Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
Nature ; 547(7663): 350-354, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28700576

RESUMEN

After liver injury, regeneration occurs through self-replication of hepatocytes. In severe liver injury, hepatocyte proliferation is impaired-a feature of human chronic liver disease. It is unclear whether other liver cell types can regenerate hepatocytes. Here we use two independent systems to impair hepatocyte proliferation during liver injury to evaluate the contribution of non-hepatocytes to parenchymal regeneration. First, loss of ß1-integrin in hepatocytes with liver injury triggered a ductular reaction of cholangiocyte origin, with approximately 25% of hepatocytes being derived from a non-hepatocyte origin. Second, cholangiocytes were lineage traced with concurrent inhibition of hepatocyte proliferation by ß1-integrin knockdown or p21 overexpression, resulting in the significant emergence of cholangiocyte-derived hepatocytes. We describe a model of combined liver injury and inhibition of hepatocyte proliferation that causes physiologically significant levels of regeneration of functional hepatocytes from biliary cells.


Asunto(s)
Conductos Biliares Intrahepáticos/citología , Hepatocitos/patología , Regeneración Hepática , Hígado/citología , Hígado/patología , Células Madre/citología , Animales , Linaje de la Célula , Proliferación Celular , Femenino , Integrina beta1/genética , Hígado/lesiones , Hepatopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Nucleic Acids Res ; 48(14): 7748-7766, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32585002

RESUMEN

Mouse embryonic stem cells (mESCs) cultured with MEK/ERK and GSK3ß (2i) inhibitors transition to ground state pluripotency. Gene expression changes, redistribution of histone H3K27me3 profiles and global DNA hypomethylation are hallmarks of 2i exposure, but it is unclear whether epigenetic alterations are required to achieve and maintain ground state or occur as an outcome of 2i signal induced changes. Here we show that ESCs with three epitypes, WT, constitutively methylated, or hypomethylated, all undergo comparable morphological, protein expression and transcriptome changes independently of global alterations of DNA methylation levels or changes in H3K27me3 profiles. Dazl and Fkbp6 expression are induced by 2i in all three epitypes, despite exhibiting hypermethylated promoters in constitutively methylated ESCs. We identify a number of activated gene promoters that undergo 2i dependent loss of H3K27me3 in all three epitypes, however genetic and pharmaceutical inhibition experiments show that H3K27me3 is not required for their silencing in non-2i conditions. By separating and defining their contributions, our data suggest that repressive epigenetic systems play minor roles in mESC self-renewal and naïve ground state establishment by core sets of dominant pluripotency associated transcription factor networks, which operate independently from these epigenetic processes.


Asunto(s)
Represión Epigenética , Redes Reguladoras de Genes , Células Madre Embrionarias de Ratones/metabolismo , Animales , Células Cultivadas , Metilación de ADN , Epigénesis Genética , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Histonas/metabolismo , Masculino , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/enzimología , Factores de Transcripción/metabolismo , Transcripción Genética
4.
Nat Methods ; 15(7): 499-504, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29941872

RESUMEN

DNA immunoprecipitation followed by sequencing (DIP-seq) is a common enrichment method for profiling DNA modifications in mammalian genomes. However, the results of independent DIP-seq studies often show considerable variation between profiles of the same genome and between profiles obtained by alternative methods. Here we show that these differences are primarily due to the intrinsic affinity of IgG for short unmodified DNA repeats. This pervasive experimental error accounts for 50-99% of regions identified as 'enriched' for DNA modifications in DIP-seq data. Correction of this error profoundly altered DNA-modification profiles for numerous cell types, including mouse embryonic stem cells, and subsequently revealed novel associations among DNA modifications, chromatin modifications and biological processes. We conclude that both matched input and IgG controls are essential in order for the results of DIP-based assays to be interpreted correctly, and that complementary, non-antibody-based techniques should be used to validate DIP-based findings to avoid further misinterpretation of genome-wide profiling data.


Asunto(s)
Dermatoglifia del ADN/métodos , ADN/genética , Genómica/métodos , Inmunoprecipitación/métodos , Animales , Islas de CpG , ADN/inmunología , Metilación de ADN , Células Madre Embrionarias , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Inmunoglobulina G , Masculino , Ratones , Análisis de Secuencia de ADN/métodos
5.
BMC Biol ; 18(1): 25, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32131813

RESUMEN

BACKGROUND: Thousands of mammalian promoters are defined by co-enrichment of the histone tail modifications H3K27me3 (repressive) and H3K4me3 (activating) and are thus termed bivalent. It was previously observed that bivalent genes in human ES cells (hESC) are frequent targets for hypermethylation in human cancers, and depletion of DNA methylation in mouse embryonic stem cells has a marked impact on H3K27me3 distribution at bivalent promoters. However, only a fraction of bivalent genes in stem cells are targets of hypermethylation in cancer, and it is currently unclear whether all bivalent promoters are equally sensitive to DNA hypomethylation and whether H3K4me3 levels play a role in the interplay between DNA methylation and H3K27me3. RESULTS: We report the sub-classification of bivalent promoters into two groups-promoters with a high H3K27me3:H3K4me3 (hiBiv) ratio or promoters with a low H3K27me3:H3K4me3 ratio (loBiv). HiBiv are enriched in canonical Polycomb components, show a higher degree of local intrachromosomal contacts and are highly sensitive to DNA hypomethylation in terms of H3K27me3 depletion from broad Polycomb domains. In contrast, loBiv promoters are enriched in non-canonical Polycomb components, show lower intrachromosomal contacts and are less sensitive to DNA hypomethylation at the same genomic resolution. Multiple systems reveal that hiBiv promoters are more depleted of Polycomb complexes than loBiv promoters following a reduction in DNA methylation, and we demonstrate that H3K27me3 re-accumulates at promoters when DNA methylation is restored. In human cancer, we show that hiBiv promoters lose H3K27me3 and are more susceptible to DNA hypermethylation than loBiv promoters. CONCLUSION: We conclude that bivalency as a general term to describe mammalian promoters is an over-simplification and our sub-classification has revealed novel insights into the interplay between the largely antagonistic presence of DNA methylation and Polycomb systems at bivalent promoters. This approach redefines molecular pathologies underlying disease in which global DNA methylation is aberrant or where Polycomb mutations are present.


Asunto(s)
Metilación de ADN , Neoplasias/genética , Regiones Promotoras Genéticas , Animales , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , Células Madre Embrionarias de Ratones/metabolismo
9.
Bioessays ; 36(2): 134-40, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24277643

RESUMEN

DNA methylation is a repressive epigenetic mark vital for normal development. Recent studies have uncovered an unexpected role for the DNA methylome in ensuring the correct targeting of the Polycomb repressive complexes throughout the genome. Here, we discuss the implications of these findings for cancer, where DNA methylation patterns are widely reprogrammed. We speculate that cancer-associated reprogramming of the DNA methylome leads to an altered Polycomb binding landscape, influencing gene expression by multiple modes. As the Polycomb system is responsible for the regulation of genes with key roles in cell fate decisions and cell cycle regulation, DNA methylation induced Polycomb mis-targeting could directly drive carcinogenesis and disease progression.


Asunto(s)
Metilación de ADN/fisiología , Neoplasias/genética , Proteínas del Grupo Polycomb/metabolismo , Animales , Metilación de ADN/genética , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Humanos , Proteínas del Grupo Polycomb/genética
10.
J Cell Biochem ; 116(3): 476-85, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25358258

RESUMEN

MBD4 is the only methyl-CpG binding protein that possesses a C-terminal glycosylase domain. It has been associated with a number of nuclear pathways including DNA repair, DNA damage response, the initiation of apoptosis, transcriptional repression, and DNA demethylation. However, the precise contribution of MBD4 to these processes in development and relevant diseases remains elusive. We identified UHRF1 and USP7 as two new interaction partners for MBD4. Both UHRF1, a E3 ubiquitin ligase, and USP7, a de-ubiquinating enzyme, regulate the stability of the DNA maintenance methyltransferase, Dnmt1. The ability of MBD4 to directly interact with and recruit USP7 to chromocenters implicates it as an additional factor that can potentially regulate Dnmt1 activity during cell proliferation.


Asunto(s)
Endodesoxirribonucleasas/metabolismo , Heterocromatina/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Replicación del ADN , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Ratones , Unión Proteica , Ubiquitina-Proteína Ligasas , Peptidasa Específica de Ubiquitina 7
11.
Hum Mol Genet ; 22(9): 1791-806, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23364048

RESUMEN

DNA methylation plays an important role in suppressing retrotransposon activity in mammalian genomes, yet there are stages of mammalian development where global hypomethylation puts the genome at risk of retrotransposition-mediated genetic instability. Hypomethylated primordial germ cells appear to limit this risk by expressing a cohort of retrotransposon-suppressing genome-defence genes whose silencing depends on promoter DNA methylation. Here, we investigate whether similar mechanisms operate in hypomethylated trophectoderm-derived components of the mammalian placenta to couple expression of genome-defence genes to the potential for retrotransposon activity. We show that the hypomethylated state of the mouse placenta results in activation of only one of the hypomethylation-sensitive germline genome-defence genes: Tex19.1. Tex19.1 appears to play an important role in placenta function as Tex19.1(-/-) mouse embryos exhibit intra-uterine growth retardation and have small placentas due to a reduction in the number of spongiotrophoblast, glycogen trophoblast and sinusoidal trophoblast giant cells. Furthermore, we show that retrotransposon mRNAs are derepressed in Tex19.1(-/-) placentas and that protein encoded by the LINE-1 retrotransposon is upregulated in hypomethylated trophectoderm-derived cells that normally express Tex19.1. This study suggests that post-transcriptional genome-defence mechanisms are operating in the placenta to protect the hypomethylated cells in this tissue from retrotransposons and suggests that imbalances between retrotransposon activity and genome-defence mechanisms could contribute to placenta dysfunction and disease.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Elementos de Nucleótido Esparcido Largo , Proteínas Nucleares/genética , Placenta/metabolismo , Animales , Metilación de ADN , Femenino , Retardo del Crecimiento Fetal/patología , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , Placenta/citología , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN , Análisis de Secuencia de ADN , Trofoblastos/metabolismo , Regulación hacia Arriba
12.
Genome Res ; 22(3): 467-77, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22106369

RESUMEN

The discovery of substantial amounts of 5-hydroxymethylcytosine (5hmC), formed by the oxidation of 5-methylcytosine (5mC), in various mouse tissues and human embryonic stem (ES) cells has necessitated a reevaluation of our knowledge of 5mC/5hmC patterns and functions in mammalian cells. Here, we investigate the tissue specificity of both the global levels and locus-specific distribution of 5hmC in several human tissues and cell lines. We find that global 5hmC content of normal human tissues is highly variable, does not correlate with global 5mC content, and decreases rapidly as cells from normal tissue adapt to cell culture. Using tiling microarrays to map 5hmC levels in DNA from normal human tissues, we find that 5hmC patterns are tissue specific; unsupervised hierarchical clustering based solely on 5hmC patterns groups independent biological samples by tissue type. Moreover, in agreement with previous studies, we find 5hmC associated primarily, but not exclusively, with the body of transcribed genes, and that within these genes 5hmC levels are positively correlated with transcription levels. However, using quantitative 5hmC-qPCR, we find that the absolute levels of 5hmC for any given gene are primarily determined by tissue type, gene expression having a secondary influence on 5hmC levels. That is, a gene transcribed at a similar level in several different tissues may have vastly different levels of 5hmC (>20-fold) dependent on tissue type. Our findings highlight tissue type as a major modifier of 5hmC levels in expressed genes and emphasize the importance of using quantitative analyses in the study of 5hmC levels.


Asunto(s)
Citosina/análogos & derivados , ADN/química , Regulación de la Expresión Génica , Transcripción Genética , 5-Metilcitosina/análogos & derivados , Animales , Línea Celular , Células Cultivadas , Mapeo Cromosómico , Análisis por Conglomerados , Citosina/análisis , Metilación de ADN , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Perfilación de la Expresión Génica , Sitios Genéticos , Humanos , Ratones , Proteínas Nucleares/genética , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Proteínas de Unión al ARN
13.
Development ; 139(19): 3623-32, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22949617

RESUMEN

Mouse primordial germ cells (PGCs) erase global DNA methylation (5mC) as part of the comprehensive epigenetic reprogramming that occurs during PGC development. 5mC plays an important role in maintaining stable gene silencing and repression of transposable elements (TE) but it is not clear how the extensive loss of DNA methylation impacts on gene expression and TE repression in developing PGCs. Using a novel epigenetic disruption and recovery screen and genetic analyses, we identified a core set of germline-specific genes that are dependent exclusively on promoter DNA methylation for initiation and maintenance of developmental silencing. These gene promoters appear to possess a specialised chromatin environment that does not acquire any of the repressive H3K27me3, H3K9me2, H3K9me3 or H4K20me3 histone modifications when silenced by DNA methylation. Intriguingly, this methylation-dependent subset is highly enriched in genes with roles in suppressing TE activity in germ cells. We show that the mechanism for developmental regulation of the germline genome-defence genes involves DNMT3B-dependent de novo DNA methylation. These genes are then activated by lineage-specific promoter demethylation during distinct global epigenetic reprogramming events in migratory (~E8.5) and post-migratory (E10.5-11.5) PGCs. We propose that genes involved in genome defence are developmentally regulated primarily by promoter DNA methylation as a sensory mechanism that is coupled to the potential for TE activation during global 5mC erasure, thereby acting as a failsafe to ensure TE suppression and maintain genomic integrity in the germline.


Asunto(s)
Metilación de ADN/fisiología , Desarrollo Embrionario/genética , Epigénesis Genética , Genoma , Células Germinativas/metabolismo , Regiones Promotoras Genéticas , Animales , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , Citoprotección/genética , Daño del ADN/genética , Embrión de Mamíferos , Epigénesis Genética/fisiología , Genoma/genética , Células Germinativas/fisiología , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Regiones Promotoras Genéticas/fisiología
14.
Cell Mol Life Sci ; 71(9): 1581-605, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24045705

RESUMEN

The viability of any species requires that the genome is kept stable as it is transmitted from generation to generation by the germ cells. One of the challenges to transgenerational genome stability is the potential mutagenic activity of transposable genetic elements, particularly retrotransposons. There are many different types of retrotransposon in mammalian genomes, and these target different points in germline development to amplify and integrate into new genomic locations. Germ cells, and their pluripotent developmental precursors, have evolved a variety of genome defence mechanisms that suppress retrotransposon activity and maintain genome stability across the generations. Here, we review recent advances in understanding how retrotransposon activity is suppressed in the mammalian germline, how genes involved in germline genome defence mechanisms are regulated, and the consequences of mutating these genome defence genes for the developing germline.


Asunto(s)
Genoma , Células Germinativas/metabolismo , Retroelementos/genética , Animales , Metilación de ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Germinativas/citología , Humanos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ARN Interferente Pequeño/metabolismo
15.
Nucleic Acids Res ; 41(11): 5639-54, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23598998

RESUMEN

Aberrant DNA methylation is a common feature of neoplastic lesions, and early detection of such changes may provide powerful mechanistic insights and biomarkers for carcinogenesis. Here, we investigate dynamic changes in the mouse liver DNA methylome associated with short (1 day) and prolonged (7, 28 and 91 days) exposure to the rodent liver non-genotoxic carcinogen, phenobarbital (PB). We find that the distribution of 5mC/5hmC is highly consistent between untreated individuals of a similar age; yet, changes during liver maturation in a transcriptionally dependent manner. Following drug treatment, we identify and validate a series of differentially methylated or hydroxymethylated regions: exposure results in staged transcriptional responses with distinct kinetic profiles that strongly correlate with promoter proximal region 5hmC levels. Furthermore, reciprocal changes for both 5mC and 5hmC in response to PB suggest that active demethylation may be taking place at each set of these loci via a 5hmC intermediate. Finally, we identify potential early biomarkers for non-genotoxic carcinogenesis, including several genes aberrantly expressed in liver cancer. Our work suggests that 5hmC profiling can be used as an indicator of cell states during organ maturation and drug-induced responses and provides novel epigenetic signatures for non-genotoxic carcinogen exposure.


Asunto(s)
Citosina/análogos & derivados , Metilación de ADN , Epigénesis Genética , Hígado/metabolismo , 5-Metilcitosina/metabolismo , Animales , Carcinógenos/toxicidad , Transformación Celular Neoplásica , Sistema Enzimático del Citocromo P-450/genética , Citosina/metabolismo , Epigénesis Genética/efectos de los fármacos , Marcadores Genéticos , Hígado/efectos de los fármacos , Hígado/crecimiento & desarrollo , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenobarbital/toxicidad , Regiones Promotoras Genéticas , Transcriptoma/efectos de los fármacos
16.
Nucleic Acids Res ; 41(14): 6857-69, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23703216

RESUMEN

LINE-1 retrotransposons are abundant repetitive elements of viral origin, which in normal cells are kept quiescent through epigenetic mechanisms. Activation of LINE-1 occurs frequently in cancer and can enable LINE-1 mobilization but also has retrotransposition-independent consequences. We previously reported that in cancer, aberrantly active LINE-1 promoters can drive transcription of flanking unique sequences giving rise to LINE-1 chimeric transcripts (LCTs). Here, we show that one such LCT, LCT13, is a large transcript (>300 kb) running antisense to the metastasis-suppressor gene TFPI-2. We have modelled antisense RNA expression at TFPI-2 in transgenic mouse embryonic stem (ES) cells and demonstrate that antisense RNA induces silencing and deposition of repressive histone modifications implying a causal link. Consistent with this, LCT13 expression in breast and colon cancer cell lines is associated with silencing and repressive chromatin at TFPI-2. Furthermore, we detected LCT13 transcripts in 56% of colorectal tumours exhibiting reduced TFPI-2 expression. Our findings implicate activation of LINE-1 elements in subsequent epigenetic remodelling of surrounding genes, thus hinting a novel retrotransposition-independent role for LINE-1 elements in malignancy.


Asunto(s)
Silenciador del Gen , Genes Supresores de Tumor , Glicoproteínas/genética , Elementos de Nucleótido Esparcido Largo , ARN sin Sentido/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación hacia Abajo , Células Madre Embrionarias/metabolismo , Femenino , Glicoproteínas/metabolismo , Humanos , Células MCF-7 , Ratones , ARN sin Sentido/química
17.
Nucleic Acids Res ; 41(22): e206, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24214958

RESUMEN

The epigenetic modification of 5-hydroxymethylcytosine (5hmC) is receiving great attention due to its potential role in DNA methylation reprogramming and as a cell state identifier. Given this interest, it is important to identify reliable and cost-effective methods for the enrichment of 5hmC marked DNA for downstream analysis. We tested three commonly used affinity-based enrichment techniques; (i) antibody, (ii) chemical capture and (iii) protein affinity enrichment and assessed their ability to accurately and reproducibly report 5hmC profiles in mouse tissues containing high (brain) and lower (liver) levels of 5hmC. The protein-affinity technique is a poor reporter of 5hmC profiles, delivering 5hmC patterns that are incompatible with other methods. Both antibody and chemical capture-based techniques generate highly similar genome-wide patterns for 5hmC, which are independently validated by standard quantitative PCR (qPCR) and glucosyl-sensitive restriction enzyme digestion (gRES-qPCR). Both antibody and chemical capture generated profiles reproducibly link to unique chromatin modification profiles associated with 5hmC. However, there appears to be a slight bias of the antibody to bind to regions of DNA rich in simple repeats. Ultimately, the increased specificity observed with chemical capture-based approaches makes this an attractive method for the analysis of locus-specific or genome-wide patterns of 5hmC.


Asunto(s)
Citosina/análogos & derivados , ADN/química , 5-Metilcitosina/análogos & derivados , Animales , Anticuerpos , Cromatina/metabolismo , Islas de CpG , Citosina/análisis , Citosina/inmunología , Proteínas de Unión al ADN/análisis , Sitios Genéticos , Impresión Genómica , Inmunoensayo/métodos , Hígado/química , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Secuencias Repetidas en Tándem
18.
Biochem J ; 451(1): 13-23, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23489368

RESUMEN

Methylation of the cytosine base in DNA, DNA methylation, is an essential epigenetic mark in mammals that contributes to the regulation of transcription. Several advances have been made in this area in recent years, leading to a leap forward in our understanding of how this pathway contributes to gene regulation during embryonic development, and the functional consequences of its perturbation in human disease. Critical to these advances is a comprehension of the genomic distribution of modified cytosine bases in unprecedented detail, drawing attention to genomic regions beyond gene promoters. In addition, we have a more complete understanding of the multifactorial manner by which DNA methylation influences gene regulation at the molecular level, and which genes rely directly on the DNA methylome for their normal transcriptional regulation. It is becoming apparent that a major role of DNA modification is to act as a relatively stable, and mitotically heritable, template that contributes to the establishment and maintenance of chromatin states. In this regard, interplay is emerging between DNA methylation and the PcG (Polycomb group) proteins, which act as evolutionarily conserved mediators of cell identity. In the present paper we review these aspects of DNA methylation, and discuss how a multifunctional view of DNA modification as an integral part of chromatin organization is influencing our understanding of this epigenetic mark's contribution to transcriptional regulation.


Asunto(s)
Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Genoma Humano/fisiología , Animales , Cromatina/genética , Cromatina/metabolismo , Humanos , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(11): 4364-9, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21368160

RESUMEN

Aberrant promoter hypermethylation is frequently observed in cancer. The potential for this mechanism to contribute to tumor development depends on whether the genes affected are repressed because of their methylation. Many aberrantly methylated genes play important roles in development and are bivalently marked in ES cells, suggesting that their aberrant methylation may reflect developmental processes. We investigated this possibility by analyzing promoter methylation in 19 breast cancer cell lines and 47 primary breast tumors. In cell lines, we defined 120 genes that were significantly repressed in association with methylation (SRAM). These genes allowed the unsupervised segregation of cell lines into epithelial (EPCAM+ve) and mesenchymal (EPCAM-ve) lineages. However, the methylated genes were already repressed in normal cells of the same lineage, and >90% could not be derepressed by treatment with 5-aza-2'-deoxycytidine. The tumor suppressor genes APC and CDH1 were among those methylated in a lineage-specific fashion. As predicted by the epithelial nature of most breast tumors, SRAM genes that were methylated in epithelial cell lines were frequently aberrantly methylated in primary tumors, as were genes specifically repressed in normal epithelial cells. An SRAM gene expression signature also correctly identified the rare claudin-low and metaplastic tumors as having mesenchymal characteristics. Our findings implicate aberrant DNA methylation as a marker of cell lineage rather than tumor progression and suggest that, in most cases, it does not cause the repression with which it is associated.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Linaje de la Célula/genética , Metilación de ADN/genética , Proteínas Represoras/genética , Transcripción Genética , Mama/metabolismo , Mama/patología , Línea Celular Tumoral , Islas de CpG/genética , Células Madre Embrionarias/metabolismo , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Histonas/metabolismo , Humanos , Mesodermo/metabolismo , Mesodermo/patología , Especificidad de Órganos/genética , Proteínas Represoras/metabolismo
20.
Hum Mol Genet ; 20(16): 3241-55, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21636528

RESUMEN

DNA methyltransferase 1 (DNMT1) maintains methylation at CpG dinucleotides, important for transcriptional silencing at many loci. It is also implicated in stabilizing repeat sequences: DNMT1 deficiency causes microsatellite instability in mouse embryonic stem cells, but it is unclear how this occurs, how repeats lacking CpG become unstable and whether the effect is confined to stem cells. To address these questions, we transfected hTERT-immortalized normal human fibroblasts (hTERT-1604) with a short hairpin RNA construct targeting DNMT1 and isolated stable integrants with different levels of protein. DNMT1 expression levels agreed well with methylation levels at imprinted genes. Knockdown cells showed two key characteristics of mismatch repair (MMR) deficiency, namely resistance to the drug 6-thioguanine and up to 10-fold elevated mutation rates at a CA(17) microsatellite reporter, but had limited viability. The likely cause of MMR defects is a matching drop in steady-state protein levels for key repair components in DNMT1 knockdown cells, affecting both the MutLα and MutSα complexes. This indirect effect on MMR proteins was also seen using a different targeting method in HT29 colon cancer cells and did not involve transcriptional silencing of the respective genes. Decreased levels of MMR components follow activation of the DNA damage response and blocking this response, and in particular poly(ADP-ribose) polymerase (PARP) overactivation, rescues cell viability in DNMT1-depleted cells. These results offer an explanation for how and why unmethylated microsatellite repeats can be destabilized in cells with decreased DNMT1 levels and uncover a novel and important role for PARP in this process.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/deficiencia , Daño del ADN , Reparación de la Incompatibilidad de ADN , Enzimas Reparadoras del ADN/metabolismo , Fibroblastos/enzimología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Endodesoxirribonucleasas/metabolismo , Humanos , Ratones , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA