Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Genet ; 49(2): 296-302, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27941798

ABSTRACT

Genes encoding subunits of SWI/SNF (BAF) chromatin-remodeling complexes are collectively mutated in ∼20% of all human cancers. Although ARID1A is the most frequent target of mutations, the mechanism by which its inactivation promotes tumorigenesis is unclear. Here we demonstrate that Arid1a functions as a tumor suppressor in the mouse colon, but not the small intestine, and that invasive ARID1A-deficient adenocarcinomas resemble human colorectal cancer (CRC). These tumors lack deregulation of APC/ß-catenin signaling components, which are crucial gatekeepers in common forms of intestinal cancer. We find that ARID1A normally targets SWI/SNF complexes to enhancers, where they function in coordination with transcription factors to facilitate gene activation. ARID1B preserves SWI/SNF function in ARID1A-deficient cells, but defects in SWI/SNF targeting and control of enhancer activity cause extensive dysregulation of gene expression. These findings represent an advance in colon cancer modeling and implicate enhancer-mediated gene regulation as a principal tumor-suppressor function of ARID1A.


Subject(s)
Colonic Neoplasms/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/genetics , HCT116 Cells , Humans , Mice , Mutation/genetics , beta Catenin/genetics
2.
Elife ; 4: e06073, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25821983

ABSTRACT

ISWI family chromatin remodelers typically organize nucleosome arrays, while SWI/SNF family remodelers (RSC) typically disorganize and eject nucleosomes, implying an antagonism that is largely unexplored in vivo. Here, we describe two independent genetic screens for rsc suppressors that yielded mutations in the promoter-focused ISW1a complex or mutations in the 'basic patch' of histone H4 (an epitope that regulates ISWI activity), strongly supporting RSC-ISW1a antagonism in vivo. RSC and ISW1a largely co-localize, and genomic nucleosome studies using rsc isw1 mutant combinations revealed opposing functions: promoters classified with a nucleosome-deficient region (NDR) gain nucleosome occupancy in rsc mutants, but this gain is attenuated in rsc isw1 double mutants. Furthermore, promoters lacking NDRs have the highest occupancy of both remodelers, consistent with regulation by nucleosome occupancy, and decreased transcription in rsc mutants. Taken together, we provide the first genetic and genomic evidence for RSC-ISW1a antagonism and reveal different mechanisms at two different promoter architectures.


Subject(s)
Adenosine Triphosphatases/genetics , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Nucleosomes/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Adenosine Triphosphatases/metabolism , Binding, Competitive , DNA-Binding Proteins/metabolism , Gene Deletion , Histones/genetics , Histones/metabolism , Multigene Family , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism
3.
Proc Natl Acad Sci U S A ; 111(8): 3128-33, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24520176

ABSTRACT

Defects in epigenetic regulation play a fundamental role in the development of cancer, and epigenetic regulators have recently emerged as promising therapeutic candidates. We therefore set out to systematically interrogate epigenetic cancer dependencies by screening an epigenome-focused deep-coverage design shRNA (DECODER) library across 58 cancer cell lines. This screen identified BRM/SMARCA2, a DNA-dependent ATPase of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling complex, as being essential for the growth of tumor cells that harbor loss of function mutations in BRG1/SMARCA4. Depletion of BRM in BRG1-deficient cancer cells leads to a cell cycle arrest, induction of senescence, and increased levels of global H3K9me3. We further demonstrate the selective dependency of BRG1-mutant tumors on BRM in vivo. Genetic alterations of the mSWI/SNF chromatin remodeling complexes are the most frequent among chromatin regulators in cancers, with BRG1/SMARCA4 mutations occurring in ∼10-15% of lung adenocarcinomas. Our findings position BRM as an attractive therapeutic target for BRG1 mutated cancers. Because BRG1 and BRM function as mutually exclusive catalytic subunits of the mSWI/SNF complex, we propose that such synthetic lethality may be explained by paralog insufficiency, in which loss of one family member unveils critical dependence on paralogous subunits. This concept of "cancer-selective paralog dependency" may provide a more general strategy for targeting other tumor suppressor lesions/complexes with paralogous subunits.


Subject(s)
DNA Helicases/deficiency , Epigenesis, Genetic/physiology , Multiprotein Complexes/genetics , Neoplasms/genetics , Nuclear Proteins/deficiency , Transcription Factors/deficiency , Transcription Factors/genetics , Blotting, Western , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cellular Senescence/genetics , Gene Knockdown Techniques , Gene Library , Histones/metabolism , Humans , Immunoprecipitation , Multiprotein Complexes/metabolism , RNA, Small Interfering/genetics , Transcription Factors/metabolism
4.
Nat Med ; 20(3): 251-4, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24562383

ABSTRACT

Recent studies have revealed that ARID1A, encoding AT-rich interactive domain 1A (SWI-like), is frequently mutated across a variety of human cancers and also has bona fide tumor suppressor properties. Consequently, identification of vulnerabilities conferred by ARID1A mutation would have major relevance for human cancer. Here, using a broad screening approach, we identify ARID1B, an ARID1A homolog whose gene product is mutually exclusive with ARID1A in SWI/SNF complexes, as the number 1 gene preferentially required for the survival of ARID1A-mutant cancer cell lines. We show that loss of ARID1B in ARID1A-deficient backgrounds destabilizes SWI/SNF and impairs proliferation in both cancer cells and primary cells. We also find that ARID1A and ARID1B are frequently co-mutated in cancer but that ARID1A-deficient cancers retain at least one functional ARID1B allele. These results suggest that loss of ARID1A and ARID1B alleles cooperatively promotes cancer formation but also results in a unique functional dependence. The results further identify ARID1B as a potential therapeutic target for ARID1A-mutant cancers.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Mutation , Neoplasms/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Alleles , Animals , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Chromatin/metabolism , False Positive Reactions , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Silencing , HEK293 Cells , Humans , Mice , RNA, Small Interfering/metabolism , Time Factors
5.
Mol Cell Biol ; 34(6): 1136-44, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24421395

ABSTRACT

Collectively, genes encoding subunits of the SWI/SNF (BAF) chromatin remodeling complex are mutated in 20% of all human cancers, with the SMARCA4 (BRG1) subunit being one of the most frequently mutated. The SWI/SNF complex modulates chromatin remodeling through the activity of two mutually exclusive catalytic subunits, SMARCA4 and SMARCA2 (BRM). Here, we show that a SMARCA2-containing residual SWI/SNF complex underlies the oncogenic activity of SMARCA4 mutant cancers. We demonstrate that a residual SWI/SNF complex exists in SMARCA4 mutant cell lines and plays essential roles in cellular proliferation. Further, using data from loss-of-function screening of 165 cancer cell lines, we identify SMARCA2 as an essential gene in SMARCA4 mutant cancer cell lines. Mechanistically, we reveal that Smarca4 inactivation leads to greater incorporation of the nonessential SMARCA2 subunit into the SWI/SNF complex. Collectively, these results reveal a role for SMARCA2 in oncogenesis caused by SMARCA4 loss and identify the ATPase and bromodomain-containing SMARCA2 as a potential therapeutic target in these cancers.


Subject(s)
Carcinogenesis/genetics , DNA Helicases/genetics , Mutation/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Carcinogenesis/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , Fibroblasts/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Transcription Factors/metabolism
6.
Proc Natl Acad Sci U S A ; 110(25): 10165-70, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23723349

ABSTRACT

Precise nucleosome-positioning patterns at promoters are thought to be crucial for faithful transcriptional regulation. However, the mechanisms by which these patterns are established, are dynamically maintained, and subsequently contribute to transcriptional control are poorly understood. The switch/sucrose non-fermentable chromatin remodeling complex, also known as the Brg1 associated factors complex, is a master developmental regulator and tumor suppressor capable of mobilizing nucleosomes in biochemical assays. However, its role in establishing the nucleosome landscape in vivo is unclear. Here we have inactivated Snf5 and Brg1, core subunits of the mammalian Swi/Snf complex, to evaluate their effects on chromatin structure and transcription levels genomewide. We find that inactivation of either subunit leads to disruptions of specific nucleosome patterning combined with a loss of overall nucleosome occupancy at a large number of promoters, regardless of their association with CpG islands. These rearrangements are accompanied by gene expression changes that promote cell proliferation. Collectively, these findings define a direct relationship between chromatin-remodeling complexes, chromatin structure, and transcriptional regulation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Promoter Regions, Genetic/physiology , Transcription Factors/metabolism , Animals , Cell Proliferation , Chromatin/physiology , Chromosomal Proteins, Non-Histone/genetics , CpG Islands/physiology , DNA Helicases/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nucleosomes/genetics , Primary Cell Culture , Protein Binding/physiology , SMARCB1 Protein , Transcription Factors/genetics , Transcriptional Activation/physiology
7.
J Clin Invest ; 121(10): 3834-45, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926465

ABSTRACT

A fundamental goal in cancer research is the identification of the cell types and signaling pathways capable of initiating and sustaining tumor growth, as this has the potential to reveal therapeutic targets. Stem and progenitor cells have been implicated in the genesis of select lymphoid malignancies. However, the identity of the cells in which mature lymphoid neoplasms are initiated remains unclear. Here, we investigate the origin of peripheral T cell lymphomas using mice in which Snf5, a chromatin remodelling-complex subunit with tumor suppressor activity, could be conditionally inactivated in developing T cells. In this model of mature peripheral T cell lymphomas, the cell of origin was a mature CD44hiCD122loCD8⁺ T cell that resembled a subset of memory cells that has capacity for self-renewal and robust expansion, features shared with stem cells. Further analysis showed that Snf5 loss led to activation of a Myc-driven signaling network and stem cell transcriptional program. Finally, lymphomagenesis and lymphoma proliferation depended upon TCR signaling, establishing what we believe to be a new paradigm for lymphoid malignancy growth. These findings suggest that the self-renewal and robust proliferative capacities of memory T cells are associated with vulnerability to oncogenic transformation. Our findings further suggest that agents that impinge upon TCR signaling may represent an effective therapeutic modality for this class of lethal human cancers.


Subject(s)
Cell Transformation, Neoplastic/immunology , Immunologic Memory , Lymphoma, T-Cell, Peripheral/etiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , Cell Differentiation/immunology , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Gene Deletion , Humans , Lymphoma, T-Cell, Peripheral/immunology , Lymphoma, T-Cell, Peripheral/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , SMARCB1 Protein , Signal Transduction
8.
Nat Rev Cancer ; 11(7): 481-92, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21654818

ABSTRACT

SWI/SNF chromatin remodelling complexes use the energy of ATP hydrolysis to remodel nucleosomes and to modulate transcription. Growing evidence indicates that these complexes have a widespread role in tumour suppression, as inactivating mutations in several SWI/SNF subunits have recently been identified at a high frequency in a variety of cancers. However, the mechanisms by which mutations in these complexes drive tumorigenesis are unclear. In this Review we discuss the contributions of SWI/SNF mutations to cancer formation, examine their normal functions and discuss opportunities for novel therapeutic interventions for SWI/SNF-mutant cancers.


Subject(s)
Chromatin Assembly and Disassembly , Neoplasms/etiology , Nucleosomes/physiology , Transcription Factors/physiology , Animals , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/physiology , DNA Helicases/genetics , DNA Helicases/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Humans , Mutation , Neoplasms/genetics , Nuclear Proteins/genetics , Nuclear Proteins/physiology , SMARCB1 Protein , Transcription Factors/genetics
9.
Cancer Cell ; 18(4): 316-28, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20951942

ABSTRACT

Epigenetic alterations have been increasingly implicated in oncogenesis. Analysis of Drosophila mutants suggests that Polycomb and SWI/SNF complexes can serve antagonistic developmental roles. However, the relevance of this relationship to human disease is unclear. Here, we have investigated functional relationships between these epigenetic regulators in oncogenic transformation. Mechanistically, we show that loss of the SNF5 tumor suppressor leads to elevated expression of the Polycomb gene EZH2 and that Polycomb targets are broadly H3K27-trimethylated and repressed in SNF5-deficient fibroblasts and cancers. Further, we show antagonism between SNF5 and EZH2 in the regulation of stem cell-associated programs and that Snf5 loss activates those programs. Finally, using conditional mouse models, we show that inactivation of Ezh2 blocks tumor formation driven by Snf5 loss.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomal Proteins, Non-Histone/genetics , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Repressor Proteins/metabolism , Animals , Cell Line, Tumor , Cell Lineage , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Embryo, Mammalian/cytology , Enhancer of Zeste Homolog 2 Protein , Fibroblasts/metabolism , Gene Silencing , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mice , Models, Genetic , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , SMARCB1 Protein , Stem Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcription, Genetic , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL