Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 169(7): 1214-1227.e18, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28622508

RESUMEN

Higher eukaryotic chromosomes are organized into topologically constrained functional domains; however, the molecular mechanisms required to sustain these complex interphase chromatin structures are unknown. A stable matrix underpinning nuclear organization was hypothesized, but the idea was abandoned as more dynamic models of chromatin behavior became prevalent. Here, we report that scaffold attachment factor A (SAF-A), originally identified as a structural nuclear protein, interacts with chromatin-associated RNAs (caRNAs) via its RGG domain to regulate human interphase chromatin structures in a transcription-dependent manner. Mechanistically, this is dependent on SAF-A's AAA+ ATPase domain, which mediates cycles of protein oligomerization with caRNAs, in response to ATP binding and hydrolysis. SAF-A oligomerization decompacts large-scale chromatin structure while SAF-A loss or monomerization promotes aberrant chromosome folding and accumulation of genome damage. Our results show that SAF-A and caRNAs form a dynamic, transcriptionally responsive chromatin mesh that organizes large-scale chromosome structures and protects the genome from instability.


Asunto(s)
Cromosomas/metabolismo , Inestabilidad Genómica , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , ARN Nuclear Pequeño/metabolismo , Secuencia de Aminoácidos , Cromatina , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo U/química , Humanos , Interfase , Modelos Moleculares , Alineación de Secuencia , Transcripción Genética
2.
EMBO J ; 40(22): e108234, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34586646

RESUMEN

DNA methylation is a fundamental epigenetic modification, important across biological processes. The maintenance methyltransferase DNMT1 is essential for lineage differentiation during development, but its functions in tissue homeostasis are incompletely understood. We show that epidermis-specific DNMT1 deletion severely disrupts epidermal structure and homeostasis, initiating a massive innate immune response and infiltration of immune cells. Mechanistically, DNA hypomethylation in keratinocytes triggered transposon derepression, mitotic defects, and formation of micronuclei. DNA release into the cytosol of DNMT1-deficient keratinocytes activated signaling through cGAS and STING, thus triggering inflammation. Our findings show that disruption of a key epigenetic mark directly impacts immune and tissue homeostasis, and potentially impacts our understanding of autoinflammatory diseases and cancer immunotherapy.


Asunto(s)
Metilación de ADN , Dermatitis/genética , Epidermis/fisiopatología , Nucleotidiltransferasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Aberraciones Cromosómicas , Citosol/fisiología , ADN (Citosina-5-)-Metiltransferasa 1/genética , Dermatitis/inmunología , Dermatitis/patología , Humanos , Inmunidad Innata/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Queratinocitos/inmunología , Queratinocitos/metabolismo , Queratinocitos/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Transgénicos , Nucleotidiltransferasas/genética
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.
Proc Natl Acad Sci U S A ; 116(30): 14995-15000, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31289233

RESUMEN

Patterns of gene expression are primarily determined by proteins that locally enhance or repress transcription. While many transcription factors target a restricted number of genes, others appear to modulate transcription levels globally. An example is MeCP2, an abundant methylated-DNA binding protein that is mutated in the neurological disorder Rett syndrome. Despite much research, the molecular mechanism by which MeCP2 regulates gene expression is not fully resolved. Here, we integrate quantitative, multidimensional experimental analysis and mathematical modeling to indicate that MeCP2 is a global transcriptional regulator whose binding to DNA creates "slow sites" in gene bodies. We hypothesize that waves of slowed-down RNA polymerase II formed behind these sites travel backward and indirectly affect initiation, reminiscent of defect-induced shockwaves in nonequilibrium physics transport models. This mechanism differs from conventional gene-regulation mechanisms, which often involve direct modulation of transcription initiation. Our findings point to a genome-wide function of DNA methylation that may account for the reversibility of Rett syndrome in mice. Moreover, our combined theoretical and experimental approach provides a general method for understanding how global gene-expression patterns are choreographed.


Asunto(s)
Metilación de ADN , Modelos Teóricos , ARN Polimerasa II/metabolismo , Animales , Línea Celular , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Unión Proteica , Elongación de la Transcripción Genética , Iniciación de la Transcripción Genética , Activación Transcripcional
5.
PLoS Genet ; 15(3): e1007605, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30856165

RESUMEN

Typical Martsolf syndrome is characterized by congenital cataracts, postnatal microcephaly, developmental delay, hypotonia, short stature and biallelic hypomorphic mutations in either RAB3GAP1 or RAB3GAP2. Genetic analysis of 85 unrelated "mutation negative" probands with Martsolf or Martsolf-like syndromes identified two individuals with different homozygous null mutations in ITPA, the gene encoding inosine triphosphate pyrophosphatase (ITPase). Both probands were from multiplex families with a consistent, lethal and highly distinctive disorder; a Martsolf-like syndrome with infantile-onset dilated cardiomyopathy. Severe ITPase-deficiency has been previously reported with infantile epileptic encephalopathy (MIM 616647). ITPase acts to prevent incorporation of inosine bases (rI/dI) into RNA and DNA. In Itpa-null cells dI was undetectable in genomic DNA. dI could be identified at a low level in mtDNA without detectable mitochondrial genome instability, mtDNA depletion or biochemical dysfunction of the mitochondria. rI accumulation was detectable in proband-derived lymphoblastoid RNA. In Itpa-null mouse embryos rI was detectable in the brain and kidney with the highest level seen in the embryonic heart (rI at 1 in 385 bases). Transcriptome and proteome analysis in mutant cells revealed no major differences with controls. The rate of transcription and the total amount of cellular RNA also appeared normal. rI accumulation in RNA-and by implication rI production-correlates with the severity of organ dysfunction in ITPase deficiency but the basis of the cellulopathy remains cryptic. While we cannot exclude cumulative minor effects, there are no major anomalies in the production, processing, stability and/or translation of mRNA.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/genética , Catarata/enzimología , Catarata/genética , Hipogonadismo/enzimología , Hipogonadismo/genética , Discapacidad Intelectual/enzimología , Discapacidad Intelectual/genética , Errores Innatos del Metabolismo/enzimología , Errores Innatos del Metabolismo/genética , Pirofosfatasas/deficiencia , Animales , Secuencia de Bases , Preescolar , Análisis Mutacional de ADN , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Femenino , Homocigoto , Humanos , Inosina/metabolismo , Masculino , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/enzimología , Mutación , Linaje , Pirofosfatasas/genética , ARN/genética , ARN/metabolismo , Secuenciación del Exoma
6.
PLoS Genet ; 13(5): e1006793, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28498846

RESUMEN

Mutations in the gene encoding the methyl-CG binding protein MeCP2 cause several neurological disorders including Rett syndrome. The di-nucleotide methyl-CG (mCG) is the classical MeCP2 DNA recognition sequence, but additional methylated sequence targets have been reported. Here we show by in vitro and in vivo analyses that MeCP2 binding to non-CG methylated sites in brain is largely confined to the tri-nucleotide sequence mCAC. MeCP2 binding to chromosomal DNA in mouse brain is proportional to mCAC + mCG density and unexpectedly defines large genomic domains within which transcription is sensitive to MeCP2 occupancy. Our results suggest that MeCP2 integrates patterns of mCAC and mCG in the brain to restrain transcription of genes critical for neuronal function.


Asunto(s)
Encéfalo/metabolismo , Metilación de ADN , Repeticiones de Dinucleótido , Proteína 2 de Unión a Metil-CpG/metabolismo , Repeticiones de Trinucleótidos , Animales , Islas de CpG , Citosina/metabolismo , Epigénesis Genética , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Síndrome de Rett/genética
7.
Nucleic Acids Res ; 44(16): 7592-604, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27179028

RESUMEN

Methylation of DNA at carbon 5 of cytosine is essential for mammalian development and implicated in transcriptional repression of genes and transposons. New patterns of DNA methylation characteristic of lineage-committed cells are established at the exit from pluripotency by de novo DNA methyltransferases enzymes, DNMT3A and DNMT3B, which are regulated by developmental signaling and require access to chromatin-organized DNA. Whether or not the capacity for de novo DNA methylation of developmentally regulated loci is preserved in differentiated somatic cells and can occur in the absence of exogenous signals is currently unknown. Here, we demonstrate that fibroblasts derived from chromatin remodeling ATPase LSH (HELLS)-null mouse embryos, which lack DNA methylation from centromeric repeats, transposons and a number of gene promoters, are capable of reestablishing DNA methylation and silencing of misregulated genes upon re-expression of LSH. We also show that the ability of LSH to bind ATP and the cellular concentration of DNMT3B are critical for cell-autonomous de novo DNA methylation in somatic cells. These data suggest the existence of cellular memory that persists in differentiated cells through many cell generations and changes in transcriptional state.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , ADN Helicasas/metabolismo , Metilación de ADN , Fibroblastos/metabolismo , 5-Metilcitosina/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Embrión de Mamíferos/citología , Fibroblastos/citología , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Ratones , Mutación/genética , Células 3T3 NIH , Regiones Promotoras Genéticas/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Retroelementos/genética , ADN Metiltransferasa 3B
8.
Nucleic Acids Res ; 40(11): 4794-803, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22323521

RESUMEN

Chemical inhibitors of histone deacetylase (HDAC) activity are used as experimental tools to induce histone hyperacetylation and deregulate gene transcription, but it is not known whether the inhibition of HDACs plays any part in the normal physiological regulation of transcription. Using both in vitro and in vivo assays, we show that lactate, which accumulates when glycolysis exceeds the cell's aerobic metabolic capacity, is an endogenous HDAC inhibitor, deregulating transcription in an HDAC-dependent manner. Lactate is a relatively weak inhibitor (IC(50) 40 mM) compared to the established inhibitors trichostatin A and butyrate, but the genes deregulated overlap significantly with those affected by low concentrations of the more potent inhibitors. HDAC inhibition causes significant up and downregulation of genes, but genes that are associated with HDAC proteins are more likely to be upregulated and less likely to be downregulated than would be expected. Our results suggest that the primary effect of HDAC inhibition by endogenous short-chain fatty acids like lactate is to promote gene expression at genes associated with HDAC proteins. Therefore, we propose that lactate may be an important transcriptional regulator, linking the metabolic state of the cell to gene transcription.


Asunto(s)
Regulación de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Ácido Láctico/farmacología , Acetilación , Aniones , Butiratos/farmacología , Línea Celular , Medios de Cultivo/química , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Humanos , Ácido Láctico/análisis
9.
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
10.
Nat Genet ; 37(11): 1274-9, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16244654

RESUMEN

Embryonic stem (ES) cells are important tools in the study of gene function and may also become important in cell therapy applications. Establishment of stable XX ES cell lines from mouse blastocysts is relatively problematic owing to frequent loss of one of the two X chromosomes. Here we show that DNA methylation is globally reduced in XX ES cell lines and that this is attributable to the presence of two active X chromosomes. Hypomethylation affects both repetitive and unique sequences, the latter including differentially methylated regions that regulate expression of parentally imprinted genes. Methylation of differentially methylated regions can be restored coincident with elimination of an X chromosome in early-passage parthenogenetic ES cells, suggesting that selection against loss of methylation may provide the basis for X-chromosome instability. Finally, we show that hypomethylation is associated with reduced levels of the de novo DNA methyltransferases Dnmt3a and Dnmt3b and that ectopic expression of these factors restores global methylation levels.


Asunto(s)
Metilación de ADN , Embrión de Mamíferos/citología , Genoma , Células Madre/fisiología , Cromosoma X/genética , Animales , Inestabilidad Cromosómica , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Impresión Genómica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , ADN Metiltransferasa 3B
11.
J Cell Biol ; 177(3): 401-11, 2007 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-17485486

RESUMEN

DNA methylation has been implicated in chromatin condensation and nuclear organization, especially at sites of constitutive heterochromatin. How this is mediated has not been clear. In this study, using mutant mouse embryonic stem cells completely lacking in DNA methylation, we show that DNA methylation affects nuclear organization and nucleosome structure but not chromatin compaction. In the absence of DNA methylation, there is increased nuclear clustering of pericentric heterochromatin and extensive changes in primary chromatin structure. Global levels of histone H3 methylation and acetylation are altered, and there is a decrease in the mobility of linker histones. However, the compaction of both bulk chromatin and heterochromatin, as assayed by nuclease digestion and sucrose gradient sedimentation, is unaltered by the loss of DNA methylation. This study shows how the complete loss of a major epigenetic mark can have an impact on unexpected levels of chromatin structure and nuclear organization and provides evidence for a novel link between DNA methylation and linker histones in the regulation of chromatin structure.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Metilación de ADN , Histonas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Acetilación , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Heterocromatina/metabolismo , Metilación , Ratones , Ratones Noqueados , Nucleosomas , Unión Proteica/fisiología
12.
Nucleic Acids Res ; 38(13): 4313-24, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20348135

RESUMEN

5-Aza-2'-deoxycytidine (5-aza-dC) is a nucleoside analogue with cytotoxic and DNA demethylating effects. Here we show that 5-aza-dC induces the proteasomal degradation of free (non-chromatin bound) DNMT1 through a mechanism which is dependent on DNA synthesis and the targeting of incorporated 5-aza-dC residues by DNMT1 itself. Thus, 5-aza-dC induces Dnmt1 degradation in wild-type mouse ES cells, but not in Dnmt [3a(-/-), 3b(-/-)] mouse ES cells which express Dnmt1 but lack DNA methylation (<0.7% of CpG methylated) and contain few hemi-methylated CpG sites, these being the preferred substrates for Dnmt1. We suggest that adducts formed between DNMT1 and 5-aza-dC molecules in DNA induce a ubiquitin-E3 ligase activity which preferentially targets free DNMT1 molecules for degradation by the proteasome. The proteasome inhibitor MG132 prevents DNMT1 degradation and reduces hypomethylation induced by 5-aza-dC.


Asunto(s)
Azacitidina/análogos & derivados , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Afidicolina/farmacología , Azacitidina/metabolismo , Azacitidina/farmacología , Línea Celular , Cromatina/enzimología , ADN/biosíntesis , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , Metilación de ADN , ADN Metiltransferasa 3A , Decitabina , Inhibidores Enzimáticos/farmacología , Ratones
13.
Life Sci Alliance ; 4(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33310759

RESUMEN

Malignant transformation depends on genetic and epigenetic events that result in a burst of deregulated gene expression and chromatin changes. To dissect the sequence of events in this process, we used a T-cell-specific lymphoma model based on the human oncogenic nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) translocation. We find that transformation of T cells shifts thymic cell populations to an undifferentiated immunophenotype, which occurs only after a period of latency, accompanied by induction of the MYC-NOTCH1 axis and deregulation of key epigenetic enzymes. We discover aberrant DNA methylation patterns, overlapping with regulatory regions, plus a high degree of epigenetic heterogeneity between individual tumors. In addition, ALK-positive tumors show a loss of associated methylation patterns of neighboring CpG sites. Notably, deletion of the maintenance DNA methyltransferase DNMT1 completely abrogates lymphomagenesis in this model, despite oncogenic signaling through NPM-ALK, suggesting that faithful maintenance of tumor-specific methylation through DNMT1 is essential for sustained proliferation and tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Epigénesis Genética , Linfoma/etiología , Linfoma/metabolismo , Proteínas Tirosina Quinasas/genética , Animales , Biomarcadores de Tumor , Biología Computacional/métodos , ADN (Citosina-5-)-Metiltransferasa 1/genética , Metilación de ADN , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Epigenómica , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Inmunohistoquímica , Inmunofenotipificación , Linfoma/tratamiento farmacológico , Linfoma/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Tirosina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Anal Chem ; 80(11): 4147-53, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18457408

RESUMEN

We describe a quantitative Fourier transform ion cyclotron resonance mass spectrometric (FTICR MS) analysis of the relative proportions of post-translational modification states (PTMs) of core histones in cultured cells and tissues. A novel preseparation process using a monolithic column interfaced to a 12 T FTICR MS equipped with electron capture dissociation (ECD) yields very high mass accuracy spectra, allowing direct assignment of the PTMs present in the dominant modification states of intact H4, resolving a well recognized ambiguity between trimethylation and acetylation states. By eliminating preseparation, we also obtain a highly quantitative analysis of the distribution of H4 PTMs. Rapid, extensive, and reversible effects on PTMs induced by a histone deacetylase inhibitor indicate that H4 and other core histones are accessible to modification throughout the chromatin, not just in regions of active transcription. These methods provide tools for analysis of the histone code and its role in chromatin function.


Asunto(s)
Ciclotrones , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Acetilación/efectos de los fármacos , Ácidos/química , Relación Dosis-Respuesta a Droga , Electrones , Células HCT116 , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/química , Histonas/aislamiento & purificación , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Isótopos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Factores de Tiempo
15.
Cell Tissue Res ; 331(1): 31-55, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18060563

RESUMEN

Mammalian development is associated with considerable changes in global DNA methylation levels at times of genomic reprogramming. Normal DNA methylation is essential for development but, despite considerable advances in our understanding of the DNA methyltransferases, the reason that development fails when DNA methylation is deficient remains unclear. Furthermore, although much is known about the enzymes that cause DNA methylation, comparatively little is known about the mechanisms or significance of active demethylation in early development. In this review, we discuss the roles of the various DNA methyltransferases and their likely functions in development.


Asunto(s)
Metilación de ADN , Desarrollo Embrionario , Células Madre Embrionarias/metabolismo , Animales , Diferenciación Celular , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Ratones , Transcripción Genética
16.
Mol Cell Biol ; 24(20): 8862-71, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15456861

RESUMEN

It has been reported that DNA methyltransferase 1-deficient (Dnmt1-/-) embryonic stem (ES) cells are hypomethylated (20% CpG methylation) and die through apoptosis when induced to differentiate. Here, we show that Dnmt[3a-/-,3b-/-] ES cells with just 0.6% of their CpG dinucleotides behave differently: the majority of cells within the culture are partially or completely blocked in their ability to initiate differentiation, remaining viable while retaining the stem cell characteristics of alkaline phosphatase and Oct4 expression. Restoration of DNA methylation levels rescues these defects. Severely hypomethylated Dnmt[3a-/-,3b-/-] ES cells have increased histone acetylation levels, and those cells that can differentiate aberrantly express extraembryonic markers of differentiation. Dnmt[3a-/-,3b-/-] ES cells with >10% CpG methylation are able to terminally differentiate, whereas Dnmt1-/- ES cells with 20% of the CpG methylated cannot differentiate. This demonstrates that successful terminal differentiation is not dependent simply on adequate methylation levels. There is an absolute requirement that the methylation be delivered by the maintenance enzyme Dnmt1.


Asunto(s)
Diferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Histonas/metabolismo , Células Madre/fisiología , Acetilación , Animales , Biomarcadores , Linaje de la Célula , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/anatomía & histología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Inhibidores de Histona Desacetilasas , Interleucina-6/metabolismo , Factor Inhibidor de Leucemia , Lisina/metabolismo , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Factor 3 de Transcripción de Unión a Octámeros , Factor de Transcripción STAT3 , Transducción de Señal/fisiología , Células Madre/citología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transgenes
17.
Mol Cell Biol ; 22(7): 2124-35, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11884600

RESUMEN

Biallelic expression of Igf2 is frequently seen in cancers because Igf2 functions as a survival factor. In many tumors the activation of Igf2 expression has been correlated with de novo methylation of the imprinted region. We have compared the intrinsic susceptibilities of the imprinted region of Igf2 and H19, other imprinted genes, bulk genomic DNA, and repetitive retroviral sequences to Dnmt1 overexpression. At low Dnmt1 methyltransferase levels repetitive retroviral elements were methylated and silenced. The nonmethylated imprinted region of Igf2 and H19 was resistant to methylation at low Dnmt1 levels but became fully methylated when Dnmt1 was overexpressed from a bacterial artificial chromosome transgene. Methylation caused the activation of the silent Igf2 allele in wild-type and Dnmt1 knockout cells, leading to biallelic Igf2 expression. In contrast, the imprinted genes Igf2r, Peg3, Snrpn, and Grf1 were completely resistant to de novo methylation, even when Dnmt1 was overexpressed. Therefore, the intrinsic difference between the imprinted region of Igf2 and H19 and of other imprinted genes to postzygotic de novo methylation may be the molecular basis for the frequently observed de novo methylation and upregulation of Igf2 in neoplastic cells and tumors. Injection of Dnmt1-overexpressing embryonic stem cells in diploid or tetraploid blastocysts resulted in lethality of the embryo, which resembled embryonic lethality caused by Dnmt1 deficiency.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Pérdida del Embrión/genética , Regulación del Desarrollo de la Expresión Génica , Genoma , Impresión Genómica/genética , Proteínas Quinasas , Factores de Transcripción , Alelos , Animales , Western Blotting , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Eliminación de Gen , Dosificación de Gen , Orden Génico/genética , Silenciador del Gen , Hibridación Fluorescente in Situ , Factores de Transcripción de Tipo Kruppel , Ratones , Poliploidía , Proteínas/genética , ARN Largo no Codificante , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética , Receptor IGF Tipo 2/genética , Células Madre/metabolismo
18.
Nucleic Acids Res ; 33(18): 5868-77, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16224102

RESUMEN

We describe a large-scale random approach termed reduced representation bisulfite sequencing (RRBS) for analyzing and comparing genomic methylation patterns. BglII restriction fragments were size-selected to 500-600 bp, equipped with adapters, treated with bisulfite, PCR amplified, cloned and sequenced. We constructed RRBS libraries from murine ES cells and from ES cells lacking DNA methyltransferases Dnmt3a and 3b and with knocked-down (kd) levels of Dnmt1 (Dnmt[1(kd),3a-/-,3b-/-]). Sequencing of 960 RRBS clones from Dnmt[1(kd),3a-/-,3b-/-] cells generated 343 kb of non-redundant bisulfite sequence covering 66212 cytosines in the genome. All but 38 cytosines had been converted to uracil indicating a conversion rate of >99.9%. Of the remaining cytosines 35 were found in CpG and 3 in CpT dinucleotides. Non-CpG methylation was >250-fold reduced compared with wild-type ES cells, consistent with a role for Dnmt3a and/or Dnmt3b in CpA and CpT methylation. Closer inspection revealed neither a consensus sequence around the methylated sites nor evidence for clustering of residual methylation in the genome. Our findings indicate random loss rather than specific maintenance of methylation in Dnmt[1(kd),3a-/-,3b-/-] cells. Near-complete bisulfite conversion and largely unbiased representation of RRBS libraries suggest that random shotgun bisulfite sequencing can be scaled to a genome-wide approach.


Asunto(s)
Metilación de ADN , Genómica/métodos , Sulfitos/química , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Embrión de Mamíferos/citología , Biblioteca Genómica , Ratones , Ratones Noqueados , Interferencia de ARN , Células Madre/metabolismo
19.
Cell Rep ; 15(1): 77-85, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-27052169

RESUMEN

DNA methylation at imprinting control regions (ICRs) is established in gametes in a sex-specific manner and has to be stably maintained during development and in somatic cells to ensure the correct monoallelic expression of imprinted genes. In addition to DNA methylation, the ICRs are marked by allele-specific histone modifications. Whether these marks are essential for maintenance of genomic imprinting is largely unclear. Here, we show that the histone H3 lysine 9 methylases G9a and GLP are required for stable maintenance of imprinted DNA methylation in embryonic stem cells; however, their catalytic activity and the G9a/GLP-dependent H3K9me2 mark are completely dispensable for imprinting maintenance despite the genome-wide loss of non-imprinted DNA methylation in H3K9me2-depleted cells. We provide additional evidence that the G9a/GLP complex protects imprinted DNA methylation by recruitment of de novo DNA methyltransferases, which antagonize TET dioxygenass-dependent erosion of DNA methylation at ICRs.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/metabolismo , Impresión Genómica , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Línea Celular , Antígenos de Histocompatibilidad/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos
20.
Stem Cells Dev ; 24(7): 879-91, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25393219

RESUMEN

Embryonic stem cells (ESCs) are maintained in an undifferentiated state through expression of the core transcriptional factors Nanog, Oct4, and Sox2. However, the epigenetic regulation of pluripotency is poorly understood. Differentiation of ESCs is accompanied by a global reduction of panacetylation of histones H3 and H4 suggesting that histone acetylation plays an important role in maintenance of ESC pluripotency. Acetylated lysine residues on histones are read by members of the bromodomain family that includes BET (bromodomain and extraterminal domain) proteins for which highly potent and selective inhibitors have been developed. In this study we demonstrate that the pan-BET bromodomain inhibitor JQ1 induces rapid spontaneous differentiation of murine ESCs by inducing marked transcriptional downregulation of Nanog as well as the stemness markers Lefty1 and Lefty2, but not Myc, often used as a marker of BET inhibitor activity in cancer. We show that the effects of JQ1 are recapitulated by knockdown of the BET family member BRD4 implicating this protein in Nanog regulation. These data are also supported by chromatin immunoprecipitation experiments which confirm BRD4 binding at the Nanog promoter that is known to require acetylation by the histone acetyltransferase MOF for transcriptional activity. In further support of our findings, we show that JQ1 antagonizes the stem cell-promoting effects of the histone deacetylase inhibitors sodium butyrate and valproic acid. Our data suggest that BRD4 is critical for the maintenance of ESC pluripotency and that this occurs primarily through the maintenance of Nanog expression.


Asunto(s)
Azepinas/farmacología , Células Madre Embrionarias/citología , Proteínas de Homeodominio/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Triazoles/farmacología , Animales , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/genética , Factores de Determinación Derecha-Izquierda/genética , Ratones , Proteína Homeótica Nanog , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA