Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 145: 88-98, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32485181

RESUMO

Class IIa histone deacetylases (HDACs) critically regulate cardiac function through the repression of the activity of myocyte enhancer factor 2 (MEF2)-dependent gene programs. Protein kinase D (PKD) and Ca2+/Calmodulin-dependent kinase II (CaMKII) activate MEF2 by phosphorylating distinct HDAC isoforms and thereby creating 14-3-3 binding sites for nucleo-cytoplasmic shuttling. Recently, it has been shown that this process is counteracted by cyclic AMP (cAMP)-dependent signaling. Here, we investigated the specific mechanisms of how cAMP-dependent signaling regulates distinct HDAC isoforms and determined their relative contributions to the protection from pathological MEF2 activation. We found that cAMP is sufficient to induce nuclear retention and to blunt phosphorylation of the 14-3-3 binding sites of HDAC5 (Ser259/498) and HDAC9 (Ser218/448) but not HDAC4 (Ser246/467/632). These regulatory events could be observed only in cardiomyocytes and myocyte-like cells but not in non-myocytes, pointing to an indirect myocyte-specific mode of action. Consistent with one previous report, we found that blunted phosphorylation of HDAC5 and HDAC9 was mediated by protein kinase A (PKA)-dependent inhibition of PKD. However, we show by the use of neonatal cardiomyocytes derived from genetic HDAC mouse models that endogenous HDAC5 but not HDAC9 contributes specifically to the repression of endogenous MEF2 activity. HDAC4 contributed significantly to the repression of MEF2 activity but based on the mechanistic findings of this study combined with previous results we attribute this to PKA-dependent proteolysis of HDAC4. Consistently, cAMP-induced repression of agonist-driven cellular hypertrophy was blunted in cardiomyocytes deficient for both HDAC5 and HDAC4. In conclusion, cAMP inhibits MEF2 through both nuclear accumulation of hypo-phosphorylated HDAC5 and through a distinct HDAC4-dependent mechanism.


Assuntos
AMP Cíclico/metabolismo , Histona Desacetilases/metabolismo , Fatores de Transcrição MEF2/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Animais Recém-Nascidos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ventrículos do Coração/patologia , Fatores de Transcrição MEF2/antagonistas & inibidores , Camundongos , Modelos Biológicos , Fosforilação , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais
2.
Eur Rev Med Pharmacol Sci ; 23(22): 9829-9839, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31799650

RESUMO

OBJECTIVE: Gallbladder carcinoma is a malignant tumor in the bile duct with poor prognosis. Although aberrant expression of miR-335 has been reported in the tumor tissues of gallbladder carcinoma, the biological role of miR-335 was still largely unknown. This study was intended to explore the role of miR-335 in the progression of gallbladder carcinoma. PATIENTS AND METHODS: The gallbladder carcinoma cell lines GBC-SD and SGC-996 were used in our study. MiR-335 mimic, miR-335 inhibitor, and si-myocyte enhancer factor 2D (MEF2D) were transfected into gallbladder carcinoma cells, respectively. (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay analysis was used to determine cell viability. The colony formation was also analyzed. Cell cycle progression was determined using flow cytometer. To verify the target gene of miR-335, the luciferase assay was used. RESULTS: MiR-335 overexpression inhibited cell viability and colony formation of GBC-SD and SGC-996 cells. The percentage of cells in first gap phase (G1)/resting phase (G0) was significantly increased, and the expression of cell division cycle 2 (cdc2) and cell division cycle 25 (cdc25) was decreased after miR-335 was overexpressed, indicating its role in inducing the cell cycle arrest of GBC-SD and SGC-996 cells. MEF2D was up-regulated in gallbladder cancer and associated with tumor size and clinical stage. Down-regulation of MEF2D inhibited cell viability and colony formation, induced cell cycle arrest in G1/G0 phase, and decreased the expression of cdc2 and cdc25 in GBC-SD and SGC-996 cells. Bioinformatics analysis by TargetScan and luciferase assay verified that MEF2D could be targeted by miR-335. Importantly, the effects of miR-335 inhibitor on cell growth were rescued by small interfering RNA of MEF2D (siMEF2D) in GBC-SD and SGC-996 cells. Besides, miR-335 overexpression increased cell sensitivity to 5-Fluoracil (Fu) treatment and decreased the expression levels of ATP-binding cassette transporter B1 (ABCB1) and ATP-binding cassette G2 (ABCG2) in GBC-SD and SGC-996 cells. CONCLUSIONS: MiR-335 participates in the progression of gallbladder carcinoma by targeting MEF2D. MiR-335 may be a potential therapeutic target for gallbladder carcinoma.


Assuntos
Fluoruracila/farmacologia , Neoplasias da Vesícula Biliar/tratamento farmacológico , MicroRNAs/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Neoplasias da Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/patologia , Humanos , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Pessoa de Meia-Idade
3.
PLoS One ; 14(10): e0223842, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31622977

RESUMO

The process by which fibroblasts are directly reprogrammed into cardiomyocytes involves two stages; initiation and maturation. Initiation represents the initial expression of factors that induce fibroblasts to transdifferentiate into cardiomyocytes. Following initiation, the cell undergoes a period of maturation before becoming a mature cardiomyocyte. We wanted to understand the role of cardiac development transcription factors in the maturation process. We directly reprogram fibroblasts into cardiomyocytes by a combination of miRNAs (miR combo). The ability of miR combo to induce cardiomyocyte-specific genes in fibroblasts was lost following the knockdown of the cardiac transcription factors Gata4, Mef2C, Tbx5 and Hand2 (GMTH). To further clarify the role of GMTH in miR combo reprogramming we utilized a modified CRISPR-Cas9 approach to activate endogenous GMTH genes. Importantly, both miR combo and the modified CRISPR-Cas9 approach induced comparable levels of GMTH expression. While miR combo was able to reprogram fibroblasts into cardiomyocyte-like cells, the modified CRISPR-Cas9 approach could not. Indeed, we found that cardiomyocyte maturation only occurred with very high levels of GMT expression. Taken together, our data indicates that while endogenous cardiac transcription factors are insufficient to reprogram fibroblasts into mature cardiomyocytes, endogenous cardiac transcription factors are necessary for expression of maturation genes.


Assuntos
Fator de Transcrição GATA4/genética , Proteínas com Domínio T/genética , Animais , Sistemas CRISPR-Cas/genética , Transdiferenciação Celular , Células Cultivadas , Reprogramação Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Fator de Transcrição GATA4/antagonistas & inibidores , Fator de Transcrição GATA4/metabolismo , Edição de Genes , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas com Domínio T/antagonistas & inibidores , Proteínas com Domínio T/metabolismo
4.
Biochem Biophys Res Commun ; 493(4): 1410-1417, 2017 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-28965945

RESUMO

Protein kinase B2 (AKT2) is implicated in diverse process of cardiomyocyte signaling including survival and metabolism. However, the role of AKT2 in myocardium development and the signaling pathway is rarely understood. Therefore, we sought to determine the effect of AKT2 deletion on heart development and its downstream targets. By using experimental animal models and neonatal rat cardiomyocytes (NRCMs), we observed that AKT2 deficiency induces retardation of heart development and increased systemic blood pressure (BP) without affecting cardiac function. Further investigation suggested that deficiency of AKT2 in myocardium results in diminished MEF2A abundance, which induced decreased size of cardiomyocytes. We additionally confirmed that EndoG, which is also regulated by AKT2, is a suppressor of MEF2A in myocardium. Finally, our results proved that AKT2 deficiency impairs the response to ß-adrenergic stimuli that normally causes hypertrophy in cardiomyocytes by downregulating MEF2A expression. Our data are the first to show the important role of AKT2 in determining the size of myocardium, its deficiency causes retardation of cardiomyocyte development. We also proved a novel pathway of heart development involving EndoG and MEF2A regulated by AKT2.


Assuntos
Endodesoxirribonucleases/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/deficiência , Animais , Diferenciação Celular , Tamanho Celular , Células Cultivadas , Endodesoxirribonucleases/antagonistas & inibidores , Endodesoxirribonucleases/genética , Técnicas de Silenciamento de Genes , Coração/crescimento & desenvolvimento , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Camundongos , Camundongos Knockout , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais
5.
JCI Insight ; 2(17)2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28878124

RESUMO

Cardiac hypertrophy, as a response to hemodynamic stress, is associated with cardiac dysfunction and death, but whether hypertrophy itself represents a pathological process remains unclear. Hypertrophy is driven by changes in myocardial gene expression that require the MEF2 family of DNA-binding transcription factors, as well as the nuclear lysine acetyltransferase p300. Here we used genetic and small-molecule probes to determine the effects of preventing MEF2 acetylation on cardiac adaptation to stress. Both nonacetylatable MEF2 mutants and 8MI, a molecule designed to interfere with MEF2-coregulator binding, prevented hypertrophy in cultured cardiac myocytes. 8MI prevented cardiac hypertrophy in 3 distinct stress models, and reversed established hypertrophy in vivo, associated with normalization of myocardial structure and function. The effects of 8MI were reversible, and did not prevent training effects of swimming. Mechanistically, 8MI blocked stress-induced MEF2 acetylation, nuclear export of class II histone deacetylases HDAC4 and -5, and p300 induction, without impeding HDAC4 phosphorylation. Correspondingly, 8MI transformed the transcriptional response to pressure overload, normalizing almost all 232 genes dysregulated by hemodynamic stress. We conclude that MEF2 acetylation is required for development and maintenance of pathological cardiac hypertrophy, and that blocking MEF2 acetylation can permit recovery from hypertrophy without impairing physiologic adaptation.


Assuntos
Cardiomegalia/prevenção & controle , Fatores de Transcrição MEF2/metabolismo , Acetilação , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Células Cultivadas , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Fatores de Transcrição MEF2/antagonistas & inibidores , Camundongos , Contração Miocárdica , Fosforilação , Ligação Proteica , Transporte Proteico , Ratos , Proteínas Repressoras/metabolismo , Estresse Fisiológico , Transcrição Gênica , Fatores de Transcrição de p300-CBP/biossíntese
6.
Sci Rep ; 6: 36146, 2016 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-27796324

RESUMO

The role of microRNA-214-3p (miR-214-3p) in cardiac hypertrophy was not well illustrated. The present study aimed to investigate the expression and potential target of miR-214-3p in angiotensin II (Ang-II)-induced mouse cardiac hypertrophy. In mice with either Ang-II infusion or transverse aortic constriction (TAC) model, miR-214-3p expression was markedly decreased in the hypertrophic myocardium. Down-regulation of miR-214-3p was observed in the myocardium of patients with cardiac hypertrophy. Expression of miR-214-3p was upregulated in Ang-II-induced hypertrophic neonatal mouse ventricular cardiomyocytes. Cardiac hypertrophy was attenuated in Ang-II-infused mice by tail vein injection of miR-214-3p. Moreover, miR-214-3p inhibited the expression of atrial natriuretic peptide (ANP) and ß-myosin heavy chain (MHC) in Ang-II-treated mouse cardiomyocytes in vitro. Myocyte-specific enhancer factor 2C (MEF2C), which was increased in Ang-II-induced hypertrophic mouse myocardium and cardiomyocytes, was identified as a target gene of miR-214-3p. Functionally, miR-214-3p mimic, consistent with MEF2C siRNA, inhibited cell size increase and protein expression of ANP and ß-MHC in Ang-II-treated mouse cardiomyocytes. The NF-κB signal pathway was verified to mediate Ang-II-induced miR-214-3p expression in cardiomyocytes. Taken together, our results revealed that MEF2C is a novel target of miR-214-3p, and attenuation of miR-214-3p expression may contribute to MEF2Cexpressionin cardiac hypertrophy.


Assuntos
Cardiomegalia/etiologia , Fatores de Transcrição MEF2/metabolismo , MicroRNAs/metabolismo , Angiotensina II/toxicidade , Animais , Antagomirs/metabolismo , Fator Natriurético Atrial/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Células Cultivadas , Modelos Animais de Doenças , Ventrículos do Coração/diagnóstico por imagem , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , NF-kappa B/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
7.
Oncotarget ; 7(21): 31053-66, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27105518

RESUMO

Cardiac fibrosis is an important pathological process of diabetic cardiomyopathy, the underlying mechanism remains elusive. This study sought to identify whether inhibition of Myocyte enhancer factor 2A (MEF2A) alleviates cardiac fibrosis by partially regulating Endothelial-to-mesenchymal transition (EndMT). We induced type 1 diabetes mellitus using the toxin streptozotocin (STZ) in mice and injected with lentivirus-mediated short-hairpin RNA (shRNA) in myocardium to inhibit MEF2A expression. Protein expression, histological and functional parameters were examined twenty-one weeks post-STZ injection. We found that Diabetes mellitus increased cardiac MEF2A expression, aggravated cardiac dysfunction and myocardial fibrosis through the accumulation of fibroblasts via EndMT. All of these features were abolished by MEF2A inhibition. MEF2A gene silencing by shRNA in cultured human umbilical vein endothelial cells (HUVECs) ameliorated high glucose-induced phenotypic transition and acquisition of mesenchymal markers through interaction with p38MAPK and Smad2. We conclude that inhibition of endothelial cell-derived MEF2A might be beneficial in the prevention of diabetes mellitus-induced cardiac fibrosis by partially inhibiting EndMT through interaction with p38MAPK and Smad2.


Assuntos
Diabetes Mellitus Experimental/terapia , Fibrose/terapia , Fatores de Transcrição MEF2/antagonistas & inibidores , Miocárdio/patologia , Animais , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Transição Epitelial-Mesenquimal , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Transdução de Sinais , Transfecção
8.
Ann N Y Acad Sci ; 1364: 62-73, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25847683

RESUMO

Parathyroid hormone (PTH) suppresses the expression of the bone formation inhibitor sclerostin (Sost) in osteocytes by inducing nuclear accumulation of histone deacetylases (HDACs) to inhibit the myocyte enhancer factor 2 (MEF2)-dependent Sost bone enhancer. Previous studies revealed that lipoprotein receptor-related protein 6 (LRP6) mediates the intracellular signaling activation and the anabolic bone effect of PTH. Here, we investigated whether LRP6 mediates the inhibitory effect of PTH on Sost using an osteoblast-specific Lrp6-knockout (LRP6-KO) mouse model. An increased level of Sost mRNA expression was detected in femur tissue from LRP6-KO mice, compared to wild-type littermates. The number of osteocytes expressing sclerostin protein was also increased in bone tissue of LRP6-KO littermates, indicating a negative regulatory role of LRP6 on Sost/sclerostin. In wild-type littermates, intermittent PTH treatment significantly suppressed Sost mRNA expression in bone and the number of sclerostin(+) osteocytes, while the effect of PTH was much less significant in LRP6-KO mice. Additionally, PTH-induced downregulation of MEF2C and 2D, as well as HDAC changes in osteocytes, were abrogated in LRP6-KO mice. These data indicate that LRP6 is required for PTH suppression of Sost expression.


Assuntos
Osso e Ossos/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Hormônio Paratireóideo/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Cruzamentos Genéticos , Fêmur , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilase 2/química , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Histona Desacetilases/química , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Injeções Subcutâneas , Peptídeos e Proteínas de Sinalização Intercelular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Fatores de Transcrição MEF2/agonistas , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Hormônio Paratireóideo/administração & dosagem , Hormônio Paratireóideo/farmacologia , RNA Mensageiro/metabolismo , Distribuição Aleatória
9.
J Steroid Biochem Mol Biol ; 148: 132-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25448741

RESUMO

Myogenic enhancer factor2 (Mef2) consists of a family of transcription factors involved in morphogenesis of skeletal, cardiac and smooth muscle cells. Among the four isoforms (Mef2A, 2B, 2C, and 2D), Mef2C was also found to play important roles in hematopoiesis. At myeloid progenitor level, Mef2C expression favors monocytic differentiation. Previous studies from our laboratory demonstrated that ERK5 was activated in 1,25-dihydroxyvitamin D3 (1,25D)-induced monocytic differentiation in AML cells and ERK5 activation was accompanied by increased Mef2C phosphorylation. We therefore examined the role of Mef2C in 1,25D-induced monocytic differentiation in AML cell lines (HL60, U937 and THP1) and found that knockdown of Mef2C with small interfering RNA (siRNA) significantly decreases the expression of the monocytic marker, CD14, without affecting the expression of the general myeloid marker, CD11b. CCAAT/enhancer-binding protein (C/EBP) ß, which can bind to CD14 promoter and increase its transcription, has been shown to be the downstream effector of 1,25D-induced monocytic differentiation in AML cells. When Mef2C was knocked down, expression of C/EBPß was reduced at both mRNA and protein levels. The protein expression levels of cell cycle regulators, p27(Kip1) and cyclin D1, were not affected by Mef2C knockdown, nor the monopoiesis related transcription factor, ATF2 (activating transcription factor 2). Thus, we conclude that 1,25D-induced monocytic differentiation, and CD14 expression in particular, are mediated through activation of ERK5-Mef2C-C/EBPß signaling pathway, and that Mef2C does not seem to modulate cell cycle progression.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Calcitriol/farmacologia , Diferenciação Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/patologia , Monócitos/patologia , Apoptose/efeitos dos fármacos , Western Blotting , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citometria de Fluxo , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Vitaminas/farmacologia
10.
J Biol Chem ; 290(2): 1256-68, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25416778

RESUMO

Skeletal muscle differentiation requires precisely coordinated transcriptional regulation of diverse gene programs that ultimately give rise to the specialized properties of this cell type. In Drosophila, this process is controlled, in part, by MEF2, the sole member of an evolutionarily conserved transcription factor family. By contrast, vertebrate MEF2 is encoded by four distinct genes, Mef2a, -b, -c, and -d, making it far more challenging to link this transcription factor to the regulation of specific muscle gene programs. Here, we have taken the first step in molecularly dissecting vertebrate MEF2 transcriptional function in skeletal muscle differentiation by depleting individual MEF2 proteins in myoblasts. Whereas MEF2A is absolutely required for proper myoblast differentiation, MEF2B, -C, and -D were found to be dispensable for this process. Furthermore, despite the extensive redundancy, we show that mammalian MEF2 proteins regulate a significant subset of nonoverlapping gene programs. These results suggest that individual MEF2 family members are able to recognize specific targets among the entire cohort of MEF2-regulated genes in the muscle genome. These findings provide opportunities to modulate the activity of MEF2 isoforms and their respective gene programs in skeletal muscle homeostasis and disease.


Assuntos
Diferenciação Celular/genética , Evolução Molecular , Fatores de Transcrição MEF2/biossíntese , Músculo Esquelético/crescimento & desenvolvimento , Isoformas de Proteínas/biossíntese , Animais , Células COS , Chlorocebus aethiops , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Mamíferos/genética , Mamíferos/crescimento & desenvolvimento , Camundongos , Desenvolvimento Muscular/genética , Mioblastos/citologia , Mioblastos/metabolismo , Isoformas de Proteínas/genética
11.
Nucleic Acids Res ; 42(1): 224-34, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24078251

RESUMO

Myocyte enhancer factor 2 (MEF2) is a family of transcription factors that regulates many processes, including muscle differentiation. Due to its many target genes, MEF2D requires tight regulation of transcription activity over time and by location. Epigenetic modifiers have been suggested to regulate MEF2-dependent transcription via modifications to histones and MEF2. However, the modulation of MEF2 activity by lysine methylation, an important posttranslational modification that alters the activities of transcription factors, has not been studied. We report the reversible lysine methylation of MEF2D by G9a and LSD1 as a regulatory mechanism of MEF2D activity and skeletal muscle differentiation. G9a methylates lysine-267 of MEF2D and represses its transcriptional activity, but LSD1 counteracts it. This residue is highly conserved between MEF2 members in mammals. During myogenic differentiation of C2C12 mouse skeletal muscle cells, the methylation of MEF2D by G9a decreased, on which MEF2D-dependent myogenic genes were upregulated. We have also identified lysine-267 as a methylation/demethylation site and demonstrate that the lysine methylation state of MEF2D regulates its transcriptional activity and skeletal muscle cell differentiation.


Assuntos
Diferenciação Celular/genética , Lisina/metabolismo , Fatores de Transcrição MEF2/metabolismo , Mioblastos Esqueléticos/metabolismo , Animais , Linhagem Celular , Cromatina/metabolismo , Células HEK293 , Histona Desmetilases , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/química , Metilação , Camundongos , Mioblastos Esqueléticos/citologia , Oxirredutases N-Desmetilantes/metabolismo , Processamento de Proteína Pós-Traducional , Transcrição Gênica
12.
Cardiovasc Toxicol ; 13(4): 316-22, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23632743

RESUMO

Histone acetylase (HAT) p300 plays an important role in the regulation of cardiac gene expression. During cardiac development, bone morphogenetic protein (BMP)-2 induces the expression of cardiac transcription factors. However, the underlying molecular mechanism(s) is not clear. In the present study, we tested the hypothesis that p300-mediated histone acetylation was essential for the regulation of cardiac transcription factors by BMP2. Cultured rat H9c2 embryonic cardiac myocytes (H9c2 cells) were transfected with recombinant adenoviruses expressing human BMP2 (AdBMP2) with or without curcumin, a specific p300-HAT inhibitor. Quantitative real-time RT-PCR analysis showed that curcumin substantially inhibited both AdBMP2-induced and basal expression levels of cardiac transcription factors GATA4 and MEF2C, but not Tbx5. Similarly, chromatin immunoprecipitation (ChIP) analysis showed that curcumin inhibited both AdBMP2-induced and basal histone H3 acetylation levels in the promoter regions of GATA4 and MEF2C, but not of Tbx5. In addition, curcumin selectively suppressed AdBMP2-induced expression of HAT p300, but not HAT GCN5 in H9c2 cells. The data indicated that inhibition of histone H3 acetylation with curcumin diminished BMP2-induced expression of GATA4 and MEF2C, suggesting that p300-mediated histone acetylation was essential for the regulation of GATA4 and MEF2C by BMP2 in H9c2 cells.


Assuntos
Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/biossíntese , Proteína p300 Associada a E1A/fisiologia , Fator de Transcrição GATA4/fisiologia , Histonas/antagonistas & inibidores , Acetilação/efeitos dos fármacos , Animais , Células Cultivadas , Curcumina/farmacologia , Fator de Transcrição GATA4/antagonistas & inibidores , Histonas/metabolismo , Humanos , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/fisiologia , Ratos , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...