Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Nucleic Acids Res ; 45(11): 6442-6458, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28431046

ABSTRACT

Mutations in SOX10 cause neurocristopathies which display varying degrees of hypopigmentation. Using a sensitized mutagenesis screen, we identified Smarca4 as a modifier gene that exacerbates the phenotypic severity of Sox10 haplo-insufficient mice. Conditional deletion of Smarca4 in SOX10 expressing cells resulted in reduced numbers of cranial and ventral trunk melanoblasts. To define the requirement for the Smarca4 -encoded BRG1 subunit of the SWI/SNF chromatin remodeling complex, we employed in vitro models of melanocyte differentiation in which induction of melanocyte-specific gene expression is closely linked to chromatin alterations. We found that BRG1 was required for expression of Dct, Tyrp1 and Tyr, genes that are regulated by SOX10 and MITF and for chromatin remodeling at distal and proximal regulatory sites. SOX10 was found to physically interact with BRG1 in differentiating melanocytes and binding of SOX10 to the Tyrp1 distal enhancer temporally coincided with recruitment of BRG1. Our data show that SOX10 cooperates with MITF to facilitate BRG1 binding to distal enhancers of melanocyte-specific genes. Thus, BRG1 is a SOX10 co-activator, required to establish the melanocyte lineage and promote expression of genes important for melanocyte function.


Subject(s)
Cell Differentiation , DNA Helicases/metabolism , Melanocytes/physiology , Nuclear Proteins/metabolism , SOXE Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Enhancer Elements, Genetic , Gene Expression , Gene Expression Regulation , Melanins/biosynthesis , Membrane Glycoproteins/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Oxidoreductases/genetics
2.
Arch Biochem Biophys ; 563: 125-35, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25026375

ABSTRACT

Brahma (BRM) and Brahma-related gene 1(BRG1) are catalytic subunits of SWItch/sucrose non-fermentable (SWI/SNF) chromatin remodeling complexes. BRM is epigenetically silenced in a wide-range of tumors. Mutations in the v-raf murine sarcoma viral oncogene homolog B1 (BRAF) gene occur frequently in melanoma and lead to constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal regulated kinase (ERK1/2) pathway. We tested the hypothesis that BRM expression is modulated by oncogenic BRAF and phosphorylation of ERK1/2 in melanocytes and melanoma cells. Expression of oncogenic BRAF in melanocytes and melanoma cells that are wild-type for BRAF decreased BRM expression and increased BRG1 expression. Inhibition of mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) or selective inhibition of BRAF in melanoma cells that harbor oncogenic BRAF increased BRM expression and decreased BRG1 expression. Increased BRM expression was associated with increased histone acetylation on the BRM promoter. Over-expression of BRM in melanoma cells that harbor oncogenic BRAF promoted changes in cell cycle progression and apoptosis consistent with a tumor suppressive role. Upon inhibition of BRAF(V600E) with PLX4032, BRM promoted survival. PLX4032 induced changes in BRM function were correlated with increased acetylation of the BRM protein. This study provides insights into the epigenetic consequences of inhibiting oncogenic BRAF in melanoma through modulation of SWI/SNF subunit expression and function.


Subject(s)
MAP Kinase Signaling System , Melanoma/genetics , Melanoma/metabolism , Transcription Factors/genetics , Amino Acid Substitution , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , DNA Helicases/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histones/metabolism , Humans , Melanocytes/cytology , Melanocytes/metabolism , Melanoma/pathology , Mutation , Nuclear Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins B-raf/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/antagonists & inhibitors
3.
Epigenetics Chromatin ; 13(1): 14, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32151278

ABSTRACT

BACKGROUND: Pharmacologic inhibition of bromodomain and extra-terminal (BET) proteins is currently being explored as a new therapeutic approach in cancer. Some studies have also implicated BET proteins as regulators of cell identity and differentiation through their interactions with lineage-specific factors. However, the role of BET proteins has not yet been investigated in melanocyte differentiation. Melanocyte inducing transcription factor (MITF) is the master regulator of melanocyte differentiation, essential for pigmentation and melanocyte survival. In this study, we tested the hypothesis that BET proteins regulate melanocyte differentiation through interactions with MITF. RESULTS: Here we show that chemical inhibition of BET proteins prevents differentiation of unpigmented melanoblasts into pigmented melanocytes and results in de-pigmentation of differentiated melanocytes. BET inhibition also slowed cell growth, without causing cell death, increasing the number of cells in G1. Transcriptional profiling revealed that BET inhibition resulted in decreased expression of pigment-specific genes, including many MITF targets. The expression of pigment-specific genes was also down-regulated in melanoma cells, but to a lesser extent. We found that RNAi depletion of the BET family members, bromodomain-containing protein 4 (BRD4) and bromodomain-containing protein 2 (BRD2) inhibited expression of two melanin synthesis enzymes, TYR and TYRP1. Both BRD4 and BRD2 were detected on melanocyte promoters surrounding MITF-binding sites, were associated with open chromatin structure, and promoted MITF binding to these sites. Furthermore, BRD4 and BRD2 physically interacted with MITF. CONCLUSION: These findings indicate a requirement for BET proteins in the regulation of pigmentation and melanocyte differentiation. We identified changes in pigmentation specific gene expression that occur upon BET inhibition in melanoblasts, melanocytes, and melanoma cells.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation , Melanocytes/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cells, Cultured , HEK293 Cells , Humans , Melanins/biosynthesis , Melanins/genetics , Melanocytes/cytology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription Factors/genetics
4.
Pigment Cell Melanoma Res ; 26(3): 377-91, 2013 May.
Article in English | MEDLINE | ID: mdl-23480510

ABSTRACT

Microphthalmia-associated transcription factor (MITF) is a survival factor in melanocytes and melanoma cells. MITF regulates expression of antiapoptotic genes and promotes lineage-specific survival in response to ultraviolet (UV) radiation and to chemotherapeutics. SWI/SNF chromatin-remodeling enzymes interact with MITF to regulate MITF target gene expression. We determined that the catalytic subunit, BRG1, of the SWI/SNF complex protects melanoma cells against UV-induced death. BRG1 prevents apoptosis in UV-irradiated melanoma cells by activating expression of the melanoma inhibitor of apoptosis (ML-IAP). Down-regulation of ML-IAP compromises BRG1-mediated survival of melanoma cells in response to UV radiation. BRG1 regulates ML-IAP expression by cooperating with MITF to promote transcriptionally permissive chromatin structure on the ML-IAP promoter. The alternative catalytic subunit, BRM, and the BRG1-associated factor, BAF180, were found to be dispensable for elevated expression of ML-IAP in melanoma cells. Thus, we illuminate a lineage-specific mechanism by which a specific SWI/SNF subunit, BRG1, modulates the cellular response to DNA damage by regulating an antiapoptotic gene and implicate this subunit of the SWI/SNF complex in mediating the prosurvival function of MITF.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA Helicases/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , Inhibitor of Apoptosis Proteins/genetics , Melanoma/genetics , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Ultraviolet Rays , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , Chromatin/metabolism , Cytoprotection/radiation effects , DNA-Binding Proteins , Histones/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Models, Biological , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/genetics , Transcription, Genetic/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL