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








Base de dados
Intervalo de ano de publicação
1.
Breast Cancer Res ; 20(1): 75, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29996866

RESUMO

The 2013 update of the American Society of Clinical Oncology-College of American Pathologists (ASCO-CAP) human epidermal growth factor receptor 2 (HER2) testing guidelines recommend using an alternative chromosome 17 probe assay to resolve HER2 results determined to be equivocal by immunohistochemistry (IHC) or fluorescence in-situ hybridization (FISH). However, it is unclear if cases considered HER2-positive (HER2+) by the alternative probe method are similar to those classified as HER2+ by traditional IHC and FISH criteria and benefit the same from HER2-targeted therapies. We studied the clinical and pathologic features of all 31 breast cancers classified as HER2+ by the alternative probe method at our institution since 2013 and determined their PAM50 intrinsic molecular subtypes. For comparison, we analyzed 19 consecutive cases that were classified as HER2+ by traditional FISH criteria during the same time period. Thirty (97%) cancers in the alternative probe cohort were estrogen receptor (ER)-positive (ER+), while only 9/19 (47%) of traditional HER2 controls were ER+ (p = 0.0002). Sufficient tissue for intrinsic subtype analysis was available for 20/31 cancers in the alternative probe cohort and 9/19 in the traditional HER2+ group. None (0%) of the 20 alternative probe-positive cases were of the HER2-enriched intrinsic subtype, while 8/9 (89%) of those HER2+ by traditional FISH criteria were HER2-enriched (p = 0.0001). These findings suggest that breast cancers classified as HER2+ only by the alternative probe method are biologically distinct from those classified as HER2+ by traditional criteria, and raises questions as to whether or not they derive the same benefit from HER2-targeted therapies.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/classificação , Neoplasias da Mama/genética , Receptor ErbB-2/genética , Neoplasias da Mama/patologia , Cromossomos Humanos Par 17/genética , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Oncologia , Pessoa de Meia-Idade , Receptores de Estrogênio/genética , Receptores de Progesterona/genética
2.
Cardiovasc Pathol ; 25(3): 258-269, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27039070

RESUMO

There has been an increasing recognition that mitochondrial perturbations play a central role in human heart failure. Mitochondrial networks, whose function is to maintain the regulation of mitochondrial biogenesis, autophagy ('mitophagy') and mitochondrial fusion/fission, are new potential therapeutic targets. Yet our understanding of the molecular underpinning of these processes is just emerging. We recently identified a role of the SWI/SNF ATP-dependent chromatin remodeling complexes in the metabolic homeostasis of the adult cardiomyocyte using cardiomyocyte-specific and inducible deletion of the SWI/SNF ATPases BRG1 and BRM in adult mice (Brg1/Brm double mutant mice). To build upon these observations in early altered metabolism, the present study looks at the subsequent alterations in mitochondrial quality control mechanisms in the impaired adult cardiomyocyte. We identified that Brg1/Brm double-mutant mice exhibited increased mitochondrial biogenesis, increases in 'mitophagy', and alterations in mitochondrial fission and fusion that led to small, fragmented mitochondria. Mechanistically, increases in the autophagy and mitophagy-regulated proteins Beclin1 and Bnip3 were identified, paralleling changes seen in human heart failure. Evidence for perturbed cardiac mitochondrial dynamics included decreased mitochondria size, reduced numbers of mitochondria, and an altered expression of genes regulating fusion (Mfn1, Opa1) and fission (Drp1). We also identified cardiac protein amyloid accumulation (aggregated fibrils) during disease progression along with an increase in pre-amyloid oligomers and an upregulated unfolded protein response including increased GRP78, CHOP, and IRE-1 signaling. Together, these findings described a role for BRG1 and BRM in mitochondrial quality control, by regulating mitochondrial number, mitophagy, and mitochondrial dynamics not previously recognized in the adult cardiomyocyte. As critical to the pathogenesis of heart failure, epigenetic mechanisms like SWI/SNF chromatin remodeling seem more intimately linked to cardiac function and mitochondrial quality control mechanisms than previously realized.


Assuntos
DNA Helicases/metabolismo , Insuficiência Cardíaca/metabolismo , Dinâmica Mitocondrial/fisiologia , Mitofagia/fisiologia , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Insuficiência Cardíaca/patologia , Homeostase/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Miócitos Cardíacos/patologia
3.
PLoS One ; 10(6): e0129280, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26076459

RESUMO

The recent FDA approval of the MiSeqDx platform provides a unique opportunity to develop targeted next generation sequencing (NGS) panels for human disease, including cancer. We have developed a scalable, targeted panel-based assay termed UNCseq, which involves a NGS panel of over 200 cancer-associated genes and a standardized downstream bioinformatics pipeline for detection of single nucleotide variations (SNV) as well as small insertions and deletions (indel). In addition, we developed a novel algorithm, NGScopy, designed for samples with sparse sequencing coverage to detect large-scale copy number variations (CNV), similar to human SNP Array 6.0 as well as small-scale intragenic CNV. Overall, we applied this assay to 100 snap-frozen lung cancer specimens lacking same-patient germline DNA (07-0120 tissue cohort) and validated our results against Sanger sequencing, SNP Array, and our recently published integrated DNA-seq/RNA-seq assay, UNCqeR, where RNA-seq of same-patient tumor specimens confirmed SNV detected by DNA-seq, if RNA-seq coverage depth was adequate. In addition, we applied the UNCseq assay on an independent lung cancer tumor tissue collection with available same-patient germline DNA (11-1115 tissue cohort) and confirmed mutations using assays performed in a CLIA-certified laboratory. We conclude that UNCseq can identify SNV, indel, and CNV in tumor specimens lacking germline DNA in a cost-efficient fashion.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Variação Genética , Genômica , Neoplasias Pulmonares/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/patologia , Biologia Computacional , Variações do Número de Cópias de DNA , Feminino , Estudos de Associação Genética , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Gradação de Tumores , Estadiamento de Neoplasias , Proteínas Proto-Oncogênicas p21(ras)/genética , Reprodutibilidade dos Testes
4.
Oncotarget ; 6(2): 732-45, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25544751

RESUMO

SWI/SNF complexes utilize BRG1 (also known as SMARCA4) or BRM (also known as SMARCA2) as alternative catalytic subunits with ATPase activity to remodel chromatin. These chromatin-remodeling complexes are required for mammalian development and are mutated in ~20% of all human primary tumors. Yet our knowledge of their tumor-suppressor mechanism is limited. To investigate the role of SWI/SNF complexes in the DNA-damage response (DDR), we used shRNAs to deplete BRG1 and BRM and then exposed these cells to a panel of 6 genotoxic agents. Compared to controls, the shRNA knockdown cells were hypersensitive to certain genotoxic agents that cause double-strand breaks (DSBs) associated with stalled/collapsed replication forks but not to ionizing radiation-induced DSBs that arise independently of DNA replication. These findings were supported by our analysis of DDR kinases, which demonstrated a more prominent role for SWI/SNF in the activation of the ATR-Chk1 pathway than the ATM-Chk2 pathway. Surprisingly, γH2AX induction was attenuated in shRNA knockdown cells exposed to a topoisomerase II inhibitor (etoposide) but not to other genotoxic agents including IR. However, this finding is compatible with recent studies linking SWI/SNF with TOP2A and TOP2BP1. Depletion of BRG1 and BRM did not result in genomic instability in a tumor-derived cell line but did result in nucleoplasmic bridges in normal human fibroblasts. Taken together, these results suggest that SWI/SNF tumor-suppressor activity involves a role in the DDR to attenuate replicative stress and genomic instability. These results may also help to inform the selection of chemotherapeutics for tumors deficient for SWI/SNF function.


Assuntos
Dano ao DNA , DNA Helicases/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Neoplasias do Colo do Útero/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , DNA Helicases/deficiência , Feminino , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteínas Nucleares/deficiência , RNA Interferente Pequeno/genética , Fatores de Transcrição/deficiência
5.
Circ Res ; 111(5): e111-22, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22740088

RESUMO

RATIONALE: Mating type switching/sucrose non-fermenting (SWI/SNF) chromatin-remodeling complexes utilize either BRG1 or BRM as a catalytic subunit to alter nucleosome position and regulate gene expression. BRG1 is required for vascular endothelial cell (VEC) development and embryonic survival, whereas BRM is dispensable. OBJECTIVE: To circumvent embryonic lethality and study Brg1 function in adult tissues, we used conditional gene targeting. To evaluate possible Brg1-Brm redundancy, we analyzed Brg1 mutant mice on wild-type and Brm-deficient backgrounds. METHODS AND RESULTS: The inducible Mx1-Cre driver was used to mutate Brg1 in adult mice. These conditional-null mutants exhibited a tissue-specific phenotype and unanticipated functional compensation between Brg1 and Brm. Brg1 single mutants were healthy and had a normal lifespan, whereas Brg1/Brm double mutants exhibited cardiovascular defects and died within 1 month. BRG1 and BRM were required for the viability of VECs but not other cell types where both genes were also knocked out. The VEC phenotype was most evident in the heart, particularly in the microvasculature of the outer myocardium, and was recapitulated in primary cells ex vivo. VEC death resulted in vascular leakage, cardiac hemorrhage, secondary death of cardiomyocytes due to ischemia, and ventricular dissections. CONCLUSIONS: BRG1-catalyzed SWI/SNF complexes are particularly important in cardiovascular tissues. However, in contrast to embryonic development, in which Brm does not compensate, Brg1 is required in adult VECs only when Brm is also mutated. These results demonstrate for the first time that Brm functionally compensates for Brg1 in vivo and that there are significant changes in the relative importance of BRG1- and BRM-catalyzed SWI/SNF complexes during the development of an essential cell lineage.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/metabolismo , Células Endoteliais/metabolismo , Cardiopatias Congênitas/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Catálise , Morte Celular/fisiologia , Linhagem da Célula/fisiologia , Sobrevivência Celular/fisiologia , Proteínas Cromossômicas não Histona/genética , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Vasos Coronários/patologia , DNA Helicases/genética , Ecocardiografia , Células Endoteliais/patologia , Coração/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Homeostase/fisiologia , Camundongos , Camundongos Transgênicos , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Proteínas Nucleares/genética , Derrame Pleural/genética , Derrame Pleural/metabolismo , Derrame Pleural/patologia , Fatores de Transcrição/genética
6.
PLoS One ; 7(2): e31346, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22363625

RESUMO

The BRG1 catalytic subunit of SWI/SNF-related complexes is required for mammalian development as exemplified by the early embryonic lethality of Brg1 null homozygous mice. BRG1 is also a tumor suppressor and, in mice, 10% of heterozygous (Brg1(null/+)) females develop mammary tumors. We now demonstrate that BRG1 mRNA and protein are expressed in both the luminal and basal cells of the mammary gland, raising the question of which lineage requires BRG1 to promote mammary homeostasis and prevent oncogenic transformation. To investigate this question, we utilized Wap-Cre to mutate both Brg1 floxed alleles in the luminal cells of the mammary epithelium of pregnant mice where WAP is exclusively expressed within the mammary gland. Interestingly, we found that Brg1(Wap-Cre) conditional homozygotes lactated normally and did not develop mammary tumors even when they were maintained on a Brm-deficient background. However, Brg1(Wap-Cre) mutants did develop ovarian cysts and uterine tumors. Analysis of these latter tissues showed that both, like the mammary gland, contain cells that normally express Brg1 and Wap. Thus, tumor formation in Brg1 mutant mice appears to be confined to particular cell types that require BRG1 and also express Wap. Our results now show that such cells exist both in the ovary and the uterus but not in either the luminal or the basal compartments of the mammary gland. Taken together, these findings indicate that SWI/SNF-related complexes are dispensable in the luminal cells of the mammary gland and therefore argue against the notion that SWI/SNF-related complexes are essential for cell survival. These findings also suggest that the tumor-suppressor activity of BRG1 is restricted to the basal cells of the mammary gland and demonstrate that this function extends to other female reproductive organs, consistent with recent observations of recurrent ARID1A/BAF250a mutations in human ovarian and endometrial tumors.


Assuntos
Linhagem da Célula , Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Neoplasias Mamárias Animais/prevenção & controle , Proteínas Nucleares/metabolismo , Cistos Ovarianos/prevenção & controle , Fatores de Transcrição/metabolismo , Neoplasias Uterinas/patologia , Neoplasias Uterinas/prevenção & controle , Animais , Apoptose , Transformação Celular Neoplásica/patologia , Células Epiteliais/metabolismo , Feminino , Deleção de Genes , Humanos , Integrases/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas do Leite/metabolismo , Cistos Ovarianos/metabolismo , Cistos Ovarianos/patologia , Fenótipo , Gravidez , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Neoplasias Uterinas/metabolismo
7.
Epigenetics ; 6(12): 1444-53, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22139574

RESUMO

Recent studies have established that two core members of the SWI/SNF chromatin remodeling complex, BRG1 and SNF5/INI1, possess tumor-suppressor activity in human and mouse cancers. While the third core member, BAF155, has been implicated by several studies as having a potential role in tumor development, direct evidence for its tumor suppressor activity has remained lacking. Therefore, we screened for BAF155 deficiency in a large number of human tumor cell lines. We identified 2 cell lines, the SNUC2B colon carcinoma and the SKOV3 ovarian carcinoma, displaying a complete loss of protein expression while maintaining normal levels of mRNA expression. The SKOV3 cell line possesses a heterozygous 4bp deletion that results in an 855AA truncated protein, while the cause of the loss of BAF155 expression in the SNUC2B cell line appears due to a post-transcriptional error. However, the lack of detectable BAF155 expression did not affect sensitivity to RB-mediated cell cycle arrest. Re-expression of full length but not a truncated form of BAF155 in the two cancer cell lines leads to reduced colony forming ability characterized by replicative senescence but not apoptosis. Collectively, these data suggest that loss of BAF155 expression represents another mechanism for inactivation of SWI/SNF complex activity in the development in human cancer. Our results further indicate that the c-terminus proline-glutamine rich domain plays a critical role in the tumor suppressor activity of this protein.


Assuntos
Proteínas Cromossômicas não Histona/genética , Genes Supressores de Tumor , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neoplasias/genética , Fatores de Transcrição/genética , Envelhecimento/genética , Sequência de Aminoácidos , Apoptose , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Dados de Sequência Molecular , Neoplasias Ovarianas/genética , RNA Mensageiro/genética , Deleção de Sequência
8.
J Cell Physiol ; 226(8): 1989-97, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21520050

RESUMO

Eukaryotic organisms package DNA into chromatin for compact storage in the cell nucleus. However, this process promotes transcriptional repression of genes. To overcome the transcriptional repression, chromatin remodeling complexes have evolved that alter the configuration of chromatin packaging of DNA into nucleosomes by histones. The SWI/SNF chromatin remodeling complex uses energy from ATP hydrolysis to reposition nucleosomes and make DNA accessible to transcription factors. Recent studies showing mutations of BRG1, one of two mutually exclusive ATPase subunits, in human tumor cell lines and primary tissue samples have implicated a role for its loss in cancer development. While most of the mutations lead to complete loss of BRG1 protein expression, others result in single amino acid substitutions. To better understand the role of these BRG1 point mutations in cancer development, we characterized SWI/SNF function in human tumor cell lines with these mutations in the absence of BRM expression, the other ATPase component. We found that the mutant BRG1 proteins still interacted with the core complex members and appeared at the promoters of target genes. However, while these mutations did not affect CD44 and CDH1 expression, known targets of the SWI/SNF complex, they did abrogate Rb-mediated cell-cycle arrest. Therefore, our results implicate that these mutations disrupt the de novo chromatin remodeling activity of the complex without affecting the status of existing nucleosome positioning.


Assuntos
Ciclo Celular/genética , DNA Helicases/genética , Proteínas Nucleares/genética , Mutação Puntual , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/genética , Adenosina Trifosfatases/genética , Antígenos CD , Caderinas/metabolismo , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo
9.
Nucleic Acids Res ; 38(20): 6906-19, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20571081

RESUMO

For DNA replication to occur, chromatin must be remodeled. Yet, we know very little about which proteins alter nucleosome occupancy at origins and replication forks and for what aspects of replication they are required. Here, we demonstrate that the BRG1 catalytic subunit of mammalian SWI/SNF-related complexes co-localizes with origin recognition complexes, GINS complexes, and proliferating cell nuclear antigen at sites of DNA replication on extended chromatin fibers. The specific pattern of BRG1 occupancy suggests it does not participate in origin selection but is involved in the firing of origins and the process of replication elongation. This latter function is confirmed by the fact that Brg1 mutant mouse embryos and RNAi knockdown cells exhibit a 50% reduction in replication fork progression rates, which is associated with decreased cell proliferation. This novel function of BRG1 is consistent with its requirement during embryogenesis and its role as a tumor suppressor to maintain genome stability and prevent cancer.


Assuntos
DNA Helicases/fisiologia , Replicação do DNA , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Proliferação de Células , Cromatina/química , DNA Helicases/análise , DNA Helicases/genética , Proteínas de Ligação a DNA/análise , Desenvolvimento Embrionário , Células Eritroides/metabolismo , Células HeLa , Humanos , Camundongos , Mutação , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Fenótipo , Fatores de Transcrição/análise , Fatores de Transcrição/genética
10.
Cancer Res ; 68(24): 10154-62, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19074882

RESUMO

Factors that drive prostate cancer progression remain poorly defined, thus hindering the development of new therapeutic strategies. Disseminated tumors are treated through regimens that ablate androgen signaling, as prostate cancer cells require androgen for growth and survival. However, recurrent, incurable tumors that have bypassed the androgen requirement ultimately arise. This study reveals that the Brm ATPase, a component of selected SWI/SNF complexes, has significant antiproliferative functions in the prostate that protect against these transitions. First, we show that targeted ablation of Brm is causative for the development of prostatic hyperplasia in mice. Second, in vivo challenge revealed that Brm-/- epithelia acquire the capacity for lobe-specific, castration-resistant cellular proliferation. Third, investigation of human specimens revealed that Brm mRNA and protein levels are attenuated in prostate cancer. Fourth, Brm down-regulation was associated with an increased proliferative index, consistent with the mouse model. Lastly, gene expression profiling showed that Brm loss alters factors upstream of E2F1; this was confirmed in murine models, wherein Brm loss induced E2F1 deregulation in a tissue-specific manner. Combined, these data identify Brm as a major effector of serum androgen-induced proliferation in the prostate that is disrupted in human disease, and indicate that loss of Brm confers a proliferative advantage in prostate cancer.


Assuntos
Adenosina Trifosfatases/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Fatores de Transcrição/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Fator de Transcrição E2F1/metabolismo , Humanos , Masculino , Camundongos , Hiperplasia Prostática/enzimologia , Hiperplasia Prostática/patologia , Receptores Androgênicos/metabolismo
11.
J Cell Physiol ; 205(2): 286-94, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15887247

RESUMO

BRG1 and Brahma are critical and mutually exclusive subunits of the multi-constituent SWI/SNF chromatin remodeling complexes. These complexes play a key role in transcriptional regulation by dynamically altering chromatin architecture. Although the two proteins are very similar in structure, murine models demonstrate a clear dichotomy in BRG1/BRM function as heterozygous loss of BRG1 results in tumor development whereas homozygous loss of BRM does not. BRG1 and/or BRM protein is absent or disrupted in approximately 17% of all human adenocarcinomas. Concomitant loss is frequent in non-small cell lung carcinomas and incurs a negative prognosis. The mechanism(s) whereby loss of BRG1 (but apparently not BRM) may contribute to tumor development and/or progression is/are ill defined. In this study, we employ MiaPaCa2, a human pancreatic adenocarcinoma cell line that lacks BRM but retains BRG1 expression to evaluate the impact of BRG1 and BRM individually on growth and tumorigenicity. We show that the MiaPaca2 cell line can apparently tolerate only very low levels of BRM after restoration of stable expression. Reduction of expression of BRG1 via shRNAi in stable clones of MiaPaCa2 results in a marked change in morphology and alterations in actin cytoskeletal organization but does not appear to exert a significant effect on in vitro growth of the cell line. Our results implicate a role for the SWI/SNF complex in the regulation of cellular differentiation.


Assuntos
Actinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , DNA Helicases , Células HeLa , Humanos , Neoplasias Pancreáticas/patologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo
12.
Mol Cell Biol ; 25(6): 2200-15, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15743818

RESUMO

Androgen receptor (AR) activity is required for prostate cancer development and progression. Thus, there is a major impetus to understand the regulation of AR action. We and others have previously shown that AR transactivation potential is dependent on the presence of an active SWI/SNF chromatin remodeling complex. However, the mechanisms underlying SWI/SNF regulation of the AR remained unsolved. We show here that the BAF57 subunit, an accessory component of the remodeling complex, is a critical regulator of AR function. We show that BAF57 is expressed in the luminal epithelia of the prostate and is required for AR-dependent transactivation in prostatic adenocarcinoma cells. Our data reveal that BAF57 can directly bind to the AR and is recruited to endogenous AR targets upon ligand activation. Loss of BAF57 or inhibition of BAF57 function severely compromised AR activity, as observed with both exogenous and endogenous AR targets. Rescue of BAF57 function restored AR activity, thus demonstrating a specific requirement of BAF57 for AR activity. This action of BAF57 proved to be dependent on SWI/SNF ATPase function. BAF57 has previously been implicated in nuclear receptor coactivator function, and we show that, although BAF57 facilitated coactivator activity, only a selected subset required BAF57 for coactivator function. Lastly, we demonstrate that both BAF57 and BRM are required for the proliferation of AR-dependent prostatic adenocarcinoma cells. In summary, these findings identify BAF57 as a critical modulator of the AR that is capable of altering AR activity, coactivator function, and AR-dependent proliferation.


Assuntos
Adenosina Trifosfatases/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/fisiologia , Fatores de Transcrição/metabolismo , Adenocarcinoma/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Transporte/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Montagem e Desmontagem da Cromatina/genética , Montagem e Desmontagem da Cromatina/fisiologia , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA , Epitélio/metabolismo , Inativação Gênica , Ligantes , Masculino , Camundongos , Proteínas Nucleares/fisiologia , Próstata/citologia , Proteínas de Ligação a RNA , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
13.
J Cell Physiol ; 194(3): 303-13, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12548550

RESUMO

The INI1 gene, which encodes a functionally uncharacterized protein component of the hSWI/SNF chromatin remodeling complex, is often mutated or deleted in malignant rhabdoid tumor (MRT). Two isoforms of INI1, that differ by the variable inclusion of nine amino acids, potentially are produced by differential RNA splicing. To determine the effect of the two INI1 isoforms on cell growth, INI1-devoid (MRT) and INI1-expressing cell lines were transfected separately with mammalian expression vectors or transduced with adenoviruses. Transfection of the short form of INI1 into either INI1-deficient or expressing cell lines resulted in complete suppression of cell growth in colony formation assays. The longer splice variant induced moderate to severe growth suppression of MRT cells, but had a far milder effect on non-MRT cells. Transduction of MRT cells with adenoviruses expressing either isoform of INI1 led to a dramatic change in morphology, growth suppression, and cell cycle arrest. Furthermore, senescence-associated proteins were up-regulated after transduction, while levels of proteins implicated in cell cycle progression were down-regulated. Adenoviral delivery of INI1 into a non-MRT cell line, however, had no demonstrable effect on any of these parameters. These results support the genetic evidence that INI1 is a tumor suppressor gene gone awry in MRT cells, and also suggest that delivery of the INI1 gene to MRT cells by adenoviruses may lead to a more effective treatment of this highly aggressive malignancy.


Assuntos
Senescência Celular/fisiologia , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Tumor Rabdoide/genética , Adenoviridae/genética , Biomarcadores , Neoplasias da Mama , Ciclo Celular/fisiologia , Proteínas Cromossômicas não Histona , Proteínas de Ligação a DNA/análise , Imunofluorescência , Genes Supressores de Tumor , Vetores Genéticos , Células HeLa , Humanos , Túbulos Renais Proximais/citologia , Proteína SMARCB1 , Fatores de Transcrição , Ensaio Tumoral de Célula-Tronco
14.
Cancer Chemother Pharmacol ; 49(2): 142-8, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11862428

RESUMO

PURPOSE: We evaluated the in vitro sensitivity of four malignant rhabdoid tumor (MRT) cell lines to six chemotherapeutic agents: 5-fluororuacil, vincristine, carboplatin, doxorubicin, etoposide, and paclitaxel. We also sought to determine whether a defect in the p53 signaling pathway may contribute to the pronounced drug resistance of MRT. METHODS: MRT cells were treated with various concentrations of each drug and the effects on DNA synthesis were quantified using a thymidine incorporation assay. In addition, the effect of various concentrations of doxorubicin on cell growth was evaluated in all four cell lines. Functionality of the p53 pathway was evaluated by incubating cells with carboplatin or doxorubicin and monitoring the effects on the levels of the p53, p21(WAF1/CIP1), and MDM 2 proteins by Western blot analyses. RESULTS: Vincristine (EC(50) 0.5-2.9 n M) and doxorubicin (EC(50) 1.9-5.7 n M) were found to be most effective in inhibiting proliferation and were within clinically relevant concentrations. However, only doxorubicin exhibited cytotoxicity (EC(50) 2.4-13.1 n M), whereas vincristine and the other drugs tested were cytostatic. Interestingly, all four cell lines had remarkably similar dose response curves to all drugs tested, despite the fact that they were derived from different patients and arose in different tissues. When challenged with DNA-damaging drugs, p53 and the downstream effectors, p21(WAF1/CIP1) and MDM 2 were upregulated. CONCLUSIONS: These studies indicate that the p53 pathway is functional and responsive to DNA-damaging drugs, and does not likely contribute to the drug resistance of MRT. The in vitro sensitivity of MRT cells to doxorubicin suggests that it may be a clinically important agent for the treatment of MRT.


Assuntos
Tumor Rabdoide/tratamento farmacológico , Dano ao DNA , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Tumor Rabdoide/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA