Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Genome Res ; 24(10): 1613-23, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25170028

RESUMO

Cytosine methylation is critical in mammalian development and plays a role in diverse biologic processes such as genomic imprinting, X chromosome inactivation, and silencing of repeat elements. Several factors regulate DNA methylation in early embryogenesis, but their precise role in the establishment of DNA methylation at a given site remains unclear. We have generated a comprehensive methylation map in fibroblasts derived from the murine DNA methylation mutant Hells(-/-) (helicase, lymphoid specific, also known as LSH). It has been previously shown that HELLS can influence de novo methylation of retroviral sequences and endogenous genes. Here, we describe that HELLS controls cytosine methylation in a nuclear compartment that is in part defined by lamin B1 attachment regions. Despite widespread loss of cytosine methylation at regulatory sequences, including promoter regions of protein-coding genes and noncoding RNA genes, overall relative transcript abundance levels in the absence of HELLS are similar to those in wild-type cells. A subset of promoter regions shows increases of the histone modification H3K27me3, suggesting redundancy of epigenetic silencing mechanisms. Furthermore, HELLS modulates CG methylation at all classes of repeat elements and is critical for repression of a subset of repeat elements. Overall, we provide a detailed analysis of gene expression changes in relation to DNA methylation alterations, which contributes to our understanding of the biological role of cytosine methylation.


Assuntos
Citosina/metabolismo , DNA Helicases/genética , Metilação de DNA , DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Animais , Núcleo Celular/genética , Células-Tronco Embrionárias , Epigênese Genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Histonas/metabolismo , Lamina Tipo B/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação , Sequências Reguladoras de Ácido Nucleico , Sequências Repetitivas de Ácido Nucleico
2.
Nucleic Acids Res ; 43(3): 1444-55, 2015 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25578963

RESUMO

Lsh, a chromatin remodeling protein of the SNF2 family, is critical for normal heterochromatin structure. In particular, DNA methylation at repeat elements, a hallmark of heterochromatin, is greatly reduced in Lsh(-/-) (KO) cells. Here, we examined the presumed nucleosome remodeling activity of Lsh on chromatin in the context of DNA methylation. We found that dynamic CG methylation was dependent on Lsh in embryonic stem cells. Moreover, we demonstrate that ATP function is critical for de novo methylation at repeat sequences. The ATP binding site of Lsh is in part required to promote stable association of the DNA methyltransferase 3b with the repeat locus. By performing nucleosome occupancy assays, we found distinct nucleosome occupancy in KO ES cells compared to WT ES cells after differentiation. Nucleosome density was restored to wild-type level by re-expressing wild-type Lsh but not the ATP mutant in KO ES cells. Our results suggest that ATP-dependent nucleosome remodeling is the primary molecular function of Lsh, which may promote de novo methylation in differentiating ES cells.


Assuntos
Trifosfato de Adenosina/metabolismo , Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Metilação de DNA , Nucleossomos/metabolismo , Sequências Repetitivas de Ácido Nucleico , Sítios de Ligação , Células Cultivadas , Humanos
3.
Proc Natl Acad Sci U S A ; 111(16): 5890-5, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24711395

RESUMO

DNA methylation patterns are established in early embryogenesis and are critical for cellular differentiation. To investigate the role of CG methylation in potential enhancer formation, we assessed H3K4me1 modification in murine embryonic fibroblasts (MEFs) derived from the DNA methylation mutant Lsh(-/-) mice. We report here de novo formation of putative enhancer elements at CG hypomethylated sites that can be dynamically altered. We found a subset of differentially enriched H3K4me1 regions clustered at neuronal lineage genes and overlapping with known cis-regulatory elements present in brain tissue. Reprogramming of Lsh(-/-) MEFs into induced pluripotent stem (iPS) cells leads to increased neuronal lineage gene expression of premarked genes and enhanced differentiation potential of Lsh(-/-) iPS cells toward the neuronal lineage pathway compared with WT iPS cells in vitro and in vivo. The state of CG hypomethylation and H3K4me1 enrichment is partially maintained in Lsh(-/-) iPS cells. The acquisition of H3K27ac and activity of subcloned fragments in an enhancer reporter assay indicate functional activity of several of de novo H3K4me1-marked sequences. Our results suggest a functional link of H3K4me1 enrichment at CG hypomethylated sites, enhancer formation, and cellular plasticity.


Assuntos
Ilhas de CpG/genética , DNA Helicases/deficiência , Metilação de DNA/genética , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem da Célula , DNA Helicases/metabolismo , Elementos Facilitadores Genéticos/genética , Epigênese Genética , Fibroblastos/citologia , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Knockout , Neurônios/citologia , Ligação Proteica , Transdução de Sinais , Fatores de Transcrição/metabolismo
4.
Proc Natl Acad Sci U S A ; 108(14): 5626-31, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21427231

RESUMO

DNA methylation is critical for normal development and plays important roles in genome organization and transcriptional regulation. Although DNA methyltransferases have been identified, the factors that establish and contribute to genome-wide methylation patterns remain elusive. Here, we report a high-resolution cytosine methylation map of the murine genome modulated by Lsh, a chromatin remodeling family member that has previously been shown to regulate CpG methylation at repetitive sequences. We provide evidence that Lsh also controls genome-wide cytosine methylation at nonrepeat sequences and relate those changes to alterations in H4K4me3 modification and gene expression. Deletion of Lsh alters the allocation of cytosine methylation in chromosomal regions of 50 kb to 2 Mb and, in addition, leads to changes in the methylation profile at the 5' end of genes. Furthermore, we demonstrate that loss of Lsh promotes--as well as prevents--cytosine methylation. Our data indicate that Lsh is an epigenetic modulator that is critical for normal distribution of cytosine methylation throughout the murine genome.


Assuntos
Citosina/metabolismo , DNA Helicases/metabolismo , Metilação de DNA , Epigenômica , Animais , Southern Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Cromatografia Líquida de Alta Pressão , Perfilação da Expressão Gênica , Genômica , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Estatísticas não Paramétricas
5.
Biochim Biophys Acta ; 1819(7): 757-62, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22381140

RESUMO

Methylation regulates DNA by altering chromatin and limiting accessibility of transcription factors and RNA polymerase. In this way, DNA methylation controls gene expression and plays a role in ES cell regulation, tissue differentiation and the development of the organism. In abnormal circumstances methylation can also induce diseases and promote cancer progression. Chromatin remodeling proteins such as the SNF2 family member Lsh regulates genome-wide cytosine methylation patterns during mammalian development. Lsh promotes methylation by targeting and repressing repeat sequences that are imbedded in heterochromatin. Lsh also regulates cytosine methylation at unique loci. Alterations in histone modifications (such as H3K4me3, histone acetylation, H3K27me3 and H2Aub) can be associated with DNA methylation changes making Lsh-mediated cytosine methylation part of a larger epigenetic network defining gene expression and cellular differentiation during development. This article is part of a Special Issue entitled: Chromatin in time and space.


Assuntos
Diferenciação Celular/genética , Metilação de DNA , Animais , Citosina/metabolismo , DNA Helicases/fisiologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Genoma Humano , Humanos , Sequências Repetitivas de Ácido Nucleico , Transcrição Gênica
6.
Nucleic Acids Res ; 39(22): 9508-20, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21880597

RESUMO

Inactivation of tumor suppressor genes plays an important role in tumorigenesis, and epigenetic modifications such as DNA methylation are frequently associated with transcriptional repression. Here, we show that gene silencing at selected genes with signs of DNA hypermethylation in breast cancer cells involves Pol II stalling. We studied several repressed genes with DNA hypermethylation within a region 1-kb upstream of the transcriptional start site that were upregulated after treatment with DNA demethylating agents, such as Azacytidine and several natural products. All those selected genes had stalled Pol II at their transcriptional start site and showed enhanced ser2 phosphorylated Pol II and elevated transcripts after drug treatment indicating successful elongation. In addition, a decrease of the epigenetic regulator LSH in a breast cancer cell line by siRNA treatment reduced DNA methylation and overcame Pol II stalling, whereas overexpression of LSH in a normal breast epithelial cell line increased DNA methylation and resulted in repression. Decrease of LSH was associated with reduced DNMT3b binding to promoter sequences, and depletion of DNMT3b by siRNA could release Pol II suggesting that DNMT3b is functionally involved. The release of paused Pol II was accompanied by a dynamic switch from repressive to active chromatin marks. Thus release of Pol II stalling can act as a mechanism for gene reactivation at specific target genes after DNA demethylating treatment in cancer cells.


Assuntos
Azacitidina/farmacologia , Neoplasias da Mama/genética , Metilação de DNA/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , RNA Polimerase II/metabolismo , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Feminino , Histonas/metabolismo , Humanos , Proteínas do Grupo Polycomb , Proteínas Repressoras/metabolismo , Sítio de Iniciação de Transcrição , Transcrição Gênica/efeitos dos fármacos
7.
Stem Cells ; 27(11): 2691-702, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19650037

RESUMO

Transcriptional control of stem cell genes is a critical step in differentiation of embryonic stem cells and in reprogramming of somatic cells into stem cells. Here we report that Lsh, a regulator of repressive chromatin at retrotransposons, also plays an important role in silencing of stem cell-specific genes such as Oct4. We found that CpG methylation is gained during in vitro differentiation of several stem cell-specific genes (in 11 of 12 promoter regions) and thus appears to be a common epigenetic mark. Lsh depletion prevents complete silencing of stem cell gene expression and moreover promotes the maintenance of stem cell characteristics in culture. Lsh is required for establishment of DNA methylation patterns at stem cell genes during differentiation, in part by regulating access of Dnmt3b to its genomic targets. Our results indicate that Lsh is involved in the control of stem cell genes and suggest that Lsh is an important epigenetic modulator during early stem cell differentiation.


Assuntos
DNA Helicases/metabolismo , Metilação de DNA , Inativação Gênica , Animais , Southern Blotting , Western Blotting , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Helicases/genética , Fator de Crescimento Epidérmico/genética , Fator 3 de Diferenciação de Crescimento/genética , Proteínas de Homeodomínio/genética , Glicoproteínas de Membrana/genética , Camundongos , Proteína Homeobox Nanog , Proteínas de Neoplasias/genética , Fator 3 de Transcrição de Octâmero/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Repressoras/genética , DNA Metiltransferase 3B
8.
PLoS One ; 5(2): e9163, 2010 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-20161795

RESUMO

DNA cytosine methylation is an important epigenetic mechanism that is involved in transcriptional silencing of developmental genes. Several molecular pathways have been described that interfere with Pol II initiation, but at individual genes the molecular mechanism of repression remains uncertain. Here, we study the molecular mechanism of transcriptional regulation at Hox genes in dependence of the epigenetic regulator Lsh that controls CpG methylation at selected Hox genes. Wild type cells show a nucleosomal deprived region around the transcriptional start site at methylated Hox genes and mediate gene silencing via Pol II stalling. Hypomethylation in Lsh-/- cells is associated with efficient transcriptional elongation and splicing, in part mediated by the chromodomain protein Chd1. Dynamic modulation of DNA methylation in Lsh-/- and wild type cells demonstrates that catalytically active DNA methyltransferase activity is required for Pol II stalling. Taken together, the data suggests that DNA methylation can be compatible with Pol II binding at selected genes and Pol II stalling can act as alternate mechanism to explain transcriptional silencing associated with DNA methylation.


Assuntos
DNA Helicases/metabolismo , Metilação de DNA , Proteínas de Homeodomínio/genética , RNA Polimerase II/metabolismo , Animais , Azacitidina/farmacologia , Western Blotting , Domínio Catalítico/genética , Células Cultivadas , Ilhas de CpG/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Helicases/deficiência , DNA Helicases/genética , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Mutação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , DNA Metiltransferase 3B
9.
Epigenetics ; 3(3): 134-42, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18487951

RESUMO

Hematopoietic malignancies are frequently associated with DNA hypomethylation but the molecular mechanisms involved in tumor formation remain poorly understood. Here we report that mice lacking Lsh develop leukemia associated with DNA hypomethylation and oncogene activation. Lsh is a member of the SNF2 chromatin remodeling family and is required for de novo methylation of genomic DNA. Mice that received Lsh deficient hematopoietic progenitors showed severe impairment of hematopoiesis, suggesting that Lsh is necessary for normal hematopoiesis. A subset of mice developed erythroleukemia, a tumor that does not spontaneously occur in mice. Tumor tissues were CpG hypomethylated and showed a modest elevation of the transcription factor PU.1, an oncogene that is crucial for Friend virus induced erythroleukemia. Analysis of Lsh(-/-) hematopoietic progenitors revealed widespread DNA hypomethylation at repetitive sequences and hypomethylation at specific retroviral elements within the PU.1 gene. Wild type cells showed Lsh and Dnmt3b binding at the retroviral elements located within the PU.1 gene. On the other hand, Lsh deficient cells had no detectable Dnmt3b association suggesting that Lsh is necessary for recruitment of Dnmt3b to its target. Furthermore, Lsh(-/-) hematopoietic precursors showed impaired suppression of retroviral elements in the PU.1 gene, an increase of PU.1 transcripts and protein levels. Thus DNA hypomethylation caused by Lsh depletion is linked to transcriptional upregulation of retroviral elements and oncogenes such as PU.1 which in turn may promote the development of erythroleukemia in mice.


Assuntos
DNA Helicases/deficiência , Metilação de DNA , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Animais , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Helicases/genética , Regulação da Expressão Gênica , Hematopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Retroviridae , Transativadores/genética , DNA Metiltransferase 3B
10.
Biochem Biophys Res Commun ; 345(2): 595-601, 2006 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-16690027

RESUMO

Transforming growth factor-beta (TGF-beta) is the prototypical member of a family of growth factors that play important roles in normal development and human diseases. We identified the gene for fibroblast growth factor-binding protein 1 (FGF-BP1) as being significantly repressed following TGF-beta treatment. FGF-BP1 is an extracellular matrix bound protein that enhances fibroblast growth factor (FGF) signaling. We demonstrate here that TGF-beta signaling significantly represses FGF-BP1 expression in mesenchymal and neural crest cells undergoing in vitro smooth muscle differentiation. Analysis of the downstream signaling pathways shows that Smad2/3 are crucial for efficient FGF-BP1 repression by TGF-beta. Furthermore, we identified a novel element in the region from -785 to -782 bp of the FGF-BP1 promoter, which represents a known binding site for Hypermethylation in Cancer-1 (Hic-1), necessary for repression of FGF-BP1 by TGF-beta. These data define the molecular mechanism of transcriptional repression of an important target of TGF-beta signaling during angiogenesis.


Assuntos
Proteínas da Matriz Extracelular/fisiologia , Inativação Gênica/fisiologia , Regiões Promotoras Genéticas/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Sequência de Bases , Sítios de Ligação , Diferenciação Celular , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like , Músculo Liso/citologia , Músculo Liso/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
J Biol Chem ; 281(3): 1765-70, 2006 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-16317010

RESUMO

We recently reported that transforming growth factor (TGF)-beta induced the neural crest stem cell line Monc-1 to differentiate into a spindle-like contractile smooth muscle cell (SMC) phenotype and that Smad signaling played an important role in this phenomenon. In addition to Smad signaling, other pathways such as mitogen-activated protein kinase (MAPK), phosphoinositol-3 kinase, and RhoA have also been shown to mediate TGF-beta actions. The objectives of this study were to examine whether these signaling pathways contribute to TGF-beta-induced SMC development and to test whether Smad signaling cross-talks with other pathway(s) during SMC differentiation induced by TGF-beta. We demonstrate here that RhoA signaling is critical to TGF-beta-induced SMC differentiation. RhoA kinase (ROCK) inhibitor Y27632 significantly blocks the expression of multiple SMC markers such as smooth muscle alpha-actin, SM22alpha, and calponin in TGF-beta-treated Monc-1 cells. In addition, Y27632 reversed the cell morphology and abolished the contractility of TGF-beta-treated cells. RhoA signaling was activated as early as 5 min following TGF-beta addition. Dominant negative RhoA blocked nuclear translocation of Smad2 and Smad3 because of the inhibition of phosphorylation of both Smads and inhibited Smad-dependent SBE promoter activity, whereas constitutively active RhoA significantly enhanced SBE promoter activity. Consistent with these results, C3 exotoxin, an inhibitor of RhoA activation, significantly attenuated SBE promoter activity and inhibited Smad nuclear translocation. Taken together, these data point to a new role for RhoA as a modulator of Smad activation while regulating TGF-beta-induced SMC differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Amidas/farmacologia , Animais , Linhagem Celular , Camundongos , Camundongos Endogâmicos C3H , Relaxantes Musculares Centrais/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Piridinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA