Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nat Commun ; 7: 12224, 2016 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-27426629

RESUMO

The trimethylation of histone H3 on lysine 9 (H3K9me3) - a mark recognized by HP1 that depends on the Suv39h lysine methyltransferases (KMTs) - has provided a basis for the reader/writer model to explain HP1 accumulation at pericentric heterochromatin in mammals. Here, we identify the Suv39h1 paralog, as a unique enhancer of HP1α sumoylation both in vitro and in vivo. The region responsible for promoting HP1α sumoylation (aa1-167) is distinct from the KMT catalytic domain and mediates binding to Ubc9. Tethering the 1-167 domain of Suv39h1 to pericentric heterochromatin, but not mutants unable to bind Ubc9, accelerates the de novo targeting of HP1α to these domains. Our results establish an unexpected feature of Suv39h1, distinct from the KMT activity, with a major role for heterochromatin formation. We discuss how linking Suv39h1 to the SUMO pathway provides conceptual implications for our general view on nuclear domain organization and physiological functions.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Heterocromatina/metabolismo , Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Aminoácidos/metabolismo , Animais , Homólogo 5 da Proteína Cromobox , Metiltransferases/química , Camundongos , Modelos Biológicos , Células NIH 3T3 , Ligação Proteica , Domínios Proteicos , Transporte Proteico , Proteínas Repressoras/química , Sumoilação , Enzimas de Conjugação de Ubiquitina/metabolismo
2.
Mol Cell Oncol ; 3(6): e1225546, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28090575

RESUMO

The Suv39h lysine methyltransferases, known as key enzymes responsible for histone H3 lysine 9 methylation, are critical for heterochromatin protein 1 enrichment at constitutive heterochromatin. Our recent findings reveal a new role for the Suv39h1 paralog that links it to SUMO pathway function at constitutive heterochromatin.

3.
Nature ; 487(7406): 249-53, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22763435

RESUMO

During immune responses, naive CD4+ T cells differentiate into several T helper (TH) cell subsets under the control of lineage-specifying genes. These subsets (TH1, TH2 and TH17 cells and regulatory T cells) secrete distinct cytokines and are involved in protection against different types of infection. Epigenetic mechanisms are involved in the regulation of these developmental programs, and correlations have been drawn between the levels of particular epigenetic marks and the activity or silencing of specifying genes during differentiation. Nevertheless, the functional relevance of the epigenetic pathways involved in TH cell subset differentiation and commitment is still unclear. Here we explore the role of the SUV39H1­H3K9me3­HP1α silencing pathway in the control of TH2 lineage stability. This pathway involves the histone methylase SUV39H1, which participates in the trimethylation of histone H3 on lysine 9 (H3K9me3), a modification that provides binding sites for heterochromatin protein 1α (HP1α) and promotes transcriptional silencing. This pathway was initially associated with heterochromatin formation and maintenance but can also contribute to the regulation of euchromatic genes. We now propose that the SUV39H1­H3K9me3­HP1α pathway participates in maintaining the silencing of TH1 loci, ensuring TH2 lineage stability. In TH2 cells that are deficient in SUV39H1, the ratio between trimethylated and acetylated H3K9 is impaired, and the binding of HP1α at the promoters of silenced TH1 genes is reduced. Despite showing normal differentiation, both SUV39H1-deficient TH2 cells and HP1α-deficient TH2 cells, in contrast to wild-type cells, expressed TH1 genes when recultured under conditions that drive differentiation into TH1 cells. In a mouse model of TH2-driven allergic asthma, the chemical inhibition or loss of SUV39H1 skewed T-cell responses towards TH1 responses and decreased the lung pathology. These results establish a link between the SUV39H1­H3K9me3­HP1α pathway and the stability of TH2 cells, and they identify potential targets for therapeutic intervention in TH2-cell-mediated inflammatory diseases.


Assuntos
Epigênese Genética , Células Th2/citologia , Células Th2/imunologia , Animais , Asma/enzimologia , Asma/imunologia , Asma/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Histonas/metabolismo , Masculino , Metiltransferases/deficiência , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Proteínas Repressoras/deficiência , Proteínas Repressoras/metabolismo , Células Th1/metabolismo , Células Th2/enzimologia
4.
Nat Struct Mol Biol ; 19(4): 458-60, 2012 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-22388734

RESUMO

SUMOylation promotes targeting of HP1α to pericentric heterochromatin. Here we identify the SUMO-specific protease SENP7 in mouse as a maintenance factor for HP1α accumulation at this location. SENP7 interacts directly with HP1α, localizes at HP1-enriched pericentric domains and can deconjugate SUMOylated HP1α in vivo. Depletion of SENP7 delocalizes HP1α from pericentric heterochromatin without affecting H3K9me3 levels. We propose that following targeting of HP1α, a subsequent deSUMOylation event enables HP1α retention at these domains.


Assuntos
Proteínas Cromossômicas não Histona/análise , Proteínas Cromossômicas não Histona/metabolismo , Endopeptidases/análise , Endopeptidases/metabolismo , Heterocromatina/metabolismo , Proteína SUMO-1/metabolismo , Animais , Homólogo 5 da Proteína Cromobox , Endopeptidases/genética , Deleção de Genes , Camundongos , Células NIH 3T3
6.
Nat Genet ; 43(3): 220-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21317888

RESUMO

HP1 enrichment at pericentric heterochromatin is considered important for centromere function. Although HP1 binding to H3K9me3 can explain its accumulation at pericentric heterochromatin, how it is initially targeted there remains unclear. Here, in mouse cells, we reveal the presence of long nuclear noncoding transcripts corresponding to major satellite repeats at the periphery of pericentric heterochromatin. Furthermore, we find that major transcripts in the forward orientation specifically associate with SUMO-modified HP1 proteins. We identified this modification as SUMO-1 and mapped it in the hinge domain of HP1α. Notably, the hinge domain and its SUMOylation proved critical to promote the initial targeting of HP1α to pericentric domains using de novo localization assays, whereas they are dispensable for maintenance of HP1 domains. We propose that SUMO-HP1, through a specific association with major forward transcript, is guided at the pericentric heterochromatin domain to seed further HP1 localization.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Heterocromatina/metabolismo , Sumoilação , Animais , Centrômero/metabolismo , Homólogo 5 da Proteína Cromobox , Camundongos , Estrutura Terciária de Proteína , RNA , Sequências Repetitivas de Ácido Nucleico
7.
EMBO J ; 26(15): 3616-28, 2007 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-17627279

RESUMO

A critical step of neuronal terminal differentiation is the permanent withdrawal from the cell cycle that requires the silencing of genes that drive mitosis. Here, we describe that the alpha isoform of the heterochromatin protein 1 (HP1) protein family exerts such silencing on several E2F-targeted genes. Among the different isoforms, HP1alpha levels progressively increase throughout differentiation and take over HP1gamma binding on E2F sites in mature neurons. When overexpressed, only HP1alpha is able to ensure a timed repression of E2F genes. Specific inhibition of HP1alpha expression drives neuronal progenitors either towards death or cell cycle progression, yet preventing the expression of the neuronal marker microtubule-associated protein 2. Furthermore, we provide evidence that this mechanism occurs in cerebellar granule neurons in vivo, during the postnatal development of the cerebellum. Finally, our results suggest that E2F-targeted genes are packaged into higher-order chromatin structures in mature neurons relative to neuroblasts, likely reflecting a transition from a 'repressed' versus 'silenced' status of these genes. Together, these data present new epigenetic regulations orchestrated by HP1 isoforms, critical for permanent cell cycle exit during neuronal differentiation.


Assuntos
Diferenciação Celular , Proteínas Cromossômicas não Histona/fisiologia , Fatores de Transcrição E2F/fisiologia , Inativação Gênica , Neurônios/citologia , Animais , Sequência de Bases , Linhagem da Célula , Células Cultivadas , Homólogo 5 da Proteína Cromobox , Citometria de Fluxo , Camundongos , RNA Interferente Pequeno
8.
J Cell Biol ; 166(4): 493-505, 2004 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-15302854

RESUMO

Heterochromatin is thought to play a critical role for centromeric function. However, the respective contributions of the distinct repetitive sequences found in these regions, such as minor and major satellites in the mouse, have remained largely unsolved. We show that these centric and pericentric repeats on the chromosomes have distinct heterochromatic characteristics in the nucleus. Major satellites from different chromosomes form clusters associated with heterochromatin protein 1alpha, whereas minor satellites are individual entities associated with centromeric proteins. Both regions contain methylated histone H3 (Me-K9 H3) but show different micrococcal nuclease sensitivities. A dinucleosome repeating unit is found specifically associated with major satellites. These domains replicate asynchronously, and chromatid cohesion is sustained for a longer time in major satellites compared with minor satellites. Such prolonged cohesion in major satellites is lost in the absence of Suv39h histone methyltransferases. Thus, we define functionally independent centromeric subdomains, which spatio-temporal isolation is proposed to be important for centromeric cohesion and dissociation during chromosome segregation.


Assuntos
Centrômero/ultraestrutura , Heterocromatina/química , Sequências Repetitivas de Ácido Nucleico , Animais , Bromodesoxiuridina/farmacologia , Linhagem Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromatina/ultraestrutura , Cromossomos/ultraestrutura , DNA Satélite , Histonas/metabolismo , Processamento de Imagem Assistida por Computador , Hibridização in Situ Fluorescente , Interfase , Cinetocoros/ultraestrutura , Metiltransferases/genética , Camundongos , Nuclease do Micrococo/metabolismo , Repetições de Microssatélites , Família Multigênica , Mutação , Células NIH 3T3 , Plasmídeos/metabolismo , Proteínas Repressoras/genética , Fase S , Troca de Cromátide Irmã , Fatores de Tempo
10.
Nat Genet ; 30(3): 329-34, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11850619

RESUMO

Post-translational modification of histone tails is thought to modulate higher-order chromatin structure. Combinations of modifications including acetylation, phosphorylation and methylation have been proposed to provide marks recognized by specific proteins. This is exemplified, in both mammalian cells and fission yeast, by transcriptionally silent constitutive pericentric heterochromatin. Such heterochromatin contains histones that are generally hypoacetylated and methylated by Suv39h methyltransferases at lysine 9 of histone H3 (H3-K9). Each of these modification states has been implicated in the maintenance of HP1 protein-binding at pericentric heterochromatin, in transcriptional silencing and in centromere function. In particular, H3-K9 methylation is thought to provide a marking system for the establishment and maintenance of stably repressed regions and heterochromatin subdomains. To address the question of how these two types of modifications, as well as other unidentified parameters, function to maintain pericentric heterochromatin, we used a combination of histone deacetylase inhibitors, RNAse treatments and an antibody raised against methylated branched H3-K9 peptides. Our results show that both H3-K9 acetylation and methylation can occur on independent sets of H3 molecules in pericentric heterochromatin. In addition, we identify an RNA- and histone modification-dependent structure that brings methylated H3-K9 tails together in a specific configuration required for the accumulation of HP1 proteins in these domains.


Assuntos
Heterocromatina/metabolismo , Histonas/metabolismo , RNA/metabolismo , Imunofluorescência , Heterocromatina/química , Conformação Proteica , RNA/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA