Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Front Cell Dev Biol ; 10: 978931, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36158208

RESUMO

Histone acetylation and methylation are epigenetic modifications that are dynamically regulated by chromatin modifiers to precisely regulate gene expression. However, the interplay by which histone modifications are synchronized to coordinate cellular differentiation is not fully understood. In this study, we demonstrate a relationship between BRD4, a reader of acetylation marks, and G9a, a writer of methylation marks in the regulation of myogenic differentiation. Using loss- and gain-of-function studies, as well as a pharmacological inhibition of its activity, we examined the mechanism by which BRD4 regulates myogenesis. Transcriptomic analysis using RNA sequencing revealed that a number of myogenic differentiation genes are downregulated in Brd4-depleted cells. Interestingly, some of these genes were upregulated upon G9a knockdown, indicating that BRD4 and G9a play opposing roles in the control of myogenic gene expression. Remarkably, the differentiation defect caused by Brd4 knockdown was rescued by inhibition of G9a methyltransferase activity. These findings demonstrate that the absence of BRD4 results in the upregulation of G9a activity and consequently impaired myogenic differentiation. Collectively, our study identifies an interdependence between BRD4 and G9a for the precise control of transcriptional outputs to regulate myogenesis.

2.
Cell Death Dis ; 10(3): 250, 2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30867409

RESUMO

The lysine methyltransferase G9a plays a role in many cellular processes. It is a potent repressor of gene expression, a function attributed to its ability to methylate histone and non-histone proteins. Paradoxically, in some instances, G9a can activate gene expression. However, regulators of G9a expression and activity are poorly understood. In this study, we report that endogenous G9a is SUMOylated in proliferating skeletal myoblasts. There are four potential SUMOylation consensus motifs in G9a. Mutation of all four acceptor lysine residues [K79, K152, K256, and K799] inhibits SUMOylation. Interestingly, SUMOylation does not impact G9a-mediated repression of MyoD transcriptional activity or myogenic differentiation. In contrast, SUMO-defective G9a is unable to enhance proliferation of myoblasts. Using complementation experiments, we show that the proliferation defect of primary myoblasts from conditional G9a-deficient mice is rescued by re-expression of wild-type, but not SUMOylation-defective, G9a. Mechanistically, SUMOylation acts as signal for PCAF (P300/CBP-associated factor) recruitment at E2F1-target genes. This results in increased histone H3 lysine 9 acetylation marks at E2F1-target gene promoters that are required for S-phase progression. Our studies provide evidence by which SUMO modification of G9a influences the chromatin environment to impact cell cycle progression.


Assuntos
Proliferação de Células/genética , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Desenvolvimento Muscular , Mioblastos Esqueléticos/metabolismo , Sumoilação , Animais , Células COS , Diferenciação Celular/genética , Chlorocebus aethiops , Cromatina/metabolismo , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Células HEK293 , Antígenos de Histocompatibilidade/química , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Humanos , Lisina/química , Lisina/metabolismo , Metilação , Camundongos , Camundongos Transgênicos , Desenvolvimento Muscular/genética , Proteína MyoD/metabolismo , Mioblastos Esqueléticos/citologia , Processamento de Proteína Pós-Traducional , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
3.
Methods Mol Biol ; 1889: 43-54, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30367408

RESUMO

Chromatin immunoprecipitation (ChIP) is a powerful and sensitive technique that is widely used to study DNA-protein interactions. It enables an unbiased genome-wide analysis of transcriptional changes during several biological processes including cellular differentiation. Here, we describe a step-by-step protocol to identify histone modifications, transcription factor, and co-factor binding to chromatin in skeletal myoblasts. We discuss critical steps during cell harvesting, sonication, and immunoprecipitation and provide notes to evade common pitfalls.


Assuntos
Imunoprecipitação da Cromatina , Mioblastos Esqueléticos/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Análise de Dados , Sequenciamento de Nucleotídeos em Larga Escala , Histonas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
4.
FASEB J ; 33(1): 219-230, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29995440

RESUMO

Aurora kinases are critical mitotic serine/threonine kinases and are often implicated in tumorigenesis. Recent studies of the interphase functions for aurora kinase (Aurk)A have considerably expanded our understanding of its role beyond mitosis. To identify the unknown targets of AurkA, we used peptide array-based screening and found E2F4 to be a novel substrate. Phosphorylation of E2F4 by AurkA at Ser75 regulates its DNA binding and subcellular localization. Because E2F4 plays an important role in skeletal muscle differentiation, we attempted to gain insight into E2F4 phosphorylation in this context. We observed that a block in E2F4 phosphorylation retained it better within the nucleus and inhibited muscle differentiation. RNA sequencing analysis revealed a perturbation of the gene network involved in the process of muscle differentiation and mitochondrial biogenesis. Collectively, our findings establish a novel role of AurkA in the process of skeletal muscle differentiation.-Dhanasekaran, K., Bose, A., Rao, V. J., Boopathi, R., Shankar, S. R., Rao, V. K., Swaminathan, A., Vasudevan, M., Taneja, R., Kundu, T. K. Unravelling the role of aurora A beyond centrosomes and spindle assembly: implications in muscle differentiation.


Assuntos
Aurora Quinase A/metabolismo , Diferenciação Celular , Centrossomo/metabolismo , Fator de Transcrição E2F4/metabolismo , Músculo Esquelético/citologia , Mioblastos/citologia , Fuso Acromático/metabolismo , Animais , Aurora Quinase A/genética , Ciclo Celular , Células Cultivadas , Fator de Transcrição E2F4/genética , Células HEK293 , Humanos , Camundongos , Mitose , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Fosforilação
5.
Nucleic Acids Res ; 44(17): 8129-43, 2016 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-27229136

RESUMO

Differentiation of skeletal muscle cells, like most other cell types, requires a permanent exit from the cell cycle. The epigenetic programming underlying these distinct cellular states is not fully understood. In this study, we provide evidence that the lysine methyltransferase G9a functions as a central axis to regulate proliferation and differentiation of skeletal muscle cells. Transcriptome analysis of G9a knockdown cells revealed deregulation of many cell cycle regulatory genes. We demonstrate that G9a enhances cellular proliferation by two distinct mechanisms. G9a blocks cell cycle exit via methylation-dependent transcriptional repression of the MyoD target genes p21(Cip/Waf1) and Rb1. In addition, it activates E2F1-target genes in a methyltransferase activity-independent manner. We show that G9a is present in the E2F1/PCAF complex, and enhances PCAF occupancy and histone acetylation marks at E2F1-target promoters. Interestingly, G9a preferentially associates with E2F1 at the G1/S phase and with MyoD at the G2/M phase. Our results provide evidence that G9a functions both as a co-activator and a co-repressor to enhance cellular proliferation and inhibit myogenic differentiation.


Assuntos
Ciclo Celular , Diferenciação Celular , Histona-Lisina N-Metiltransferase/metabolismo , Desenvolvimento Muscular , Animais , Ciclo Celular/genética , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/genética , Fator de Transcrição E2F1/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Lisina/metabolismo , Metilação , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular/genética , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição de p300-CBP/metabolismo
6.
J Cell Mol Med ; 18(2): 344-54, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24393246

RESUMO

Clinically reported reparative benefits of mesenchymal stromal cells (MSCs) are majorly attributed to strong immune-modulatory abilities not exactly shared by fibroblasts. However, MSCs remain heterogeneous populations, with unique tissue-specific subsets, and lack of clear-cut assays defining therapeutic stromal subsets adds further ambiguity to the field. In this context, in-depth evaluation of cellular characteristics of MSCs from proximal oro-facial tissues: dental pulp (DPSCs) and periodontal ligament (PDLSCs) from identical donors provides an opportunity to evaluate exclusive niche-specific influences on multipotency and immune-modulation. Exhaustive cell surface profiling of DPSCs and PDLSCs indicated key differences in expression of mesenchymal (CD105) and pluripotent/multipotent stem cell-associated cell surface antigens: SSEA4, CD117, CD123 and CD29. DPSCs and PDLSCs exhibited strong chondrogenic potential, but only DPSCs exhibited adipogenic and osteogenic propensities. PDLSCs expressed immuno-stimulatory/immune-adhesive ligands like HLA-DR and CD50, upon priming with IFNγ, unlike DPSCs, indicating differential response patterns to pro-inflammatory cytokines. Both DPSCs and PDLSCs were hypo-immunogenic and did not elicit robust allogeneic responses despite exposure to IFNγ or TNFα. Interestingly, only DPSCs attenuated mitogen-induced lympho-proliferative responses and priming with either IFNγ or TNFα enhanced immuno-modulation capacity. In contrast, primed or unprimed PDLSCs lacked the ability to suppress polyclonal T cell blast responses. This study indicates that stromal cells from even topographically related tissues do not necessarily share identical MSC properties and emphasizes the need for a thorough functional testing of MSCs from diverse sources with respect to multipotency, immune parameters and response to pro-inflammatory cytokines before translational usage.


Assuntos
Polpa Dentária/citologia , Células-Tronco Mesenquimais/citologia , Ligamento Periodontal/citologia , Adipócitos/citologia , Adipócitos/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Polpa Dentária/efeitos dos fármacos , Polpa Dentária/metabolismo , Expressão Gênica , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Humanos , Imunofenotipagem , Interferon gama/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Especificidade de Órgãos , Osteócitos/citologia , Osteócitos/metabolismo , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Antígenos Embrionários Estágio-Específicos/genética , Antígenos Embrionários Estágio-Específicos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
7.
J Cell Sci ; 127(Pt 3): 599-608, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24357723

RESUMO

Sharp-1 is a basic helix-loop-helix (bHLH) transcriptional repressor that is involved in a number of cellular processes. Our previous studies have demonstrated that Sharp-1 is a negative regulator of skeletal myogenesis and it blocks differentiation of muscle precursor cells by modulating the activity of MyoD. In order to understand its role in pre- and post-natal myogenesis, we assessed skeletal muscle development and freeze-injury-induced regeneration in Sharp-1-deficient mice. We show that embryonic skeletal muscle development is not impaired in the absence of Sharp-1; however, post-natally, the regenerative capacity is compromised. Although the initial phases of injury-induced regeneration proceed normally in Sharp-1(-/-) mice, during late stages, the mutant muscle exhibits necrotic fibers, calcium deposits and fibrosis. TGF-ß expression, as well as levels of phosphorylated Smad2 and Smad3, are sustained in the mutant tissue and treatment with decorin, which blocks TGF-ß signaling, improves the histopathology of Sharp-1(-/-) injured muscles. In vitro, Sharp-1 associates with Smad3, and its overexpression inhibits TGF-ß- and Smad3-mediated expression of extracellular matrix genes in myofibroblasts. These results demonstrate that Sharp-1 regulates muscle regenerative capacity, at least in part, by modulation of TGF-ß signaling.


Assuntos
Músculo Esquelético/metabolismo , Regeneração/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Embrião de Mamíferos , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Músculo Esquelético/crescimento & desenvolvimento , Miofibroblastos/metabolismo , Transdução de Sinais , Proteína Smad3 , Fatores de Transcrição/biossíntese , Fator de Crescimento Transformador beta/biossíntese
8.
J Biol Chem ; 288(24): 17654-62, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23637228

RESUMO

Sumoylation is an important post-translational modification that alters the activity of many transcription factors. However, the mechanisms that link sumoylation to alterations in chromatin structure, which culminate in tissue specific gene expression, are not fully understood. In this study, we demonstrate that SUMO modification of the transcription factor Sharp-1 is required for its full transcriptional repression activity and function as an inhibitor of skeletal muscle differentiation. Sharp-1 is modified by sumoylation at two conserved lysine residues 240 and 255. Mutation of these SUMO acceptor sites in Sharp-1 does not impact its subcellular localization but attenuates its ability to act as a transcriptional repressor and inhibit myogenic differentiation. Consistently, co-expression of the SUMO protease SENP1 with wild type Sharp-1 abrogates Sharp-1-dependent inhibition of myogenesis. Interestingly, sumoylation acts as a signal for recruitment of the co-repressor G9a. Thus, enrichment of G9a, and histone H3 lysine 9 dimethylation (H3K9me2), a signature of G9a activity, is dramatically reduced at muscle promoters in cells expressing sumoylation-defective Sharp-1. Our findings demonstrate how sumoylation of Sharp-1 exerts an impact on chromatin structure and transcriptional repression of muscle gene expression through recruitment of G9a.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Diferenciação Celular , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Sumoilação , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Sítios de Ligação , Células COS , Linhagem Celular , Chlorocebus aethiops , Sequência Conservada , Cisteína Endopeptidases , Endopeptidases/metabolismo , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Desenvolvimento Muscular , Proteína MyoD/metabolismo , Ligação Proteica , Transporte Proteico , Transcrição Gênica
9.
Epigenetics ; 8(1): 16-22, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23257913

RESUMO

Lysine methylation of histone and non-histone substrates by the methyltransferase G9a is mostly associated with transcriptional repression. Recent studies, however, have highlighted its role as an activator of gene expression through mechanisms that are independent of its methyltransferase activity. Here we review the growing repertoire of molecular mechanisms and substrates through which G9a regulates gene expression. We also discuss emerging evidence for its wide-ranging functions in development, pluripotency, cellular differentiation and cell cycle regulation that underscore the complexity of its functions. The deregulated expression of G9a in cancers and other human pathologies suggests that it may be a viable therapeutic target in various diseases.


Assuntos
Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Senescência Celular , Histona-Lisina N-Metiltransferase/química , Humanos , Neoplasias/enzimologia , Neoplasias/patologia , Proteínas Repressoras/metabolismo , Transativadores/metabolismo
10.
Mol Biol Cell ; 23(24): 4778-85, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23087213

RESUMO

Sharp-1, a basic helix-loop-helix transcription factor, is a potent repressor of skeletal muscle differentiation and is dysregulated in muscle pathologies. However, the mechanisms by which it inhibits myogenesis are not fully understood. Here we show that G9a, a lysine methyltransferase, is involved in Sharp-1-mediated inhibition of muscle differentiation. We demonstrate that G9a directly interacts with Sharp-1 and enhances its ability to transcriptionally repress the myogenin promoter. Concomitant with a differentiation block, G9a-dependent histone H3 lysine 9 dimethylation (H3K9me2) and MyoD methylation are apparent upon Sharp-1 overexpression in muscle cells. RNA interference-mediated reduction of G9a or pharmacological inhibition of its activity erases these repressive marks and rescues the differentiation defect imposed by Sharp-1. Our findings provide new insights into Sharp-1-dependent regulation of myogenesis and identify epigenetic mechanisms that could be targeted in myopathies characterized by elevated Sharp-1 levels.


Assuntos
Diferenciação Celular , Histona-Lisina N-Metiltransferase/metabolismo , Músculo Esquelético/metabolismo , Fatores de Transcrição/metabolismo , Animais , Western Blotting , Linhagem Celular , Regulação da Expressão Gênica , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Humanos , Luciferases/genética , Luciferases/metabolismo , Lisina/metabolismo , Metilação , Camundongos , Microscopia de Fluorescência , Músculo Esquelético/citologia , Mutação , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Miogenina/genética , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Fatores de Transcrição/genética
11.
PLoS One ; 5(2): e9016, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20126406

RESUMO

BACKGROUND: Wharton's jelly derived stem cells (WJMSCs) are gaining attention as a possible clinical alternative to bone marrow derived mesenchymal stem cells (BMMSCs) owing to better accessibility, higher expansion potential and low immunogenicity. Usage of allogenic mesenchymal stem cells (MSC) could be permissible in vivo only if they retain their immune properties in an inflammatory setting. Thus the focus of this study is to understand and compare the immune properties of BMMSCs and WJMSCs primed with key pro-inflammatory cytokines, Interferon-gamma (IFNgamma) and Tumor Necrosis Factor-alpha (TNFalpha). METHODOLOGY/PRINCIPAL FINDINGS: Initially the effect of priming on MSC mediated suppression of alloantigen and mitogen induced lymphoproliferation was evaluated in vitro. Treatment with IFNgamma or TNFalpha, did not ablate the immune-suppression caused by both the MSCs. Extent of immune-suppression was more with WJMSCs than BMMSCs in both the cases. Surprisingly, priming BMMSCs enhanced suppression of mitogen driven lymphoproliferation only; whereas IFNgamma primed WJMSCs were better suppressors of MLRs. Further, kinetic analysis of cytokine profiles in co-cultures of primed/unprimed MSCs and Phytohematoagglutinin (PHA) activated lymphocytes was evaluated. Results indicated a decrease in levels of pro-inflammatory cytokines. Interestingly, a change in kinetics and thresholds of Interleukin-2 (IL-2) secretion was observed only with BMMSCs. Analysis of activation markers on PHA-stimulated lymphocytes indicated different expression patterns in co-cultures of primed/unprimed WJMSCs and BMMSCs. Strikingly, co-culture with WJMSCs resulted in an early activation of a negative co-stimulatory molecule, CTLA4, which was not evident with BMMSCs. A screen for immune suppressive factors in primed/unprimed WJMSCs and BMMSCs indicated inherent differences in IFNgamma inducible Indoleamine 2, 3-dioxygenase (IDO) activity, Hepatocyte growth factor (HGF) and Prostaglandin E-2 (PGE2) levels which could possibly influence the mechanism of immune-modulation. CONCLUSION/SIGNIFICANCE: This study demonstrates that inflammation affects the immune properties of MSCs distinctly. Importantly different tissue derived MSCs could utilize unique mechanisms of immune-modulation.


Assuntos
Interferon gama/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Técnicas de Cocultura , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Teste de Cultura Mista de Linfócitos , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Células Estromais/imunologia , Células Estromais/metabolismo , Cordão Umbilical/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA