Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 284(15): 10067-75, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19144648

ABSTRACT

The mammalian SWI/SNF chromatin-remodeling complex is essential for the multiple changes in gene expression that occur during differentiation. However, the basis within the complex for specificity in effecting positive versus negative changes in gene expression has only begun to be elucidated. The catalytic core of the complex can be either of two closely related ATPases, BRM or BRG1, with the potential that the choice of alternative subunits is a key determinant of specificity. Short hairpin RNA-mediated depletion of the ATPases was used to explore their respective roles in the well characterized multistage process of osteoblast differentiation. The results reveal an unexpected role for BRM-specific complexes. Instead of impeding differentiation as was seen with BRG1 depletion, depletion of BRM caused accelerated progression to the differentiation phenotype. Multiple tissue-specific differentiation markers, including the tightly regulated late stage marker osteocalcin, become constitutively up-regulated in BRM-depleted cells. Chromatin immunoprecipitation analysis of the osteocalcin promoter as a model for the behavior of the complexes indicates that the promoter is a direct target of both BRM- and BRG1-containing complexes. BRG1 complexes, which are required for activation, are associated with the promoter well before induction, but the concurrent presence of BRM-specific complexes overrides their activation function. BRM-specific complexes are present only on the repressed promoter and are required for association of the co-repressor HDAC1. These findings reveal an unanticipated degree of specialization of function linked with the choice of ATPase and suggest a new paradigm for the roles of the alternative subunits during differentiation.


Subject(s)
DNA Helicases/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Cell Differentiation , Chromatin/chemistry , Chromatin Immunoprecipitation , Gene Expression Regulation , Humans , Models, Biological , Osteoblasts/metabolism , Osteocalcin/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Distribution
2.
EMBO J ; 26(3): 752-63, 2007 Feb 07.
Article in English | MEDLINE | ID: mdl-17255939

ABSTRACT

The mammalian SWI/SNF chromatin remodeling complex is becoming increasingly recognized for its role in tumor suppression, based on its ability to regulate accessibility of proliferation-associated genes to transcription factors. However, understanding the biological role of the complex is complicated because the same complex seemingly plays both positive and negative roles in gene expression. Work described here reveals that a choice between two independently encoded, closely related variants of a major subunit of the ARID protein family determines whether the SWI/SNF complex forms further associations with activator versus repressor complexes. The choice distinguishes assemblies with opposite effects on cell-cycle activity. The specific complexes control access of factors such as E2F1, Tip60, and HDAC1/2/3 to the promoters of various cell-cycle-specific genes, with c-Myc emerging as a particularly critical target.


Subject(s)
Cell Cycle/physiology , Chromatin Assembly and Disassembly/physiology , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Protein Subunits/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle/genetics , Cell Line , Chromatin Assembly and Disassembly/genetics , Chromatin Immunoprecipitation , DNA Primers , DNA-Binding Proteins/genetics , E2F Transcription Factors/metabolism , Genes, myc/genetics , Mice , Nuclear Proteins/genetics , Protein Subunits/genetics
3.
Cancer Res ; 66(3): 1289-93, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452181

ABSTRACT

The activity of mammalian SWI/SNF-related chromatin remodeling complexes is crucial for differentiation, development, and tumor suppression. Cell cycle-regulating activities dependent on the complexes include induction of the p21(WAF1/CIP1) kinase inhibitor and repression of E2F-responsive promoters. These responses are linked through effects on pRb phosphorylation, but the direct role of the SWI/SNF-related complexes in their regulation is not fully understood. Results presented here reveal that the complexes are required for regulation of a distinct pathway of proliferation control involving repression of c-myc expression in differentiating cells. This involves direct promoter targeting of the c-myc gene by the complexes. Induction of p21(WAF1/CIP1) is specifically dependent on prior repression of c-myc, but repression of E2F-responsive genes is dissociable from the regulation of c-myc and p21(WAF1/CIP1).


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Genes, myc/physiology , Transcription Factors/physiology , 3T3 Cells , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Growth Processes/genetics , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/deficiency , E2F Transcription Factors/physiology , Gene Expression Regulation/physiology , Mice , Nuclear Proteins/deficiency , Promoter Regions, Genetic
4.
Cancer Res ; 65(20): 9236-44, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230384

ABSTRACT

Mammalian SWI/SNF-related complexes are ATPase-powered nucleosome remodeling assemblies crucial for proper development and tissue-specific gene expression. The ATPase activity of the complexes is also critical for tumor suppression. The complexes contain seven or more noncatalytic subunits; only one of which, hSNF5/Ini1/BAF47, has been individually identified as a tumor suppressor thus far. The noncatalytic subunits include p270/ARID1A, which is of particular interest because tissue array analysis corroborated by screening of tumor cell lines indicates that p270 may be deficient in as many as 30% of renal carcinomas and 10% of breast carcinomas. The complexes can also include an alternative ARID1B subunit, which is closely related to p270, but the product of an independent gene. The respective importance of p270 and ARID1B in the control of cell proliferation was explored here using a short interfering RNA approach and a cell system that permits analysis of differentiation-associated cell cycle arrest. The p270-depleted cells fail to undergo normal cell cycle arrest on induction, as evidenced by continued synthesis of DNA. These lines fail to show other characteristics typical of arrested cells, including up-regulation of p21 and down-regulation of cyclins. The requirement for p270 is evident separately in both the up-regulation of p21 and the down-regulation of E2F-responsive products. In contrast, the ARID1B-depleted lines behaved like the parental cells in these assays. Thus, p270-containing complexes are functionally distinct from ARID1B-containing complexes. These results provide a direct biological basis to support the implication from tumor tissue screens that deficiency of p270 plays a causative role in carcinogenesis.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , 3T3 Cells , Alkaline Phosphatase/biosynthesis , Animals , Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DNA-Binding Proteins/deficiency , E2F Transcription Factors/genetics , Enzyme Induction , Mice , Nuclear Proteins/deficiency , Promoter Regions, Genetic , Transcription Factors
5.
Int J Cancer ; 112(4): 636, 2004 Nov 20.
Article in English | MEDLINE | ID: mdl-15382044

ABSTRACT

Human SWI/SNF complexes use the energy of ATP hydrolysis to remodel chromosomes and alter gene expression patterns. The activity of the complexes generally promotes tissue-specific gene expression and restricts cell proliferation. The ATPase that drives the complexes, BRG1, is essential for tumor suppression in mice and deficient in a variety of established human tumor cell lines. The complex contains at least 7 other core components, one of which is a large subunit designated p270. p270 RNA is expressed in all normal human tissues examined, but protein expression is severely reduced in at least 2 human tumor lines, C33A and T47D. We show here that loss of p270 in the C33A and T47D cell lines is evident at the RNA level as well as the protein level. The implication that p270 can be informatively screened at the RNA level made a high-efficiency cancer profiling array approach to screening human tumors feasible. Expression was screened in an array containing RNA-derived cDNA from 241 tumor and corresponding matched normal tissues from individual patients. p270 deficiency was observed at a higher overall frequency than BRG1 deficiency, but all tissues were not equally affected. Deficiency of p270 was observed most frequently in carcinomas of the breast and kidney. The results were most striking in kidney, where p270 expression was deficient in 30% of carcinoma samples screened. Screening of a panel of established human renal carcinoma-derived cell lines supports the frequency observed in the primary tumor tissue samples.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Renal Cell/genetics , Gene Expression Profiling , Kidney Neoplasms/genetics , Nuclear Proteins/biosynthesis , Transcription Factors/biosynthesis , Cell Division , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins , Humans , Immunoblotting , Oligonucleotide Array Sequence Analysis , RNA/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Biochem J ; 383(Pt 2): 319-25, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15170388

ABSTRACT

p270 (ARID1A) is a member of the ARID family of DNA-binding proteins and a subunit of human SWI/SNF-related complexes, which use the energy generated by an integral ATPase subunit to remodel chromatin. ARID1B is an independent gene product with an open reading frame that is more than 60% identical with p270. We have generated monoclonal antibodies specific for either p270 or ARID1B to facilitate the investigation of ARID1B and its potential interaction with human SWI/SNF complexes in vivo. Immunocomplex analysis provides direct evidence that endogenous ARID1B is associated with SWI/SNF-related complexes and indicates that p270 and ARID1B, similar to the ATPase subunits BRG1 and hBRM, are alternative, mutually exclusive subunits of the complexes. The ARID-containing subunits are not specific to the ATPases. Each associates with both BRG1 and hBRM, thus increasing the number of distinct subunit combinations known to be present in cells. Analysis of the panels of cell lines indicates that ARID1B, similar to p270, has a broad tissue distribution. The ratio of p270/ARID1B in typical cells is approx. 3.5:1, and BRG1 is distributed proportionally between the two ARID subunits. Analysis of DNA-binding behaviour indicates that ARID1B binds DNA in a non-sequence-specific manner similar to p270.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Subunits/metabolism , Transcription Factors/metabolism , Adenosine Triphosphatases/metabolism , Cell Line, Tumor , DNA/metabolism , DNA Helicases , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Substrate Specificity
7.
Biol Reprod ; 70(5): 1419-27, 2004 May.
Article in English | MEDLINE | ID: mdl-14724134

ABSTRACT

One of the most critical events of preimplantation development is the successful activation of gene transcription. Both the timing and the array of genes activated must be controlled. The ability to regulate gene transcription appears to be reduced just prior to the time of the major genome activation event, and changes in chromatin structure appear essential for establishing this ability. Major molecules that modulate chromatin structure are the linker and core histones, enzymes that modify histones, and a wide variety of other factors that associate with DNA and mediate either repressive or activating changes. Among the latter are chromatin accessibility complexes, SWI/SNF complexes, and the YY1 protein and its associated factors. Detailed information about the expression and regulation of these factors in preimplantation stage embryos has not been published for any species. In order to ascertain which of these factors may participate in chromatin remodeling, genome activation, and DNA replication during early primate embryogenesis, we determined the temporal expression patterns of mRNA encoding these factors. Our data identify the predominant members of these different functional classes of factors expressed in oocytes and embryos, and reveal patterns of expression distinct from those patterns seen in somatic cells. Among each of four classes of mRNAs examined, some mRNAs were expressed predominantly in the oocyte, with these largely giving way to others expressed stage specifically in the embryo. This transition may be part of a global mechanism underlying the transition from maternal to embryonic control of development, wherein the oocyte program is silenced and an embryonic pattern of gene expression becomes established. Possible roles for these mRNAs in chromatin remodeling, genome activation, DNA replication, cell lineage determination, and nuclear reprogramming are discussed.


Subject(s)
Blastocyst/physiology , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Developmental , Genome , Macaca mulatta/embryology , Macaca mulatta/genetics , Oocytes/physiology , Acetylation , Animals , Cellular Senescence , Female , Histones/metabolism , RNA, Messenger/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...