Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055045

ABSTRACT

The neural crest transcription factor BRN3A is essential for the proliferation and survival of melanoma cells. It is frequently expressed in melanoma but not in normal melanocytes or benign nevi. The mechanisms underlying the aberrant expression of BRN3A are unknown. Here, we investigated the epigenetic regulation of BRN3A in melanocytes and melanoma cell lines treated with DNA methyltransferase (DNMT), histone acetyltransferase (HAT), and histone deacetylase (HDAC) inhibitors. DNMT and HAT inhibition did not significantly alter BRN3A expression levels, whereas panHDAC inhibition by trichostatin A led to increased expression. Treatment with the isoform-specific HDAC inhibitor mocetinostat, but not with PCI-34051, also increased BRN3A expression levels, suggesting that class I HDACs HDAC1, HDAC2, and HDAC3, and class IV HDAC11, were involved in the regulation of BRN3A expression. Transient silencing of HDACs 1, 2, 3, and 11 by siRNAs revealed that, specifically, HDAC2 inhibition was able to increase BRN3A expression. ChIP-Seq analysis uncovered that HDAC2 inhibition specifically increased H3K27ac levels at a distal enhancer region of the BRN3A gene. Altogether, our data suggest that HDAC2 is a key epigenetic regulator of BRN3A in melanocytes and melanoma cells. These results highlight the importance of epigenetic mechanisms in regulating melanoma oncogenes.


Subject(s)
Gene Expression Regulation , Histone Deacetylase 2/metabolism , Melanocytes/metabolism , Melanoma/etiology , Melanoma/metabolism , Transcription Factor Brn-3A/genetics , Cell Line , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Gene Silencing , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Melanocytes/pathology , Melanoma/pathology , Transcription Factor Brn-3A/metabolism
2.
Oncol Res Treat ; 39(10): 635-642, 2016.
Article in English | MEDLINE | ID: mdl-27710977

ABSTRACT

Melanoma is a common type of skin cancer with a high propensity to metastasize. Tyrosine kinase inhibitors targeting the mitogen-activated protein kinase (MAPK) pathway and immune checkpoint blockade have recently revolutionized the management of unresectable and metastatic disease. However, acquired resistance and primary non-response to therapy require novel treatment strategies and combinations. The purpose of this review is to provide a brief and up-to-date overview on the clinical management and current trial landscape in melanoma. We summarize the most pertinent studies on BRAF/MEK inhibitors and blockade of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1). Although most agents show robust antitumor efficacy as single agents, further improvements have been achieved by the combination of both approved and developing drugs. We discuss ongoing trials and evaluate future approaches that may provide additional efficacy with less toxicity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Melanoma/drug therapy , Molecular Targeted Therapy/methods , Skin Neoplasms/drug therapy , Dose-Response Relationship, Drug , Evidence-Based Medicine , Humans , Melanoma/pathology , Treatment Outcome
3.
Exp Dermatol ; 25(11): 831-838, 2016 11.
Article in English | MEDLINE | ID: mdl-27792246

ABSTRACT

Histone deacetylases (HDACs) are critically involved in epigenetic gene regulation through alterations of the chromatin status of DNA. Aberrant expression, dysregulation of their enzymatic activity or imbalances between HDACs and histone acetyltransferases are likely involved in the development and progression of cancer. Pharmacologic inhibition of HDACs shows potent antitumor activity in a panel of malignancies such as colon or gastric cancer and multiple myeloma. In this review, we summarize the current knowledge of HDACs in melanoma and evaluate the application of HDAC inhibition from an experimental and clinical perspective. The molecular functions of HDACs can be classified into histone and non-histone effects with diverse implications in proliferation, cell cycle progression and apoptosis. HDAC inhibition results in G1 cell cycle arrest, induces apoptosis and increases the immunogenicity of melanoma cells. Some studies proposed that HDAC inhibition may overcome the resistance of melanoma cells to BRAF inhibition. Several inhibitors such as vorinostat, entinostat and valproic acid have recently been tested in phase I and early phase II trials, yet most agents show limited efficacy and tolerability as single agents. The most frequent adverse events of HDAC inhibition comprise haematological toxicity, fatigue, nausea and laboratory abnormalities. Existing evidence supports the hypothesis that HDAC inhibitors (HDACi) may sensitize melanoma cells to immunotherapy and targeted therapy and hence bear therapeutic potential concurrent with immune checkpoint blockade or BRAF and MEK inhibition.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Melanoma/drug therapy , Animals , Clinical Trials as Topic , Histone Deacetylases/classification , Histone Deacetylases/metabolism , Humans , Melanoma/enzymology
4.
J Invest Dermatol ; 136(12): 2475-2484, 2016 12.
Article in English | MEDLINE | ID: mdl-27498344

ABSTRACT

Significant advancements of mutation-based targeted therapy and immune checkpoint blockade have been achieved in melanoma. Nevertheless, acquired resistance and nonresponders to therapy require different strategies. An innovative approach is presented here that is based on the combination of innate immune system activation and simultaneous targeting of the oncogene urokinase-type plasminogen activator receptor (uPAR). We generated two triphosphate-conjugated siRNAs targeting uPAR (ppp-uPAR) by in vitro transcription. Specific uPAR knockdown and simultaneous activation of the retinoic acid-inducible gene 1 (RIG-I) was shown in different human melanoma cells, fibroblasts, and melanocytes. The compounds induced massive apoptosis in melanoma cells, whereas fibroblasts and melanocytes were less sensitive. The effects were less pronounced when the IFN receptor was blocked. Treatment with ppp-uPAR led to accumulation of p53 and induction of RIG-I-dependent proapoptotic signaling. The apoptotic effects induced by ppp-uPAR were maintained in melanoma cell lines that had acquired double resistance to B-RAF and MEK/extracellular signal-regulated kinase inhibition. Systemic intraperitoneal application of ppp-uPAR in nude mice significantly reduced growth of human melanoma xenografts and elicited a systemic innate immune response with increased serum cytokine levels. Our data suggest that ppp-uPAR represents a therapeutically attractive compound that may help overcome the strong therapy resistance of melanoma.


Subject(s)
Immunization/methods , Melanoma/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Skin Neoplasms/genetics , Animals , Apoptosis/genetics , Cell Survival/genetics , Disease Models, Animal , Female , Humans , Melanocytes/cytology , Melanocytes/pathology , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Knockout , Mice, Nude , RNA, Small Interfering/genetics , Random Allocation , Receptors, Cell Surface , Reference Values , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Tumor Cells, Cultured
5.
Curr Opin Oncol ; 27(2): 134-40, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25602684

ABSTRACT

PURPOSE OF REVIEW: Treatment options for metastatic melanoma depend on the clinical course of the disease and the molecular profile such as mutations of the BRAF gene. In this article, we review the current state of targeted therapy with kinase inhibitors. RECENT FINDINGS: Despite major advancements in targeted therapy of metastatic melanoma, most patients relapse and show progressive disease after 5-7 months with single inhibition of BRAF or MEK. Acquired resistance is virtually universal and mediated by diverse mitogen-activated protein kinase-dependent or independent mechanisms. Recent evidence favours concurrent targeting of BRAF and MEK in patients with BRAFV600-mutated melanoma instead of BRAF inhibitor monotherapy. The combination delays the onset of acquired resistance, resulting in increased progression-free and overall survival. A growing number of early trials evaluate the efficacy of inhibitors targeting additional pathways such as phospho-inositide 3-kinase/AKT in conjunction with BRAF or MEK. Even though consistent and mature phase III study results are not yet available for these combinations, the repertoire of targeted therapy in metastatic melanoma is wide and promising. SUMMARY: The short era of single BRAF inhibition in BRAF-mutated melanoma is soon taken over by dual concurrent inhibition of MEK and BRAF.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Combined Modality Therapy , Disease-Free Survival , Drug Resistance, Neoplasm , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/genetics , Mutation/drug effects , Neoplasm Recurrence, Local/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms , Treatment Outcome , Melanoma, Cutaneous Malignant
6.
J Invest Dermatol ; 134(8): 2212-2220, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24608986

ABSTRACT

The transcription factor SOX10 (SRY (sex determining region Y)-box 10) has a key role in the embryonic development of melanocytes. Recently, it has been suggested that SOX10 is highly relevant for melanoma development and survival. However, the distinct functions and downstream targets of SOX10 in melanoma remain widely unknown. In this study, we inhibited SOX10 via RNA interference in different human melanoma cell lines and found a significantly reduced invasion capacity in vitro and in the chick embryo model. At later time points, SOX10 inhibition reduced proliferation and induced cell death. We identified melanoma inhibitory activity (MIA) as a direct target gene of SOX10, which is an essential protein for melanoma cell migration and invasion. Expression levels of SOX10 and MIA strictly correlated in melanoma cell lines, and SOX10 inhibition reduced MIA expression and promoter activity. Direct binding of SOX10 to the MIA promoter was demonstrated by electrophoretic mobility shift assay and chromatin immunoprecipitation. Ectopic expression of MIA in SOX10-inhibited melanoma cells restored the invasion capacity, supporting the hypothesis that MIA is responsible for SOX10-mediated melanoma cell invasion. Our data provide evidence for a critical role of SOX10 in melanoma cell invasion through the regulation of MIA and highlight its role as a therapeutic target in melanoma.


Subject(s)
Extracellular Matrix Proteins/genetics , Melanoma/pathology , Neoplasm Proteins/genetics , SOXE Transcription Factors/physiology , Apoptosis , Cell Line, Tumor , Humans , Neoplasm Invasiveness , Promoter Regions, Genetic , SOXE Transcription Factors/antagonists & inhibitors
7.
PLoS One ; 7(1): e30821, 2012.
Article in English | MEDLINE | ID: mdl-22292048

ABSTRACT

Melanoma is an often fatal form of skin cancer which is remarkably resistant against radio- and chemotherapy. Even new strategies that target RAS/RAF signaling and display unprecedented efficacy are characterized by resistance mechanisms. The targeting of survival pathways would be an attractive alternative strategy, if tumor-specific cell death can be achieved. Bcl-2 proteins play a central role in regulating survival of tumor cells. In this study, we systematically investigated the relevance of antiapoptotic Bcl-2 proteins, i.e., Bcl-2, Bcl-xL, Bcl-w, Mcl-1, and A1, in melanoma cell lines and non-malignant cells using RNAi. We found that melanoma cells required the presence of specific antiapoptotic Bcl-2 proteins: Inhibition of Mcl-1 and A1 strongly induced cell death in some melanoma cell lines, whereas non-malignant cells, i.e., primary human fibroblasts or keratinocytes were not affected. This specific sensitivity of melanoma cells was further enhanced by the combined inhibition of Mcl-1 and A1 and resulted in 60% to 80% cell death in all melanoma cell lines tested. This treatment was successfully combined with chemotherapy, which killed a substantial proportion of cells that survived Mcl-1 and A1 inhibition. Together, these results identify antiapoptotic proteins on which specifically melanoma cells rely on and, thus, provide a basis for the development of new Bcl-2 protein-targeting therapies.


Subject(s)
Melanoma/drug therapy , Melanoma/pathology , Molecular Targeted Therapy/methods , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/genetics , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference/physiology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Skin/cytology , Skin/drug effects , Skin Neoplasms/genetics , Substrate Specificity/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...