Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Cycle ; 11(10): 1956-65, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22544318

ABSTRACT

Emerging evidence demonstrates that subunits of the SWI/SNF chromatin remodeling complex are specifically mutated at high frequency in a variety of human cancer types. SNF5 (SMARCB1/INI1/BAF47), a core subunit of the SWI/SNF complex, is inactivated in the vast majority of rhabdoid tumors (RT), an aggressive type of pediatric cancer. SNF5-deficient cancers are diploid and genomically stable, suggesting that epigenetically based changes in transcription are key drivers of tumor formation caused by SNF5 loss. However, there is limited understanding of the target genes that drive cancer formation following SNF5 loss. Here we performed comparative expression analyses upon three independent SNF5-deficient cancer data sets from both human and mouse and identify downregulation of the BIN1 tumor suppressor as a conserved event in primary SNF5-deficient cancers. We show that SNF5 recruits the SWI/SNF complex to the BIN1 promoter, and that the marked reduction of BIN1 expression in RT correlates with decreased SWI/SNF occupancy. Functionally, we demonstrate that re-expression of BIN1 specifically compromises the proliferation of SNF5-deficient RT cell lines. Identification of BIN1 as a SNF5 target gene reveals a novel tumor suppressive regulatory mechanism whose disruption can drive cancer formation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Proliferation , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Down-Regulation , Humans , Mice , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA Splicing , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , SMARCB1 Protein , Transcription Factors/deficiency , Transcription Factors/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics
2.
Nat Med ; 16(12): 1429-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21076395

ABSTRACT

Aberrant activation of the Hedgehog (Hh) pathway can drive tumorigenesis. To investigate the mechanism by which glioma-associated oncogene family zinc finger-1 (GLI1), a crucial effector of Hh signaling, regulates Hh pathway activation, we searched for GLI1-interacting proteins. We report that the chromatin remodeling protein SNF5 (encoded by SMARCB1, hereafter called SNF5), which is inactivated in human malignant rhabdoid tumors (MRTs), interacts with GLI1. We show that Snf5 localizes to Gli1-regulated promoters and that loss of Snf5 leads to activation of the Hh-Gli pathway. Conversely, re-expression of SNF5 in MRT cells represses GLI1. Consistent with this, we show the presence of a Hh-Gli-activated gene expression profile in primary MRTs and show that GLI1 drives the growth of SNF5-deficient MRT cells in vitro and in vivo. Therefore, our studies reveal that SNF5 is a key mediator of Hh signaling and that aberrant activation of GLI1 is a previously undescribed targetable mechanism contributing to the growth of MRT cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Rhabdoid Tumor/genetics , Signal Transduction/genetics , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling , Humans , Immunoblotting , In Situ Hybridization , Mass Spectrometry , Mice , Microarray Analysis , SMARCB1 Protein , Transcription Factors/genetics , Zinc Finger Protein GLI1
3.
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
4.
Cell Cycle ; 8(1): 23-6, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19098432

ABSTRACT

Genomic instability is often considered a hallmark of cancer. However, it remains unclear whether chromosomal disorganization is most frequently a cause or a consequence of tumorigenesis. The fact that subsets of many cancers lack chromosomal or microsatellite instability argues against the hypothesis that genomic instability plays an essential role in the initiation and maintenance of oncogenesis. Cancer-specific, epigenetically-based changes in gene expression caused by abnormalities in DNA methylation, in histone modifications, and in nucleosome positioning are gaining recognition as driving events in tumorigenesis. But are these changes merely a small piece of the oncogenesis puzzle or could they be the major component, or even sufficient for cancer? Here we discuss evidence that the progression of aggressive cancers can be driven by epigenetic events without genomic instability.


Subject(s)
Epigenesis, Genetic , Genomic Instability/genetics , Neoplasms/genetics , Animals , DNA Methylation , Histones/metabolism , Humans
5.
Mol Cell Biol ; 28(20): 6223-33, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710953

ABSTRACT

There is a growing appreciation of the role that epigenetic alterations can play in oncogenesis. However, given the large number of genetic anomalies present in most cancers, it has been difficult to evaluate the extent to which epigenetic changes contribute to cancer. SNF5 (INI1/SMARCB1/BAF47) is a tumor suppressor that regulates the epigenome as a core member of the SWI/SNF chromatin remodeling complex. While the SWI/SNF complex displays potent tumor suppressor activity, it is unknown whether this activity is exerted genetically via maintenance of genome integrity or epigenetically via transcriptional regulation. Here we show that Snf5-deficient primary cells do not show altered sensitivity to DNA damaging agents, defects in gamma-H2AX induction, or an abrogated DNA damage checkpoint. Further, the aggressive malignancies that arise following SNF5 loss are diploid and genomically stable. Remarkably, we demonstrate that most human SNF5-deficient cancers lack genomic amplifications/deletions and, aside from SNF5 loss, are indistinguishable from normal cells on single-nucleotide polymorphism arrays. Finally, we show that epigenetically based changes in transcription that occur following SNF5 loss correlate with the tumor phenotype. Collectively, our results provide novel insight into the mechanisms of oncogenesis by demonstrating that disruption of a chromatin remodeling complex can largely, if not completely, substitute for genomic instability in the genesis of aggressive cancer.


Subject(s)
Chromosomal Proteins, Non-Histone/deficiency , DNA-Binding Proteins/deficiency , Epigenesis, Genetic , Genomic Instability , Neoplasms/genetics , Transcription Factors/deficiency , Animals , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Cisplatin/pharmacology , Cyclin D1/metabolism , DNA Damage , DNA Repair/drug effects , DNA-Binding Proteins/metabolism , Diploidy , Epigenesis, Genetic/drug effects , Etoposide/pharmacology , G2 Phase/drug effects , Genomic Instability/drug effects , Histones/metabolism , Humans , Mice , Phenotype , Polymorphism, Single Nucleotide/genetics , Protein Transport/drug effects , Rhabdoid Tumor/genetics , SMARCB1 Protein , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL