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

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 449(7164): 933-7, 2007 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17898714

RESUMEN

Eukaryotic genomes are organized into active (euchromatic) and inactive (heterochromatic) chromatin domains. Post-translational modifications of histones (or 'marks') are key in defining these functional states, particularly in promoter regions. Mutual regulatory interactions between these marks--and the enzymes that catalyse them--contribute to the shaping of this epigenetic landscape, in a manner that remains to be fully elucidated. We previously observed that asymmetric di-methylation of histone H3 arginine 2 (H3R2me2a) counter-correlates with di- and tri- methylation of H3 lysine 4 (H3K4me2, H3K4me3) on human promoters. Here we show that the arginine methyltransferase PRMT6 catalyses H3R2 di-methylation in vitro and controls global levels of H3R2me2a in vivo. H3R2 methylation by PRMT6 was prevented by the presence of H3K4me3 on the H3 tail. Conversely, the H3R2me2a mark prevented methylation of H3K4 as well as binding to the H3 tail by an ASH2/WDR5/MLL-family methyltransferase complex. Chromatin immunoprecipitation showed that H3R2me2a was distributed within the body and at the 3' end of human genes, regardless of their transcriptional state, whereas it was selectively and locally depleted from active promoters, coincident with the presence of H3K4me3. Hence, the mutual antagonism between H3R2 and H3K4 methylation, together with the association of MLL-family complexes with the basal transcription machinery, may contribute to the localized patterns of H3K4 tri-methylation characteristic of transcriptionally poised or active promoters in mammalian genomes.


Asunto(s)
Arginina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Epigénesis Genética , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Metilación , Ratones , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , Proteína-Arginina N-Metiltransferasas/genética , Factores de Transcripción/metabolismo
2.
Cancer Res ; 68(3): 749-58, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18245475

RESUMEN

Regulation of chromatin is an important aspect of controlling promoter activity and gene expression. Posttranslational modifications of core histones allow proteins associated with gene transcription to access chromatin. Closely associated with promoters of actively transcribed genes, trimethylation of histone H3 at lysine 4 (H3K4me3) is a core histone mark set by several protein complexes. Some of these protein complexes contain the trithorax protein ASH2 combined with the MLL oncoproteins. We identified human ASH2 in a complex with the oncoprotein MYC. This finding, together with the observation that hASH2 interacts with MLL, led us to test whether hASH2 itself is involved in transformation. We observed that hASH2 cooperates with Ha-RAS to transform primary rat embryo fibroblasts (REF). Furthermore, transformation of REFs by MYC and Ha-RAS required the presence of rAsh2. In an animal model, the hASH2/Ha-RAS-transformed REFs formed rapidly growing tumors characteristic of fibrosarcomas that, compared with tumors derived from MYC/Ha-RAS transformed cells, were poorly differentiated. This finding suggests that ASH2 functions as an oncoprotein. Although hASH2 expression at the mRNA level was generally not deregulated, hASH2 protein expression was increased in most human tumors and tumor cell lines. In addition, knockdown of hASH2 inhibited tumor cell proliferation. Taken together, these observations define hASH2 as a novel oncoprotein.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Neoplasias/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Procesos de Crecimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/biosíntesis , Fibroblastos , Regulación Neoplásica de la Expresión Génica , Genes ras , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Endogámicas F344 , Factores de Transcripción/biosíntesis , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA