Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1803(10): 1186-97, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20621129

RESUMO

Class IIa HDACs including HDAC7 play a role in gene expression, cell differentiation, and animal development through their association with transcription factors such as myogenic enhancer factors 2 (MEF2s). In this study, we show that endogenous HDAC7 localizes to both the nucleus and the cytoplasm of C2C12 myoblasts but is exclusively retained in the cytoplasm of myotubes after completion of differentiation process. To elucidate the role of differential distribution of HDAC7 during myogenesis, we examined the effects of stably expressed HDAC7 mutants on myogenesis. Expression of nuclear-retained HDAC7 mutants significantly inhibits myogenesis in C2C12 cells and reduces the expression of muscle-specific myosin heavy chain (MHC) and myogenin. The inhibition in myocyte differentiation can be partially relieved by introduction of a mutation disrupting HDAC7:MEF2 interaction. Since phosphorylation of HDAC7 plays an important role in its nucleocytoplasmic shuttling, we further investigated the expression and distribution of phosphorylated HDAC7. To our surprise, the phosphorylation levels of HDAC7 at S344 and S479 were slightly decreased upon differentiation, whereas the phosphorylation of S178 was unchanged. Interestingly, a significant fraction of pS344- and/or pS479-HDAC7 localized to plasma membrane of myotubes. In addition, Ser178-phosphorylated (pS178) HDAC7 displays a predominantly actin filament-like structure before muscle differentiation. Consistent with this notion, HDAC7 partially colocalized with actin filaments; in particular, pS178-HDAC7 largely colocalized with actin filaments as indicated by phalloidin counter staining in myocytes. Furthermore, C2C12 cells expressing nuclear-retained HDAC7 display defects in migration. Our results provide novel insight into the mechanisms that regulate myocyte differentiation and migration by controlling the subcellular distribution of HDAC7 in differentiating myoblasts.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Histona Desacetilases/metabolismo , Mioblastos/metabolismo , Actinas/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Movimento Celular/genética , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Histona Desacetilases/genética , Immunoblotting , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia Confocal , Mutação , Mioblastos/citologia , Miogenina/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fosforilação , Serina/genética , Serina/metabolismo
2.
FEBS Lett ; 580(21): 5096-104, 2006 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-16956611

RESUMO

CRM1, 14-3-3 proteins, and CaMK play important roles in trafficking of HDAC7, but the interplay between these proteins in this process is not clearly understood. Here, we show that CRM1 is capable of promoting cytoplasmic localization of wild-type and mutant HDAC7 (S178A/S344A/S479A), which is normally found in the nucleus. Using phospho-specific antibodies to HDAC7, we demonstrate that CaMK I promotes phosphorylation of S178, S344, and S479 of HDAC7. We also show that endogenous S178-phosphorylated HDAC7 is localized in both the nucleus and the cytoplasm, whereas S344- and S479-phosphorylated HDAC7 are exclusively localized in the nucleus. An HDAC7 mutant, S178E/S344E/S479E, which lost the ability to bind 14-3-3s, is localized in both the nucleus and the cytoplasm. Furthermore, the nuclear export of S178E/S344E/S479E is inhibited by LMB, but is enhanced by the CRM1. Taken together, these results strongly suggest that CRM1 mediated-nuclear export of HDAC7 is independent of HDAC7 phosphorylation and its association with 14-3-3s.


Assuntos
Proteínas 14-3-3/metabolismo , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Histona Desacetilases/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Citoplasma/metabolismo , Células HeLa , Histona Desacetilases/química , Humanos , Camundongos , Proteínas Mutantes/metabolismo , Fosforilação , Ligação Proteica , Transporte Proteico , Proteína Exportina 1
3.
Mol Cell Biol ; 33(8): 1546-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23401860

RESUMO

Nucleocytoplasmic shuttling of class IIa of histone deacetylases (HDACs) is a key mechanism that controls cell fate and animal development. We have identified the filamin B (FLNB) as a novel HDAC7-interacting protein that is required for temporal and spatial regulation of vascular endothelial growth factor (VEGF)-mediated HDAC7 cytoplasmic sequestration. This interaction occurs in the cytoplasm and requires monoubiquitination of an evolutionarily conserved lysine 1147 (K1147) in the immunoglobulin (Ig)-like repeat 10 (R10) of FLNB and the nuclear localization sequence of HDAC7. Inhibition of protein kinase C (PKC) blocks VEGF-induced ubiquitination of FLNB and its interaction with HDAC7. Small interfering RNA (siRNA) knockdown of FLNB or ubiquitin (Ub) in human primary endothelial cells blocks VEGF-mediated cytoplasmic accumulation of HDAC7, reduces VEGF-induced expression of the HDAC7 target genes Mmp-10 and Nur77, and inhibits VEGF-induced vascular permeability. Using dominant negative mutants and rescue experiments, we demonstrate the functional significance of FLNB K1147 to interfere with the ability of phorbol myristate acetate (PMA) to promote FLNB-mediated cytoplasmic accumulation of HDAC7. Taken together, our data show that VEGF and PKC promote degradation-independent protein ubiquitination of FLNB to control intracellular trafficking of HDAC7.


Assuntos
Proteínas Contráteis/metabolismo , Histona Desacetilases/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteína Quinase C/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Sequência de Aminoácidos , Carbazóis/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Proteínas Contráteis/genética , Células Endoteliais/metabolismo , Filaminas , Células HeLa , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Metaloproteinase 10 da Matriz/biossíntese , Proteínas dos Microfilamentos/genética , Neovascularização Fisiológica , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/biossíntese , Proteína Quinase C/antagonistas & inibidores , Estrutura Terciária de Proteína , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno , Acetato de Tetradecanoilforbol/farmacologia , Ubiquitina/genética , Ubiquitinação , Fatores de Crescimento do Endotélio Vascular/genética
4.
Mol Biol Cell ; 19(7): 3020-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18463162

RESUMO

Promyelocytic leukemia protein (PML) nuclear bodies (NBs) are dynamic subnuclear compartments that play roles in several cellular processes, including apoptosis, transcriptional regulation, and DNA repair. Histone deacetylase (HDAC) 7 is a potent corepressor that inhibits transcription by myocyte enhancer factor 2 (MEF2) transcription factors. We show here that endogenous HDAC7 and PML interact and partially colocalize in PML NBs. Tumor necrosis factor (TNF)-alpha treatment recruits HDAC7 to PML NBs and enhances association of HDAC7 with PML in human umbilical vein endothelial cells. Consequently, TNF-alpha promotes dissociation of HDAC7 from MEF2 transcription factors and the promoters of MEF2 target genes such as matrix metalloproteinase (MMP)-10, leading to accumulation of MMP-10 mRNA. Conversely, knockdown of PML enhances the association between HDAC7 and MEF2 and decreases MMP-10 mRNA accumulation. Accordingly, ectopic expression of PML recruits HDAC7 to PML NBs and leads to activation of MEF2 reporter activity. Notably, small interfering RNA knockdown of PML decreases basal and TNF-alpha-induced MMP-10 mRNA accumulation. Our results reveal a novel mechanism by which PML sequesters HDAC7 to relieve repression and up-regulate gene expression.


Assuntos
Regulação Enzimológica da Expressão Gênica , Histona Desacetilases/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Animais , Apoptose , Células HeLa , Humanos , Fatores de Transcrição MEF2 , Metaloproteinase 10 da Matriz/metabolismo , Camundongos , Microscopia Confocal , Fatores de Regulação Miogênica/metabolismo , Mapeamento de Interação de Proteínas , RNA Interferente Pequeno/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
5.
Mol Cell Biol ; 28(18): 5658-67, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18625722

RESUMO

Promyelocytic leukemia protein (PML) sumoylation has been proposed to control the formation of PML nuclear bodies (NBs) and is crucial for PML-dependent cellular processes, including apoptosis and transcriptional regulation. However, the regulatory mechanisms of PML sumoylation and its specific roles in the formation of PML NBs remain largely unknown. Here, we show that histone deacetylase 7 (HDAC7) knockdown reduces the size and the number of the PML NBs in human umbilical vein endothelial cells (HUVECs). HDAC7 coexpression stimulates PML sumoylation independent of its HDAC activity. Furthermore, HDAC7 associates with the E2 SUMO ligase, Ubc9, and stimulates PML sumoylation in vitro, suggesting that it possesses a SUMO E3 ligase-like activity to promote PML sumoylation. Importantly, HDAC7 knockdown inhibits tumor necrosis factor alpha-induced PML sumoylation and the formation of PML NBs in HUVECs. These results demonstrate a novel function of HDAC7 and provide a regulatory mechanism of PML sumoylation.


Assuntos
Histona Desacetilases/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Proteínas Nucleares/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células HeLa , Histona Desacetilases/genética , Humanos , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Supressoras de Tumor/genética , Veias Umbilicais/citologia
6.
J Biol Chem ; 281(46): 35070-80, 2006 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-16980305

RESUMO

Histone deacetylase 7 (HDAC7) is a member of class IIa HDACs that regulate myocyte enhancer factor-2 (MEF2)-mediated transcription and participate in multiple cellular processes such as T cell apoptosis. We have identified alpha-actinin 1 and 4 as class IIa HDAC-interacting proteins. The interaction domains are mapped to C terminus of alpha-actinin 4 and amino acids 72-172 of HDAC7. A point mutation in HDAC7 that disrupts its association with MEF2A also disrupts its association with alpha-actinin 4, indicating that MEF2A and alpha-actinin 4 binding sites largely overlap. We have also isolated a novel splice variant of alpha-actinin 4 that is predominantly localized in the nucleus, a pattern distinct from the full-length alpha-actinin 4, which is primarily distributed in the cytoplasm and plasma membrane. Using small interfering RNA, chromatin immunoprecipitation, and transient transfection assays, we show that alpha-actinin 4 potentiates expression of TAF55, a putative MEF2 target gene. Loss of MEF2A interaction correlates with loss of the ability of alpha-actinin 4 to potentiate TAF55 promoter activity. Ectopic expression of alpha-actinin 4, but not the mutant defective in MEF2A association, leads to disruption of HDAC7.MEF2A association and enhancement of MEF2-mediated transcription. Taken together, we have identified a novel mechanism by which HDAC7 activity is negatively regulated and uncovered a previously unknown function of alpha-actinin 4.


Assuntos
Actinina/metabolismo , Histona Desacetilases/metabolismo , Proteínas de Domínio MADS/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fatores de Regulação Miogênica/metabolismo , Sequência de Aminoácidos , Células HeLa , Humanos , Proteínas de Domínio MADS/genética , Fatores de Transcrição MEF2 , Fatores de Regulação Miogênica/genética , Transcrição Gênica , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA