent adhesion. These results indicate that tenascin may play a critical role in cell-substrate interactions of melanoma cells. Topics: Antibodies, Monoclonal; Antibody Specificity; Cell Adhesion; Cell Adhesion Molecules, Neuronal; Epitopes; Extracellular Matrix Proteins; Humans; Immunization; Melanocytes; Melanoma; Tenascin; Transforming Growth Factor beta; Tumor Cells, Cultured | 1991 |
Reduced tumor incidence, metastatic potential, and cytokine responsiveness of nm23-transfected melanoma cells.
Reduced expression of the nm23 gene in certain rodent model systems and human breast tumors has been correlated with high tumor metastatic potential. To investigate the functional effects of nm23 expression, we have transfected a constitutive murine nm23-1 expression construct into highly metastatic K-1735 TK murine melanoma cells. TK clones expressing the exogenous nm23-1 construct exhibited a reduced incidence of primary tumor formation, significant reductions in tumor metastatic potential independent of tumor cell growth, and altered responses to the cytokine transforming growth factor beta 1 in soft agar colonization assays, compared with control-transfected TK clones. In contrast, nm23-1-transfected TK clones exhibited no significant differences in intrinsic tumor cell growth, i.e., primary tumor size in vivo, anchorage-dependent growth rate in vitro, and anchorage-independent colony formation in soft agar in vitro. The data demonstrate a suppressive effect of nm23 on several aspects of the cancer process, including tumor metastasis. Topics: Amino Acid Sequence; Animals; Base Sequence; Cloning, Molecular; DNA, Neoplasm; Gene Expression; Genes, Tumor Suppressor; Melanoma; Mice; Molecular Sequence Data; Monomeric GTP-Binding Proteins; Neoplasm Metastasis; Neoplasm Transplantation; NM23 Nucleoside Diphosphate Kinases; Nucleoside-Diphosphate Kinase; Phenotype; Protein Biosynthesis; Proteins; Transcription Factors; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured | 1991 |
Comparison of transforming growth factor beta and a human tumour-derived suppressor factor.
Serum-free supernatants from the human melanoma cell line G361 contain a factor that can potently suppress the generation of tumouricidal lymphokine-activated killer (LAK) cells in response to interleukin-2. To characterise the suppressive factor of tumour origin we performed a number of physicochemical and functional comparisons with another immunosuppressive protein, transforming growth factor beta (TGF beta). The bioactivity of tumour-derived suppressor factor (TDSF), assayed by suppression of LAK cell generation, was unaffected by a reducing agent but lost when denatured with a chaotropic agent. In contrast, TGF beta was inactivated by reduction but not denaturation. TDSF lost bioactivity in conditions of pH less than 4, whereas TGF beta showed no loss of activity. The TDSF moiety has an estimated pI of 4.3 and a molecular mass of 69-87 kDa. This differs from published values of pI 9.5, and 25 kDa molecular mass for TGF beta. Anti-TGF beta antiserum reversed the effects of TGF beta but did not affect the suppression of LAK cell generation caused by TDSF. These findings provide compelling evidence that the TDSF moiety is not TGF beta, and may be a novel immunoregulatory cytokine. Topics: Humans; Hydrogen-Ion Concentration; Isoelectric Point; Killer Cells, Lymphokine-Activated; Melanoma; Molecular Weight; Suppressor Factors, Immunologic; Transforming Growth Factor beta; Tumor Cells, Cultured | 1991 |
Alternative splicing of endothelial cell fibronectin mRNA in the IIICS region. Functional significance.
Transforming growth factor-beta 1 (TGF-beta 1) is thought to play a role in modulating vascular cell function in vivo. In vitro, it decreases endothelial cell proliferation and migration. We postulated that these biologic activities could be mediated through TGF-beta 1 modulation of specific gene expression. Therefore we differentially screened a human umbilical vein endothelial cell cDNA library with cDNAs prepared from both untreated and TGF-beta 1-treated bovine aortic endothelial cells. Using this technique, we isolated many TGF-beta 1-induced cDNA clones. Sequence analysis of these cDNAs showed that many of them corresponded to alternatively spliced fibronectin mRNAs. These fibronectin clones all contained the extradomain I (ED I) but three different forms of the type III connecting segment (IIICS). These different fibronectin cDNAs were expressed in bacteria and the recombinant proteins used to study the effects of IIICS alternative splicing on cell attachment, spreading, and migration in bovine aortic endothelial and smooth muscle cells and B16F10 melanoma cells. The results of these experiments show that attachment and spreading of bovine aortic endothelial and smooth muscle cells depend primarily on the presence of the Arg-Gly-Asp-Ser (RGDS) sequence in the recombinant fibronectin proteins. However attachment and spreading of bovine aortic endothelial cells are modulated by alternative splicing in the IIICS region. Specifically splicing of the IIICS region decreases spreading and increases migration rates of the endothelial cells. On the contrary, using a cell line (B16F10 melanoma cells) that is known not to require the RGDS sequence for adhesion confirmed previous findings that B16F10 melanoma cells do not require the presence of the RGDS sequence for attachment and spreading. Indeed B16F10 cells were able to attach and spread on two recombinant proteins that did not contain the RGDS sequence. However attachment and spreading of B16F10 were dramatically inhibited when a 75-base pair DNA fragment was removed from the 5' end of the IIICS region. These results suggest that various regions of the fibronectin molecule may be able to interact with different cell populations to promote cell attachment and spreading, and that alternative splicing may modulate this process. Topics: Animals; Aorta; Cell Movement; Cloning, Molecular; DNA; Endothelium, Vascular; Fibronectins; Fluorescent Antibody Technique; Gene Library; Integrins; Melanoma; Muscle, Smooth, Vascular; Peptides; Plasmids; Recombinant Proteins; RNA Splicing; RNA, Messenger; Transforming Growth Factor beta; Tumor Cells, Cultured | 1990 |