Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Stem Cells ; 36(4): 514-526, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29282804

RESUMO

The Nkx2-5 gene codes for a transcription factor that plays a critical role in heart development. Heterozygous mutations in NKX2-5 in both human and mice result in congenital heart defects (CHDs). However, the molecular mechanisms by which these mutations cause the disease are still unknown. Recently, we have generated the heterozygous mouse model of the human CHDs associated mutation NKX2-5 R142C (Nkx2-5R141C/+ mouse ortholog of human NKX2-5 R142C variant) that developed septal and conduction defects. This study generated a heterozygous Nkx2-5 R141C mouse embryonic stem cell line (Nkx2-5R141C/+ mESCs) to model CHDs in vitro. We observed that Nkx2-5R141C/+ mESCs display an alteration in the expression of genes that are essential for normal heart development. Furthermore, the reduced cardiomyogenesis is paralleled by a reduction in nuclear import of Nkx2-5 protein. Examination of the Nkx2-5R141C/+ embryos at E8.5 revealed a transient loss of cardiomyogenesis, which is consistent with the phenotype observed in vitro. Moreover, gene expression profiling of Nkx2-5R141C/+ cells at an early stage of cardiac differentiation revealed pronounced deregulation of several cardiac differentiation and function genes. Collectively, our data showed that heterozygosity for the R141C mutation results in disruption of the cellular distribution of Nkx2-5 protein, a transient reduction in cardiomyogenesis that may disrupt the early patterning of the heart, and this, in turn, affects the intricate orchestration of signaling pathways leading to downregulation of Bone morphogenetic protein (BMP) and Notch signaling. Therefore, we have developed mESCs model of a human CHD, providing an in vitro system to examine early stages of heart development, which are otherwise difficult to study in vivo. Stem Cells 2018;36:514-526.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Cardiopatias Congênitas/metabolismo , Proteína Homeobox Nkx-2.5/metabolismo , Modelos Cardiovasculares , Células-Tronco Embrionárias Murinas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Substituição de Aminoácidos , Animais , Proteínas Morfogenéticas Ósseas/genética , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Proteína Homeobox Nkx-2.5/genética , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/patologia , Mutação de Sentido Incorreto , Receptores Notch/genética
2.
J Mol Cell Cardiol ; 105: 89-98, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28302382

RESUMO

The Nkx2.5 gene encodes a transcription factor that plays a critical role in heart development. In humans, heterozygous mutations in NKX2.5 result in congenital heart defects (CHDs). However, the molecular mechanisms by which these mutations cause the disease remain unknown. NKX2.5-R142C is a mutation that was reported to be associated with atrial septal defect (ASD) and atrioventricular (AV) block in 13-patients from one family. The R142C mutation is located within both the DNA-binding domain and the nuclear localization sequence of NKX2.5 protein. The pathogenesis of CHDs in humans with R142C point mutation is not well understood. To examine the functional deficit associated with this mutation in vivo, we generated and characterized a knock-in mouse that harbours the human mutation R142C. Systematic structural and functional examination of the embryonic, newborn, and adult mice revealed that the homozygous embryos Nkx2.5R141C/R141C are developmentally arrested around E10.5 with delayed heart morphogenesis and downregulation of Nkx2.5 target genes, Anf, Mlc2v, Actc1 and Cx40. Histological examination of Nkx2.5R141C/+ newborn hearts showed that 36% displayed ASD, with at least 80% 0f adult heterozygotes displaying a septal defect. Moreover, heterozygous Nkx2.5R141C/+ newborn mice have downregulation of ion channel genes with 11/12 adult mice manifesting a prolonged PR interval that is indicative of 1st degree AV block. Collectively, the present study demonstrates that mice with the R141C point mutation in the Nkx2.5 allele phenocopies humans with the NKX2.5 R142C point mutation.


Assuntos
Estudos de Associação Genética , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/fisiopatologia , Proteína Homeobox Nkx-2.5/genética , Mutação , Animais , Modelos Animais de Doenças , Ecocardiografia , Feminino , Expressão Gênica , Marcação de Genes , Vetores Genéticos/genética , Genótipo , Cardiopatias Congênitas/diagnóstico , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo
3.
Methods ; 101: 73-84, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26404920

RESUMO

Human pluripotent stem cells provide a developmental model to study early embryonic and tissue development, tease apart human disease processes, perform drug screens to identify potential molecular effectors of in situ regeneration, and provide a source for cell and tissue based transplantation. Highly efficient differentiation protocols have been established for many cell types and tissues; however, until very recently robust differentiation into skeletal muscle cells had not been possible unless driven by transgenic expression of master regulators of myogenesis. Nevertheless, several breakthrough protocols have been published in the past two years that efficiently generate cells of the skeletal muscle lineage from pluripotent stem cells. Here, we present an updated version of our recently described 50-day protocol in detail, whereby chemically defined media are used to drive and support muscle lineage development from initial CHIR99021-induced mesoderm through to PAX7-expressing skeletal muscle progenitors and mature skeletal myocytes. Furthermore, we report an optional method to passage and expand differentiating skeletal muscle progenitors approximately 3-fold every 2weeks using Collagenase IV and continued FGF2 supplementation. Both protocols have been optimized using a variety of human pluripotent stem cell lines including patient-derived induced pluripotent stem cells. Taken together, our differentiation and expansion protocols provide sufficient quantities of skeletal muscle progenitors and myocytes that could be used for a variety of studies.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Humanas/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Técnicas de Cultura de Células , Células Cultivadas , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Microscopia de Fluorescência , Células Musculares/fisiologia , Mioblastos/fisiologia
4.
BMC Dev Biol ; 16(1): 27, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27484899

RESUMO

BACKGROUND: The Hedgehog (HH) signalling pathway regulates cardiomyogenesis in vivo and in differentiating P19 embryonal carcinoma (EC) cells, a mouse embryonic stem (mES) cell model. To further assess the transcriptional role of HH signalling during cardiomyogenesis in stem cells, we studied the effects of overexpressing GLI2, a primary transducer of the HH signalling pathway, in mES cells. RESULTS: Stable GLI2 overexpression resulted in an enhancement of cardiac progenitor-enriched genes, Mef2c, Nkx2-5, and Tbx5 during mES cell differentiation. In contrast, pharmacological blockade of the HH pathway in mES cells resulted in lower expression of these genes. Mass spectrometric analysis identified the chromatin remodelling factor BRG1 as a protein which co-immunoprecipitates with GLI2 in differentiating mES cells. We then determined that BRG1 is recruited to a GLI2-specific Mef2c gene element in a HH signalling-dependent manner during cardiomyogenesis in P19 EC cells, a mES cell model. CONCLUSIONS: Thus, we propose a mechanism where HH/GLI2 regulates the expression of Mef2c by recruiting BRG1 to the Mef2c gene, most probably via chromatin remodelling, to ultimately regulate in vitro cardiomyogenesis.


Assuntos
DNA Helicases/metabolismo , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Miócitos Cardíacos/citologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Montagem e Desmontagem da Cromatina , DNA Helicases/genética , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Proteínas Hedgehog/genética , Técnicas In Vitro , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Espectrometria de Massas , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Transdução de Sinais , Fatores de Transcrição/genética , Proteína Gli2 com Dedos de Zinco
5.
EMBO J ; 30(12): 2477-89, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21556048

RESUMO

The MEF2 factors regulate transcription during cardiac and skeletal myogenesis. MEF2 factors establish skeletal muscle commitment by amplifying and synergizing with MyoD. While phosphorylation is known to regulate MEF2 function, lineage-specific regulation is unknown. Here, we show that phosphorylation of MEF2C on T(80) by skeletal myosin light chain kinase (skMLCK) enhances skeletal and not cardiac myogenesis. A phosphorylation-deficient MEF2C mutant (MEFT80A) enhanced cardiac, but not skeletal myogenesis in P19 stem cells. Further, MEFT80A was deficient in recruitment of p300 to skeletal but not cardiac muscle promoters. In gain-of-function studies, skMLCK upregulated myogenic regulatory factor (MRF) expression, leading to enhanced skeletal myogenesis in P19 cells and more efficient myogenic conversion. In loss-of-function studies, MLCK was essential for efficient MRF expression and subsequent myogenesis in embryonic stem (ES) and P19 cells as well as for proper activation of quiescent satellite cells. Thus, skMLCK regulates MRF expression by controlling the MEF2C-dependent recruitment of histone acetyltransferases to skeletal muscle promoters. This work identifies the first kinase that regulates MyoD and Myf5 expression in ES or satellite cells.


Assuntos
Proteínas de Domínio MADS/metabolismo , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/enzimologia , Fatores de Regulação Miogênica/metabolismo , Quinase de Cadeia Leve de Miosina/fisiologia , Sequência de Aminoácidos , Animais , Carcinoma Embrionário/enzimologia , Carcinoma Embrionário/patologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Proteínas de Domínio MADS/fisiologia , Fatores de Transcrição MEF2 , Camundongos , Dados de Sequência Molecular , Fatores de Regulação Miogênica/fisiologia , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Especificidade de Órgãos , Fosforilação , Treonina/metabolismo
6.
Circ Res ; 112(2): 257-66, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23168335

RESUMO

RATIONALE: Nkx2.5 is a transcription factor that regulates cardiomyogenesis in vivo and in embryonic stem cells. It is also a common target in congenital heart disease. Although Nkx2.5 has been implicated in the regulation of many cellular processes that ultimately contribute to cardiomyogenesis and morphogenesis of the mature heart, relatively little is known about how it is regulated at a functional level. OBJECTIVE: We have undertaken a proteomic screen to identify novel binding partners of Nkx2.5 during cardiomyogenic differentiation in an effort to better understand the regulation of its transcriptional activity. METHODS AND RESULTS: Purification of Nkx2.5 from differentiating cells identified the myosin phosphatase subunits protein phosphatase 1ß and myosin phosphatase targeting subunit 1 (Mypt1) as novel binding partners. The interaction with protein phosphatase 1 ß/Mypt1 resulted in exclusion of Nkx2.5 from the nucleus and, consequently, inhibition of its transcriptional activity. Exclusion of Nkx2.5 was inhibited by treatment with leptomycin B and was dependent on an Mypt1 nuclear export signal. Furthermore, in transient transfection experiments, Nkx2.5 colocalized outside the nucleus with phosphorylated Mypt1 in a manner dependent on Wnt signaling and Rho-associated protein kinase. Treatment of differentiating mouse embryonic stem cells with Wnt3a resulted in enhanced phosphorylation of endogenous Mypt1, increased nuclear exclusion of endogenous Nkx2.5, and a failure to undergo terminal cardiomyogenesis. Finally, knockdown of Mypt1 resulted in rescue of Wnt3a-mediated inhibition of cardiomyogenesis, indicating that Mypt1 is required for this process. CONCLUSIONS: We have identified a novel interaction between Nkx2.5 and myosin phosphatase. Promoting this interaction represents a novel mechanism whereby Wnt3a regulates Nkx2.5 and inhibits cardiomyogenesis.


Assuntos
Inibidores do Crescimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Miócitos Cardíacos/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt3A/fisiologia , Quinases Associadas a rho/fisiologia , Animais , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Células HEK293 , Proteína Homeobox Nkx-2.5 , Humanos , Camundongos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Proteína Fosfatase 1/metabolismo , Frações Subcelulares/enzimologia , Frações Subcelulares/metabolismo
7.
J Biol Chem ; 288(6): 4389-404, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23266826

RESUMO

The inhibition of MyoD expression is important for obtaining muscle progenitors that can replenish the satellite cell niche during muscle repair. Progenitors could be derived from either embryonic stem cells or satellite cells. Hedgehog (Hh) signaling is important for MyoD expression during embryogenesis and adult muscle regeneration. To date, the mechanistic understanding of MyoD regulation by Hh signaling is unclear. Here, we demonstrate that the Hh effector, Gli2, regulates MyoD expression and associates with MyoD gene elements. Gain- and loss-of-function experiments in pluripotent P19 cells show that Gli2 activity is sufficient and required for efficient MyoD expression during skeletal myogenesis. Inhibition of Hh signaling reduces MyoD expression during satellite cell activation in vitro. In addition to regulating MyoD expression, Hh signaling regulates MyoD transcriptional activity, and MyoD activates Hh signaling in myogenic conversion assays. Finally, Gli2, MyoD, and MEF2C form a protein complex, which enhances MyoD activity on skeletal muscle-related promoters. We therefore link Hh signaling to the function and expression of MyoD protein during myogenesis in stem cells.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas Hedgehog/metabolismo , Proteína MyoD/biossíntese , Células-Tronco Pluripotentes/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Proteínas Hedgehog/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição MEF2 , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Desenvolvimento Muscular/fisiologia , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Células-Tronco Pluripotentes/citologia , Células Satélites de Músculo Esquelético/citologia , Proteína Gli2 com Dedos de Zinco
8.
Nucleic Acids Res ; 40(8): 3329-47, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22199256

RESUMO

The transcription factors Gli2 (glioma-associated factor 2), which is a transactivator of Sonic Hedgehog (Shh) signalling, and myocyte enhancer factor 2C (MEF2C) play important roles in the development of embryonic heart muscle and enhance cardiomyogenesis in stem cells. Although the physiological importance of Shh signalling and MEF2 factors in heart development is well known, the mechanistic understanding of their roles is unclear. Here, we demonstrate that Gli2 and MEF2C activated each other's expression while enhancing cardiomyogenesis in differentiating P19 EC cells. Furthermore, dominant-negative mutant proteins of either Gli2 or MEF2C repressed each other's expression, while impairing cardiomyogenesis in P19 EC cells. In addition, chromatin immunoprecipitation (ChIP) revealed association of Gli2 to the Mef2c gene, and of MEF2C to the Gli2 gene in differentiating P19 cells. Finally, co-immunoprecipitation studies showed that Gli2 and MEF2C proteins formed a complex, capable of synergizing on cardiomyogenesis-related promoters containing both Gli- and MEF2-binding elements. We propose a model whereby Gli2 and MEF2C bind each other's regulatory elements, activate each other's expression and form a protein complex that synergistically activates transcription, enhancing cardiac muscle development. This model links Shh signalling to MEF2C function during cardiomyogenesis and offers mechanistic insight into their in vivo functions.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Desenvolvimento Muscular/genética , Miócitos Cardíacos/metabolismo , Fatores de Regulação Miogênica/metabolismo , Ativação Transcricional , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Coração/embriologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição MEF2 , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Fatores de Regulação Miogênica/genética , Regiões Promotoras Genéticas , Proteína Gli2 com Dedos de Zinco
9.
J Mol Cell Cardiol ; 60: 164-71, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23598283

RESUMO

Since a previous study (Goldman-Johnson et al., 2008 [4]) has shown that androgens can stimulate increased differentiation of mouse embryonic stem (mES) cells into cardiomyocytes using a genomic pathway, the aim of our study is to elucidate the molecular mechanisms regulating testosterone-enhanced cardiomyogenesis. Testosterone upregulated cardiomyogenic transcription factors, including GATA4, MEF2C, and Nkx2.5, muscle structural proteins, and the pacemaker ion channel HCN4 in a dose-dependent manner, in mES cells and P19 embryonal carcinoma cells. Knock-down of the androgen receptor (AR) or treatment with anti-androgenic compounds inhibited cardiomyogenesis, supporting the requirement of the genomic pathway. Chromatin immunoprecipitation (ChIP) studies showed that testosterone enhanced recruitment of AR to the regulatory regions of MEF2C and HCN4 genes, which was associated with increased histone acetylation. In summary, testosterone upregulated cardiomyogenic transcription factor and HCN4 expression in stem cells. Further, testosterone induced cardiomyogenesis, at least in part, by recruiting the AR receptor to the regulatory regions of the MEF2C and HCN4 genes. These results provide a detailed molecular analysis of the function of testosterone in stem cells and may offer molecular insight into the role of steroids in the heart.


Assuntos
Androgênios/farmacologia , Células-Tronco Embrionárias/metabolismo , Coração/embriologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/biossíntese , Organogênese/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Testosterona/farmacologia , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Organogênese/fisiologia , Receptores Androgênicos/genética , Elementos de Resposta/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
10.
Cell Mol Life Sci ; 69(2): 215-21, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21947442

RESUMO

Aberrant or elevated levels of reactive oxygen species (ROS) can mediate deleterious cellular effects, including neuronal toxicity and degeneration observed in the etiology of a number of pathological conditions, including Alzheimer's and Parkinson's diseases. Nevertheless, ROS can be generated in a controlled manner and can regulate redox sensitive transcription factors such as NFκB, AP-1 and NFAT. Moreover, ROS can modulate the redox state of tyrosine phosphorylated proteins, thereby having an impact on many transcriptional networks and signaling cascades important for neurogenesis. A large body of literature links the controlled generation of ROS at low-to-moderate levels with the stimulation of differentiation in certain developmental programs such as neurogenesis. In this regard, ROS are involved in governing the acquisition of the neural fate-from neural induction to the elaboration of axons. Here, we summarize and discuss the growing body of literature that describe a role for ROS signaling in neuronal development.


Assuntos
Sistema Nervoso/metabolismo , Neurogênese , Espécies Reativas de Oxigênio/metabolismo , Animais , Feminino , Humanos , Masculino , Camundongos , NADPH Oxidases/metabolismo , Sistema Nervoso/crescimento & desenvolvimento , Neurônios/enzimologia , Transdução de Sinais
11.
J Biol Chem ; 286(4): 2517-25, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21078671

RESUMO

Gain- and loss-of-function experiments have illustrated that the family of myogenic regulatory factors is necessary and sufficient for the formation of skeletal muscle. Furthermore, MyoD required cellular aggregation to induce myogenesis in P19 embryonal carcinoma stem cells. To determine the mechanism by which stem cells can be directed into skeletal muscle, a time course of P19 cell differentiation was examined in the presence and absence of exogenous MyoD. By quantitative PCR, the first MyoD up-regulated transcripts were the premyogenic mesoderm factors Meox1, Pax7, Six1, and Eya2 on day 4 of differentiation. Subsequently, the myoblast markers myogenin, MEF2C, and Myf5 were up-regulated, leading to skeletal myogenesis. These results were corroborated by Western blot analysis, showing up-regulation of Pax3, Six1, and MEF2C proteins, prior to myogenin protein expression. To determine at what stage a dominant-negative MyoD/EnR mutant could inhibit myogenesis, stable cell lines were created and examined. Interestingly, the premyogenic mesoderm factors, Meox1, Pax3/7, Six1, Eya2, and Foxc1, were down-regulated, and as expected, skeletal myogenesis was abolished. Finally, to identify direct targets of MyoD in this system, chromatin immunoprecipitation experiments were performed. MyoD was observed associated with regulatory regions of Meox1, Pax3/7, Six1, Eya2, and myogenin genes. Taken together, MyoD directs stem cells into the skeletal muscle lineage by binding and activating the expression of premyogenic mesoderm genes, prior to activating myoblast genes.


Assuntos
Antígenos de Diferenciação/biossíntese , Diferenciação Celular/fisiologia , Proteínas de Homeodomínio/biossíntese , Desenvolvimento Muscular/fisiologia , Proteína MyoD/metabolismo , Mioblastos Esqueléticos/metabolismo , Células-Tronco/metabolismo , Regulação para Cima/fisiologia , Animais , Antígenos de Diferenciação/genética , Linhagem Celular Tumoral , Regulação para Baixo/fisiologia , Proteínas de Homeodomínio/genética , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Mutação , Proteína MyoD/genética , Mioblastos Esqueléticos/citologia , Células-Tronco/citologia
12.
FASEB J ; 25(3): 960-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21135039

RESUMO

The SET1 family of methyltransferases carries out the bulk of histone H3 Lys-4 methylation in vivo. One of the common features of this family is the regulation of their methyltransferase activity by a tripartite complex composed of WDR5, RbBP5, and Ash2L. To selectively probe the role of the SET1 family of methyltransferases, we have developed a library of histone H3 peptide mimetics and report herein the characterization of an Nα acetylated form of histone H3 peptide (NαH3). Binding and inhibition studies reveal that the addition of an acetyl moiety to the N terminus of histone H3 significantly enhances its binding to WDR5 and prevents the stimulation of MLL1 methyltransferase activity by the WDR5-RbBP5-Ash2L complex. The crystal structure of NαH3 in complex with WDR5 reveals that a high-affinity hydrophobic pocket accommodates the binding of the acetyl moiety. These results provide the structural basis to control WDR5-RbBP5-Ash2L-MLL1 activity and a tool to manipulate stem cell differentiation programs.


Assuntos
Metilação de DNA/fisiologia , Epigenômica , Histonas/metabolismo , Proteína de Leucina Linfoide-Mieloide , Células-Tronco/enzimologia , Acetilação , Diferenciação Celular/fisiologia , Células Cultivadas , Cristalografia , Proteínas de Ligação a DNA/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/química , Histonas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Químicos , Mimetismo Molecular , Proteína de Leucina Linfoide-Mieloide/química , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas Nucleares/metabolismo , Nucleossomos/fisiologia , Biblioteca de Peptídeos , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Células-Tronco/citologia , Fatores de Transcrição/metabolismo
13.
J Biol Chem ; 285(23): 17974-85, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20233719

RESUMO

In this study, we describe a role for the mammalian Numb-interacting protein 1 (Nip1) in regulation of neuronal differentiation in stem cells. The expression of Nip1 was detected in the developing mouse brain, embryonic stem cells, primary neuronal stem cells, and retinoic acid-treated P19 embryonal carcinoma cells. The highest expression of Nip1 was observed in undifferentiated neuronal stem cells and was associated with Duox1-mediated reactive oxygen species ROS production. Ectopic nip1 expression in P19 embryonal carcinoma cells induced neuronal differentiation, and this phenotype was also linked to elevated ROS production. The neuronal differentiation in nip1-overexpressing P19 cells was achieved in a retinoic acid-independent manner and was corroborated by an increase in the expression of the neuronal basic helix-loop-helix transcription factors and neural-lineage cell markers. Furthermore, depletion of nip1 by short hairpin RNA led to a decrease in the expression of neuronal basic helix-loop-helix transcription factors and ROS. However, inhibition of ROS production in nip1-overexpressing P19 cells restricted but did not extinguish neuronal differentiation. Microarray and mass spectrometry analysis identified intermediate filaments as the principal cytoskeletal elements affected by up-regulation of nip1. We show here the first evidence for a functional interaction between Nip1 and a component of the nuclear lamina, lamin A/C. associated with a neuronal-specific phenotype. Taken together, our data reveal an important role for Nip1 in the guidance of neuronal differentiation through ROS generation and modulation of intermediate filaments and implicate Nip1 as a novel intrinsic regulator of neuronal cell fate.


Assuntos
NADPH Oxidases/metabolismo , Neurônios/metabolismo , Células-Tronco/citologia , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Linhagem da Célula , Citoesqueleto/metabolismo , Oxidases Duais , Lamina Tipo A/química , Camundongos , Complexo Proteico Nuclear de Ligação ao Cap/metabolismo , Fenótipo , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio , Células-Tronco/metabolismo
14.
Dev Cell ; 11(4): 535-46, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17011492

RESUMO

The transcriptional mechanisms underlying lineage specification and differentiation of embryonic stem (ES) cells remain elusive. Oct-3/4 (POU5f1) is one of the earliest transcription factors expressed in the embryo. Both the pluripotency and the fate of ES cells depend upon a tight control of Oct-3/4 expression. We report that transgene- or TGFbeta-induced increase in Oct-3/4 mRNA and protein levels in undifferentiated ES cells and at early stages of differentiation triggers expression of mesodermal and cardiac specific genes through Smad2/4. cDNA antisense- and siRNA-mediated inhibition of upregulation of Oct-3/4 in ES cells prevent their specification toward the mesoderm and their differentiation into cardiomyocytes. Similarly, Oct-3/4 siRNA injected in the inner cell mass of blastocysts impairs cardiogenesis in early embryos. Thus, quantitative Oct-3/4 expression is regulated by a morphogen, pointing to a pivotal and physiological function of the POU factor in mesodermal and cardiac commitments of ES cells and of the epiblast.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/crescimento & desenvolvimento , Miócitos Cardíacos/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Linhagem da Célula , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Microinjeções , Miócitos Cardíacos/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Técnicas de Cultura de Órgãos , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/metabolismo
15.
Differentiation ; 79(1): 31-40, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19782461

RESUMO

FOXC1 and FOXC2 are forkhead/winged-helix transcription factors expressed in paraxial mesoderm and somites. Emphasizing the importance of FOXC1/2 during embryonic development, double-knockout mice lacking the alleles for both Foxc1 and Foxc2 failed to form segmented somites and undergo myogenesis. The present study aims to determine upstream factors that regulate Foxc1/2 expression during the differentiation of P19 cells into skeletal muscle. Previous work had shown that dominant-negative forms of beta-catenin, Gli2, and Meox1 could inhibit distinct stages of skeletal myogenesis in P19 cells. In the presence of a dominant-negative beta-catenin fusion protein, Foxc1/2 transcripts were not upregulated and neither were markers of somitogenesis/myogenesis, including Meox1, Pax3 and MyoD. Conversely, inhibition of GSK3 by LiCl or overexpression of activated beta-catenin in aggregated P19 cells resulted in enhancement of Foxc1/2 expression, indicating that FOX transcription may be under the control of Wnt signaling. Supporting this hypothesis, beta-catenin bound to conserved regions upstream of Foxc1 during P19 cell differentiation and drove transcription from this region in a promoter assay. In addition, ectopic expression of a dominant-negative Meox1 or Gli2 resulted in decreased Foxc1/2 transcript levels, correlating with inhibition of skeletal myogenesis. Overexpression of Gli2 was also sufficient to upregulate Foxc1/2 transcript levels and induce skeletal myogenesis. In summary, Foxc1/2 expression is dependent on a complex interplay from various signaling inputs from the Wnt and Shh pathways during early stages of in vitro skeletal myogenesis.


Assuntos
Carcinoma Embrionário/metabolismo , Fatores de Transcrição Forkhead/genética , Músculo Esquelético/citologia , Transdução de Sinais/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/metabolismo , Animais , Northern Blotting , Carcinoma Embrionário/patologia , Diferenciação Celular , Imunoprecipitação da Cromatina , Imunofluorescência , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição , Células Tumorais Cultivadas , Proteína Gli2 com Dedos de Zinco , beta Catenina/genética
16.
Stem Cells ; 27(6): 1231-43, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489079

RESUMO

In this study, we have identified novel roles for Sox15 and Sox7 as regulators of muscle precursor cell fate in P19 cells. To examine the role of Sox15 and Sox7 during skeletal myogenesis, we isolated populations of P19 cells with either gene stably integrated into the genome, termed P19[Sox15] and P19[Sox7]. Both SOX proteins were sufficient to upregulate the expression of the muscle precursor markers Pax3/7, Meox1, and Foxc1 in aggregated cells. In contrast to the P19[Sox7] cell lines, which subsequently differentiated into skeletal muscle, myogenesis failed to progress past the precursor stage in P19[Sox15] cell lines, shown by the lack of MyoD and myosin heavy chain (MHC) expression. P19[Sox15] clones showed elevated and sustained levels of the inhibitory factors Msx1 and Id1, which may account for the lack of myogenic progression in these cells. Stable expression of a Sox15 dominant-negative protein resulted in the loss of Pax3/7 and Meox1 transcripts, as well as myogenic regulatory factor (MRF) and MHC expression. These results suggest that Sox15, or genes that are bound by Sox15, are necessary and sufficient for the acquisition of the muscle precursor cell fate. On the other hand, knockdown of endogenous Sox15 caused a decrease in Pax3 and Meox1, but not MRF expression, suggesting that other factors can compensate in the absence of Sox15. Taken together, these results show that both Sox7 and Sox15 are able to induce the early stages of myogenesis, but only Sox7 is sufficient to initiate the formation of fully differentiated skeletal myocytes.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Muscular/genética , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Fatores de Transcrição SOX/metabolismo , Fatores de Transcrição SOXF/metabolismo , Animais , Linhagem Celular , Imunofluorescência , Expressão Gênica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Imunoprecipitação , Camundongos , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Interferência de RNA , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOXF/genética , Células-Tronco/fisiologia , Transfecção
17.
Mol Cell Proteomics ; 7(3): 560-72, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18065756

RESUMO

Very little is known about SET- and MYND-containing protein 2 (SMYD2), a member of the SMYD protein family. However, the interest in better understanding the roles of SMYD2 has grown because of recent reports indicating that SMYD2 methylates p53 and histone H3. In this study, we present a combined proteomics and genomics study of SMYD2 designed to elucidate its molecular roles. We report the cytosolic and nuclear interactome of SMYD2 using a combination of immunoprecipitation coupled with high throughput MS, chromatin immunoprecipitation coupled with high throughput MS, and co-immunoprecipitation methods. In particular, we report that SMYD2 interacted with HSP90alpha independently of the SET and MYND domains, with EBP41L3 through the MYND domain, and with p53 through the SET domain. We demonstrated that the interaction of SMYD2 with HSP90alpha enhances SMYD2 histone methyltransferase activity and specificity for histone H3 at lysine 4 (H3K4) in vitro. Interestingly histone H3K36 methyltransferase activity was independent of its interaction with HSP90alpha similar to LSD1 dependence on the androgen receptor. We also showed that the SET domain is required for the methylation at H3K4. We demonstrated using a modified chromatin immunoprecipitation protocol that the SMYD2 gain of function leads to an increase in H3K4 methylation in vivo, whereas no observable levels of H3K36 were detected. We also report that the SMYD2 gain of function was correlated with the up-regulation of 37 and down-regulation of four genes, the majority of which are involved in the cell cycle, chromatin remodeling, and transcriptional regulation. TACC2 is one of the genes up-regulated as a result of SMYD2 gain of function. Up-regulation of TACC2 by SMYD2 occurred as a result of SMYD2 binding to the TACC2 promoter where it methylates H3K4. Furthermore the combination of the SMYD2 interactome with the gene expression data suggests that some of the genes regulated by SMYD2 are closely associated with SMYD2-interacting proteins.


Assuntos
Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Proteômica/métodos , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas de Transporte/genética , Linhagem Celular , DNA/metabolismo , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP90/metabolismo , Histonas/metabolismo , Humanos , Metilação , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Reprodutibilidade dos Testes , Especificidade por Substrato , Proteínas Supressoras de Tumor/genética
18.
BMC Biol ; 7: 67, 2009 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-19814781

RESUMO

BACKGROUND: Understanding stem cell differentiation is essential for the future design of cell therapies. While retinoic acid (RA) is the most potent small molecule enhancer of skeletal myogenesis in stem cells, the stage and mechanism of its function has not yet been elucidated. Further, the intersection of RA with other signalling pathways that stimulate or inhibit myogenesis (such as Wnt and BMP4, respectively) is unknown. Thus, the purpose of this study is to examine the molecular mechanisms by which RA enhances skeletal myogenesis and interacts with Wnt and BMP4 signalling during P19 or mouse embryonic stem (ES) cell differentiation. RESULTS: Treatment of P19 or mouse ES cells with low levels of RA led to an enhancement of skeletal myogenesis by upregulating the expression of the mesodermal marker, Wnt3a, the skeletal muscle progenitor factors Pax3 and Meox1, and the myogenic regulatory factors (MRFs) MyoD and myogenin. By chromatin immunoprecipitation, RA receptors (RARs) bound directly to regulatory regions in the Wnt3a, Pax3, and Meox1 genes and RA activated a beta-catenin-responsive promoter in aggregated P19 cells. In the presence of a dominant negative beta-catenin/engrailed repressor fusion protein, RA could not bypass the inhibition of skeletal myogenesis nor upregulate Meox1 or MyoD. Thus, RA functions both upstream and downstream of Wnt signalling. In contrast, it functions downstream of BMP4, as it abrogates BMP4 inhibition of myogenesis and Meox1, Pax3, and MyoD expression. Furthermore, RA downregulated BMP4 expression and upregulated the BMP4 inhibitor, Tob1. Finally, RA inhibited cardiomyogenesis but not in the presence of BMP4. CONCLUSION: RA can enhance skeletal myogenesis in stem cells at the muscle specification/progenitor stage by activating RARs bound directly to mesoderm and skeletal muscle progenitor genes, activating beta-catenin function and inhibiting bone morphogenetic protein (BMP) signalling. Thus, a signalling pathway can function at multiple levels to positively regulate a developmental program and can function by abrogating inhibitory pathways. Finally, since RA enhances skeletal muscle progenitor formation, it will be a valuable tool for designing future stem cell therapies.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Músculo Esquelético/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Tretinoína/farmacologia , beta Catenina/metabolismo , Animais , Padronização Corporal/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Dominantes , Humanos , Camundongos , Modelos Biológicos , Desenvolvimento Muscular/efeitos dos fármacos , Proteína MyoD/genética , Proteína MyoD/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Organogênese/efeitos dos fármacos , Paxilina/genética , Paxilina/metabolismo
19.
Curr Protoc Stem Cell Biol ; 48(1): e75, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30548532

RESUMO

We have established an in vitro model of the human congenital heart defect (CHD)-associated mutation NKX2.5 R141C. We describe the use of the hanging drop method to differentiate Nkx2.5R141C/+ murine embryonic stem cells (mESCs) along with Nkx2.5+/+ control cells. This method allows us to recapitulate the early stages of embryonic heart development in tissue culture. We also use qRT-PCR and immunofluorescence to examine samples at different time points during differentiation to validate our data. The in vivo model is a mouse line with a knock-in of the same mutation. We describe the isolation of RNA from embryonic day 8.5 (E8.5) embryos and E9.5 hearts of wild-type and mutant mice. We found that the in vitro model shows reduced cardiomyogenesis, similar to Nkx2.5R141C/+ embryos at E8.5, indicating a transient loss of cardiomyogenesis at this time point. These results suggest that our in vitro model can be used to study very early changes in heart development that cause CHD. © 2018 by John Wiley & Sons, Inc.


Assuntos
Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Cardiopatias Congênitas/genética , Proteína Homeobox Nkx-2.5/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Coração/crescimento & desenvolvimento , Humanos , Camundongos , Miócitos Cardíacos/citologia , RNA/isolamento & purificação
20.
PLoS One ; 14(9): e0222946, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31560727

RESUMO

Human embryonic stem cell (hESC)-derived skeletal muscle progenitors (SMP)-defined as PAX7-expressing cells with myogenic potential-can provide an abundant source of donor material for muscle stem cell therapy. As in vitro myogenesis is decoupled from in vivo timing and 3D-embryo structure, it is important to characterize what stage or type of muscle is modeled in culture. Here, gene expression profiling is analyzed in hESCs over a 50 day skeletal myogenesis protocol and compared to datasets of other hESC-derived skeletal muscle and adult murine satellite cells. Furthermore, day 2 cultures differentiated with high or lower concentrations of CHIR99021, a GSK3A/GSK3B inhibitor, were contrasted. Expression profiling of the 50 day time course identified successively expressed gene subsets involved in mesoderm/paraxial mesoderm induction, somitogenesis, and skeletal muscle commitment/formation which could be regulated by a putative cascade of transcription factors. Initiating differentiation with higher CHIR99021 concentrations significantly increased expression of MSGN1 and TGFB-superfamily genes, notably NODAL, resulting in enhanced paraxial mesoderm and reduced ectoderm/neuronal gene expression. Comparison to adult satellite cells revealed that genes expressed in 50-day cultures correlated better with those expressed by quiescent or early activated satellite cells, which have the greatest therapeutic potential. Day 50 cultures were similar to other hESC-derived skeletal muscle and both expressed known and novel SMP surface proteins. Overall, a putative cascade of transcription factors has been identified which regulates four stages of myogenesis. Subsets of these factors were upregulated by high CHIR99021 or their binding sites were significantly over-represented during SMP activation, ranging from quiescent to late-activated stages. This analysis serves as a resource to further study the progression of in vitro skeletal myogenesis and could be mined to identify novel markers of pluripotent-derived SMPs or regulatory transcription/growth factors. Finally, 50-day hESC-derived SMPs appear similar to quiescent/early activated satellite cells, suggesting they possess therapeutic potential.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Humanas/metabolismo , Desenvolvimento Muscular/genética , Músculo Esquelético/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Perfilação da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Músculo Esquelético/citologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Células Satélites de Músculo Esquelético/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA