Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
BMC Cancer ; 23(1): 775, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596538

RESUMEN

BACKGROUND: Inhibitors of Poly (ADP-Ribose) Polymerases (PARP) provide clinical benefit to patients with breast and ovarian cancers, by compromising the DNA repair activity of cancer cells. Although these agents extend progression-free survival in many patients, responses can be short lived with many patients ultimately progressing. Identification of combination partners that increase dependence of cancer cells to the DNA repair activity of PARPs may represent a strategy to increase the utility of PARP inhibitors. Protein arginine methyltransferase 5 (PRMT5) regulates DNA damage response pathways through splicing and protein modification, and inhibitors of PRMT5 have recently entered clinical trials. METHODS: The effect of PRMT5 inhibition on the levels of DNA damage and repair markers including γH2AX, RAD51, and 53BP1 was determined using high content immunofluorescent imaging. The anti-proliferative activity of the combination of PRMT5 and PARP inhibitors was evaluated using in vitro models of breast and ovarian cancers using both cell lines and ex vivo patient derived xenografts. Finally, the combinations of PRMT5 and PARP inhibitors were evaluated in cell line xenograft models in vivo. RESULTS: Inhibition of PRMT5 by GSK3326595 led to increased levels of markers of DNA damage. The addition of GSK3326595 to the PARP inhibitor, niraparib, resulted in increased growth inhibition of breast and ovarian cancer cell lines and patient derived spheroids. In vivo, the combination improved the partial effects on tumor growth inhibition achieved by either single agent, producing complete tumor stasis and regression. CONCLUSION: These data demonstrate that inhibition of PRMT5 induced signatures of DNA damage in models of breast and ovarian cancer. Furthermore, combination with the PARP inhibitor, Niraparib, resulted in increased anti-tumor activity in vitro and in vivo. Overall, these data suggest inhibition of PRMT5 as a mechanism to broaden and enhance the clinical application of PARP inhibitors.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Humanos , Femenino , Animales , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Daño del ADN , Modelos Animales de Enfermedad , Proteína-Arginina N-Metiltransferasas
2.
Nat Genet ; 49(2): 296-302, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27941798

RESUMEN

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.


Asunto(s)
Neoplasias del Colon/genética , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Células HCT116 , Humanos , Ratones , Mutación/genética , beta Catenina/genética
3.
Elife ; 4: e06073, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25821983

RESUMEN

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.


Asunto(s)
Adenosina Trifosfatasas/genética , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/genética , Regulación Fúngica de la Expresión Génica , Nucleosomas/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Adenosina Trifosfatasas/metabolismo , Unión Competitiva , Proteínas de Unión al ADN/metabolismo , Eliminación de Gen , Histonas/genética , Histonas/metabolismo , Familia de Multigenes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Regiones Promotoras Genéticas , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo
4.
Nat Med ; 20(3): 251-4, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24562383

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Alelos , Animales , Línea Celular , Núcleo Celular/metabolismo , Proliferación Celular , Cromatina/metabolismo , Reacciones Falso Positivas , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen , Células HEK293 , Humanos , Ratones , ARN Interferente Pequeño/metabolismo , Factores de Tiempo
5.
Proc Natl Acad Sci U S A ; 111(8): 3128-33, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24520176

RESUMEN

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.


Asunto(s)
ADN Helicasas/deficiencia , Epigénesis Genética/fisiología , Complejos Multiproteicos/genética , Neoplasias/genética , Proteínas Nucleares/deficiencia , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Western Blotting , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Senescencia Celular/genética , Técnicas de Silenciamiento del Gen , Biblioteca de Genes , Histonas/metabolismo , Humanos , Inmunoprecipitación , Complejos Multiproteicos/metabolismo , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo
6.
Mol Cell Biol ; 34(6): 1136-44, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24421395

RESUMEN

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.


Asunto(s)
Carcinogénesis/genética , ADN Helicasas/genética , Mutación/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Carcinogénesis/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Ensamble y Desensamble de Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Fibroblastos/metabolismo , Humanos , Ratones , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
7.
Proc Natl Acad Sci U S A ; 110(25): 10165-70, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23723349

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Regiones Promotoras Genéticas/fisiología , Factores de Transcripción/metabolismo , Animales , Proliferación Celular , Cromatina/fisiología , Proteínas Cromosómicas no Histona/genética , Islas de CpG/fisiología , ADN Helicasas/genética , Fibroblastos/citología , Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Nucleosomas/genética , Cultivo Primario de Células , Unión Proteica/fisiología , Proteína SMARCB1 , Factores de Transcripción/genética , Activación Transcripcional/fisiología
8.
J Clin Invest ; 121(10): 3834-45, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21926465

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Memoria Inmunológica , Linfoma de Células T Periférico/etiología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/patología , Animales , Diferenciación Celular/inmunología , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/genética , Eliminación de Gen , Humanos , Linfoma de Células T Periférico/inmunología , Linfoma de Células T Periférico/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína SMARCB1 , Transducción de Señal
9.
Nat Rev Cancer ; 11(7): 481-92, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-21654818

RESUMEN

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.


Asunto(s)
Ensamble y Desensamble de Cromatina , Neoplasias/etiología , Nucleosomas/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/fisiología , ADN Helicasas/genética , ADN Helicasas/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Humanos , Mutación , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Proteína SMARCB1 , Factores de Transcripción/genética
10.
Nat Med ; 16(12): 1429-33, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21076395

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Tumor Rabdoide/genética , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/genética , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Hibridación in Situ , Espectrometría de Masas , Ratones , Análisis por Micromatrices , Proteína SMARCB1 , Factores de Transcripción/genética , Proteína con Dedos de Zinc GLI1
11.
Cancer Cell ; 18(4): 316-28, 2010 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-20951942

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas Cromosómicas no Histona/genética , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/genética , Proteínas Represoras/metabolismo , Animales , Línea Celular Tumoral , Linaje de la Célula , Proliferación Celular , Transformación Celular Neoplásica/patología , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Embrión de Mamíferos/citología , Proteína Potenciadora del Homólogo Zeste 2 , Fibroblastos/metabolismo , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilación , Ratones , Modelos Genéticos , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Proteína SMARCB1 , Células Madre/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Transcripción Genética , Regulación hacia Arriba/genética
12.
Mol Cell ; 22(5): 633-44, 2006 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-16762836

RESUMEN

Nutrient deprivation and various stress conditions repress RNA polymerase III (Pol III) transcription in S. cerevisiae. The signaling pathways that relay stress and nutrient conditions converge on the conserved protein Maf1, but how Maf1 integrates environmental conditions and couples them to transcriptional repression is largely unknown. Here, we demonstrate that Maf1 is phosphorylated in favorable conditions, whereas diverse unfavorable conditions lead to rapid Maf1 dephosphorylation, nuclear localization, physical association of dephosphorylated Maf1 with Pol III, and Maf1 targeting to Pol III-transcribed genes genome wide. Furthermore, Maf1 mutants defective in full dephosphorylation display maf1Delta phenotypes and are compromised for both nuclear localization and Pol III association. Repression conditions also promote TFIIIB-TFIIIC interactions in crosslinked chromatin. Taken together, Maf1 appears to integrate environmental conditions and signaling pathways through its phosphorylation state, with stress leading to dephosphorylation, association with Pol III at target loci, alterations in basal factor interactions, and transcriptional repression.


Asunto(s)
ARN Polimerasa III/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , Represión Enzimática/genética , Represión Enzimática/fisiología , Regulación Fúngica de la Expresión Génica , Genoma Fúngico , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Mutación Puntual , ARN Polimerasa III/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética
13.
Genetics ; 172(2): 795-809, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16204215

RESUMEN

RSC is an essential and abundant ATP-dependent chromatin remodeling complex from Saccharomyces cerevisiae. Here we show that the RSC components Rsc7/Npl6 and Rsc14/Ldb7 interact physically and/or functionally with Rsc3, Rsc30, and Htl1 to form a module important for a broad range of RSC functions. A strain lacking Rsc7 fails to properly assemble RSC, which confers sensitivity to temperature and to agents that cause DNA damage, microtubule depolymerization, or cell wall stress (likely via transcriptional misregulation). Cells lacking Rsc14 display sensitivity to cell wall stress and are deficient in the assembly of Rsc3 and Rsc30. Interestingly, certain rsc7delta and rsc14delta phenotypes are suppressed by an increased dosage of Rsc3, an essential RSC member with roles in cell wall integrity and spindle checkpoint pathways. Thus, Rsc7 and Rsc14 have different roles in the module as well as sharing physical and functional connections to Rsc3. Using a genetic array of nonessential null mutations (SGA) we identified mutations that are sick/lethal in combination with the rsc7delta mutation, which revealed connections to a surprisingly large number of chromatin remodeling complexes and cellular processes. Taken together, we define a protein module on the RSC complex with links to a broad spectrum of cellular functions.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/fisiología , Factores de Transcripción/fisiología , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Regulación Fúngica de la Expresión Génica/fisiología , Ligamiento Genético , Mutación , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Huso Acromático/fisiología , Factores de Transcripción/genética
14.
Mol Cell ; 9(3): 563-73, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11931764

RESUMEN

The cellular response to environmental changes includes widespread modifications in gene expression. Here we report the identification and characterization of Rsc9, a member of the RSC chromatin-remodeling complex in yeast. The genome-wide localization of Rsc9 indicated a relationship between genes targeted by Rsc9 and genes regulated by stress; treatment with hydrogen peroxide or rapamycin, which inhibits TOR signaling, resulted in genome-wide changes in Rsc9 occupancy. We further show that Rsc9 is involved in both repression and activation of mRNAs regulated by TOR as well as the synthesis of rRNA. Our results illustrate the response of a chromatin-remodeling factor to signaling cascades and suggest that changes in the activity of chromatin-remodeling factors are reflected in changes in their localization in the genome.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Fúngica de la Expresión Génica , Genoma Fúngico , Fosfatidilinositol 3-Quinasas , Proteínas de Saccharomyces cerevisiae , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Levaduras/genética , Secuencia de Aminoácidos , Animales , Antifúngicos/farmacología , Separación Celular , Cromatina/metabolismo , Proteínas de Unión al ADN/química , Citometría de Flujo , Proteínas Fúngicas/metabolismo , Peróxido de Hidrógeno/farmacología , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidantes/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Sirolimus/farmacología , Factores de Transcripción/química , Levaduras/efectos de los fármacos , Levaduras/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...