Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Carcinog ; 48(12): 1139-48, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19676100

RESUMEN

Malignant rhabdoid tumors (MRTs) are poorly differentiated pediatric cancers that arise in various anatomical locations and have a very poor outcome. The large majority of these malignancies are caused by loss of function of the SNF5/INI1 component of the SWI/SNF chromatin remodeling complex. However, the mechanism of tumor development associated with SNF5 loss remains unclear. Multiple studies have demonstrated a role for SNF5 in the regulation of cyclin D1, p16(INK4A), and pRb(f) activities suggesting it functions through the SWI/SNF complex to affect transcription of genes involved in cell cycle control. Previous studies in genetically engineered mouse models (GEMM) have shown that loss of SNF5 on a p53-null background significantly accelerates tumor development. Here, we use established GEMM to further define the relationship between the SNF5 and p53 tumor suppressor pathways. Combined haploinsufficiency of p53 and Snf5 leads to decreased latency for MRTs arising in alternate anatomical locations but not for the standard facial MRTs. We also observed acceleration in the appearance of T-cell lymphomas in the p53(+/-);Snf5(+/-) mice. Our studies suggest that loss of SNF5 activity does not bestow a selective advantage on the p53 spectrum of tumors in the p53(+/-);Snf5(+/-) mice. However, reduced p53 expression specifically accelerated the growth of a subset of MRTs in these mice.


Asunto(s)
Neoplasias Óseas/patología , Proteínas Cromosómicas no Histona/fisiología , Linfoma de Células T/patología , Osteosarcoma/patología , Tumor Rabdoide/patología , Proteína p53 Supresora de Tumor/fisiología , Animales , Neoplasias Óseas/genética , Proteínas de Ciclo Celular/metabolismo , Femenino , Linfoma de Células T/genética , Masculino , Ratones , Ratones Noqueados , Osteosarcoma/genética , Tumor Rabdoide/genética , Proteína SMARCB1 , Tasa de Supervivencia
2.
Epigenetics ; 6(12): 1444-53, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22139574

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Genes Supresores de Tumor , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neoplasias/genética , Factores de Transcripción/genética , Envejecimiento/genética , Secuencia de Aminoácidos , Apoptosis , Línea Celular Tumoral , Neoplasias del Colon/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Neoplasias Ováricas/genética , ARN Mensajero/genética , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA