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
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
J Virol ; 86(9): 5165-78, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22357270

RESUMEN

Epstein-Barr virus (EBV) establishes a persistent latent infection in B lymphocytes and is associated with the development of numerous human tumors. Epstein-Barr nuclear antigen 3C (EBNA 3C) is essential for B-cell immortalization, has potent cell cycle deregulation capabilities, and functions as a regulator of both viral- and cellular-gene expression. We performed transcription profiling on EBNA 3C-expressing B cells and identified several chemokines and members of integrin receptor-signaling pathways, including CCL3, CCL4, CXCL10, CXCL11, ITGA4, ITGB1, ADAM28, and ADAMDEC1, as cellular target genes that could be repressed by the action of EBNA 3C alone. Chemotaxis assays demonstrated that downregulation of CXCL10 and -11 by EBNA 3C is sufficient to reduce the migration of cells expressing the CXCL10 and -11 receptor CXCR3. Gene repression by EBNA 3C was accompanied by decreased histone H3 lysine 9/14 acetylation and increased histone H3 lysine 27 trimethylation. In an EBV-positive cell line expressing all latent genes, we identified binding sites for EBNA 3C at ITGB1 and ITGA4 and in a distal regulatory region between ADAMDEC1 and ADAM28, providing the first demonstration of EBNA 3C association with cellular-gene control regions. Our data implicate indirect mechanisms in CXCL10 and CXCL11 repression by EBNA 3C. In summary, we have unveiled key cellular pathways repressed by EBNA 3C that are likely to contribute to the ability of EBV-immortalized cells to modulate immune responses, adhesion, and B-lymphocyte migration to facilitate persistence in the host.


Asunto(s)
Antígenos Virales/metabolismo , Regulación hacia Abajo/genética , Integrinas/genética , Regiones Promotoras Genéticas , Transducción de Señal , Proteínas ADAM/genética , Animales , Sitios de Unión , Adhesión Celular/genética , Línea Celular , Inhibición de Migración Celular/genética , Quimiocinas/genética , Quimiotaxis/genética , Antígenos Nucleares del Virus de Epstein-Barr , Regulación de la Expresión Génica , Humanos , Ratones , Receptores CXCR3/metabolismo , Elementos Reguladores de la Transcripción
2.
PLoS One ; 6(12): e28638, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22163048

RESUMEN

Epstein-Barr virus (EBV) is implicated in the pathogenesis of multiple human tumours of lymphoid and epithelial origin. The virus infects and immortalizes B cells establishing a persistent latent infection characterized by varying patterns of EBV latent gene expression (latency 0, I, II and III). The CDK1 activator, Response Gene to Complement-32 (RGC-32, C13ORF15), is overexpressed in colon, breast and ovarian cancer tissues and we have detected selective high-level RGC-32 protein expression in EBV-immortalized latency III cells. Significantly, we show that overexpression of RGC-32 in B cells is sufficient to disrupt G2 cell-cycle arrest consistent with activation of CDK1, implicating RGC-32 in the EBV transformation process. Surprisingly, RGC-32 mRNA is expressed at high levels in latency I Burkitt's lymphoma (BL) cells and in some EBV-negative BL cell-lines, although RGC-32 protein expression is not detectable. We show that RGC-32 mRNA expression is elevated in latency I cells due to transcriptional activation by high levels of the differentially expressed RUNX1c transcription factor. We found that proteosomal degradation or blocked cytoplasmic export of the RGC-32 message were not responsible for the lack of RGC-32 protein expression in latency I cells. Significantly, analysis of the ribosomal association of the RGC-32 mRNA in latency I and latency III cells revealed that RGC-32 transcripts were associated with multiple ribosomes in both cell-types implicating post-initiation translational repression mechanisms in the block to RGC-32 protein production in latency I cells. In summary, our results are the first to demonstrate RGC-32 protein upregulation in cells transformed by a human tumour virus and to identify post-initiation translational mechanisms as an expression control point for this key cell-cycle regulator.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Herpesvirus Humano 4/metabolismo , Proteínas Musculares/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Regulación hacia Arriba , Linfocitos B/metabolismo , Linfocitos B/virología , Proteína Quinasa CDC2/biosíntesis , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/biosíntesis , Citometría de Flujo/métodos , Fase G2 , Perfilación de la Expresión Génica , Humanos , Plásmidos/metabolismo , Polirribosomas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA