Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros




Base de datos
Intervalo de año de publicación
1.
Oncogene ; 36(10): 1328-1338, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27593931

RESUMEN

Cancer is characterised by DNA hypermethylation and gene silencing of CpG island-associated promoters, including tumour-suppressor genes. The methyl-CpG-binding domain (MBD) family of proteins bind to methylated DNA and can aid in the mediation of gene silencing through interaction with histone deacetylases and histone methyltransferases. However, the mechanisms responsible for eliciting CpG island hypermethylation in cancer, and the potential role that MBD proteins play in modulation of the methylome remain unclear. Our previous work demonstrated that MBD2 preferentially binds to the hypermethylated GSTP1 promoter CpG island in prostate cancer cells. Here, we use functional genetic approaches to investigate if MBD2 plays an active role in reshaping the DNA methylation landscape at this locus and genome-wide. First, we show that loss of MBD2 results in inhibition of both maintenance and spread of de novo methylation of a transfected construct containing the GSTP1 promoter CpG island in prostate cancer cells and Mbd2-/- mouse fibroblasts. De novo methylation was rescued by transient expression of Mbd2 in Mbd2-/- cells. Second, we show that MBD2 depletion triggers significant hypomethylation genome-wide in prostate cancer cells with concomitant loss of MBD2 binding at promoter and enhancer regulatory regions. Finally, CpG islands and shores that become hypomethylated after MBD2 depletion in LNCaP cancer cells show significant hypermethylation in clinical prostate cancer samples, highlighting a potential active role of MBD2 in promoting cancer-specific hypermethylation. Importantly, co-immunoprecipiation of MBD2 shows that MBD2 associates with DNA methyltransferase enzymes 1 and 3A. Together our results demonstrate that MBD2 has a critical role in 'rewriting' the cancer methylome at specific regulatory regions.


Asunto(s)
Islas de CpG , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Análisis por Conglomerados , Proteínas de Unión al ADN/genética , ADN-Citosina Metilasas/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Gutatión-S-Transferasa pi/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Unión Proteica
2.
Oncogene ; 32(23): 2891-9, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22869146

RESUMEN

Deregulation of microRNA (miRNA) expression can have a critical role in carcinogenesis. Here we show in prostate cancer that miRNA-205 (miR-205) transcription is commonly repressed and the MIR-205 locus is hypermethylated. LOC642587, the MIR-205 host gene of unknown function, is also concordantly inactivated. We show that miR-205 targets mediator 1 (MED1, also called TRAP220 and PPARBP) for transcriptional silencing in normal prostate cells, leading to reduction in MED1 mRNA levels, and in total and active phospho-MED1 protein. Overexpression of miR-205 in prostate cancer cells negatively affects cell viability, consistent with a tumor suppressor function. We found that hypermethylation of the MIR-205 locus was strongly related with a decrease in miR-205 expression and an increase in MED1 expression in primary tumor samples (n=14), when compared with matched normal prostate (n=7). An expanded patient cohort (tumor n=149, matched normal n=30) also showed significant MIR-205 DNA methylation in tumors compared with normal, and MIR-205 hypermethylation is significantly associated with biochemical recurrence (hazard ratio=2.005, 95% confidence interval (1.109, 3.625), P=0.02), in patients with low preoperative prostate specific antigen. In summary, these results suggest that miR-205 is an epigenetically regulated tumor suppressor that targets MED1 and may provide a potential biomarker in prostate cancer management.


Asunto(s)
Adenocarcinoma/metabolismo , Silenciador del Gen , Subunidad 1 del Complejo Mediador/metabolismo , MicroARNs/genética , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Anciano , Secuencia de Bases , Línea Celular Tumoral , Supervivencia Celular , Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Datos de Secuencia Molecular , Fosforilación , Pronóstico , Regiones Promotoras Genéticas , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/mortalidad , Procesamiento Proteico-Postraduccional
4.
Nucleic Acids Res ; 25(21): 4422-6, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9336479

RESUMEN

Methylation analysis of individual cytosines in genomic DNA can be determined quantitatively by bisulphite treatment and PCR amplification of the target DNA sequence, followed by restriction enzyme digestion or sequencing. Methylated and unmethylated molecules, however, have different sequences after bisulphite conversion. For some sequences this can result in bias during the PCR amplification leading to an inaccurate estimate of methylation. PCR bias is sequence dependent and often strand-specific. This study presents a simple method for detection and measurement of PCR bias for any set of primers, and investigates parameters for overcoming PCR bias.


Asunto(s)
Citosina/análogos & derivados , Metilación de ADN , ADN/química , Reacción en Cadena de la Polimerasa/métodos , Sulfitos , 5-Metilcitosina , Animales , Composición de Base , Línea Celular , Citosina/análisis , Cartilla de ADN , Genes de Retinoblastoma/genética , Humanos , Ratones , Sensibilidad y Especificidad
5.
Cancer Res ; 57(11): 2229-37, 1997 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9187126

RESUMEN

The retinoblastoma gene (Rb) is one of the best characterized tumor suppressor genes, and its inactivation is associated with a number of cancers. Previous studies have shown, by restriction enzyme analysis, that the promoter region of the Rb gene is methylated in a significant proportion of primary retinoblastoma tumors. We now report the first detailed methylation sequence analysis of the CpG island spanning the retinoblastoma promoter from hypermethylated retinoblastoma tumors. Our results show methylation is not confined to a specific CpG site, as detected by restriction enzyme studies, but extends to essentially all 27 CpG dinucleotides spanning the retinoblastoma CpG island, including the core promoter. The methylation pattern from each tumor DNA sample is different, ranging from densely to sparsely methylated profiles. Single CpG sites, in particular the E2F transcription factor binding site, as well as blocks of CpGs, were undermethylated in some tumor samples. Possible interference of methylation could be due to the binding of sequence-specific protein factors at these sites in the tumor cells. This study highlights that the dynamics of DNA methylation in cancer cells are clearly different from normal cells and gives an insight into the mechanism of abnormal methylation of CpG islands in cancer cells.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Metilación de ADN , Proteínas de Unión al ADN , Neoplasias del Ojo/genética , Genes de Retinoblastoma , Regiones Promotoras Genéticas , Retinoblastoma/genética , Secuencia de Bases , Southern Blotting , Clonación Molecular , Islas de CpG , Factores de Transcripción E2F , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Proteína 1 de Unión a Retinoblastoma , Análisis de Secuencia de ADN , Factor de Transcripción DP1 , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA