Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Development ; 145(21)2018 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-30266829

RESUMO

The transcription factor Nfix belongs to the nuclear factor one family and has an essential role in prenatal skeletal muscle development, where it is a master regulator of the transition from embryonic to foetal myogenesis. Recently, Nfix was shown to be involved in adult muscle regeneration and in muscular dystrophies. Here, we have investigated the signalling that regulates Nfix expression, and show that JunB, a member of the AP-1 family, is an activator of Nfix, which then leads to foetal myogenesis. Moreover, we demonstrate that their expression is regulated through the RhoA/ROCK axis, which maintains embryonic myogenesis. Specifically, RhoA and ROCK repress ERK kinase activity, which promotes JunB and Nfix expression. Notably, the role of ERK in the activation of Nfix is conserved postnatally in satellite cells, which represent the canonical myogenic stem cells of adult muscle. As lack of Nfix in muscular dystrophies rescues the dystrophic phenotype, the identification of this pathway provides an opportunity to pharmacologically target Nfix in muscular dystrophies.


Assuntos
Sistema de Sinalização das MAP Quinases , Desenvolvimento Muscular , Mioblastos/metabolismo , Fatores de Transcrição NFI/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Animais Recém-Nascidos , Embrião de Mamíferos/metabolismo , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Masculino , Camundongos , Fatores de Transcrição NFI/genética , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Quinases Associadas a rho/metabolismo
2.
Nat Commun ; 8(1): 1055, 2017 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-29057908

RESUMO

Muscular dystrophies are severe disorders due to mutations in structural genes, and are characterized by skeletal muscle wasting, compromised patient mobility, and respiratory functions. Although previous works suggested enhancing regeneration and muscle mass as therapeutic strategies, these led to no long-term benefits in humans. Mice lacking the transcription factor Nfix have delayed regeneration and a shift toward an oxidative fiber type. Here, we show that ablating or silencing the transcription factor Nfix ameliorates pathology in several forms of muscular dystrophy. Silencing Nfix in postnatal dystrophic mice, when the first signs of the disease already occurred, rescues the pathology and, conversely, Nfix overexpression in dystrophic muscles increases regeneration and markedly exacerbates the pathology. We therefore offer a proof of principle for a novel therapeutic approach for muscular dystrophies based on delaying muscle regeneration.


Assuntos
Músculos/fisiologia , Distrofias Musculares/genética , Fatores de Transcrição NFI/fisiologia , Regeneração , Animais , Feminino , Inativação Gênica , Masculino , Camundongos , Músculos/patologia , Distrofias Musculares/patologia , Sarcoglicanas/genética
3.
Cell Rep ; 17(9): 2354-2366, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27880909

RESUMO

Sox6 belongs to the Sox gene family and plays a pivotal role in fiber type differentiation, suppressing transcription of slow-fiber-specific genes during fetal development. Here, we show that Sox6 plays opposite roles in MyHC-I regulation, acting as a positive and negative regulator of MyHC-I expression during embryonic and fetal myogenesis, respectively. During embryonic myogenesis, Sox6 positively regulates MyHC-I via transcriptional activation of Mef2C, whereas during fetal myogenesis, Sox6 requires and cooperates with the transcription factor Nfix in repressing MyHC-I expression. Mechanistically, Nfix is necessary for Sox6 binding to the MyHC-I promoter and thus for Sox6 repressive function, revealing a key role for Nfix in driving Sox6 activity. This feature is evolutionarily conserved, since the orthologs Nfixa and Sox6 contribute to repression of the slow-twitch phenotype in zebrafish embryos. These data demonstrate functional cooperation between Sox6 and Nfix in regulating MyHC-I expression during prenatal muscle development.


Assuntos
Feto/embriologia , Músculo Esquelético/embriologia , Cadeias Pesadas de Miosina/metabolismo , Fatores de Transcrição NFI/metabolismo , Fatores de Transcrição SOXD/genética , Transcrição Gênica , Proteínas de Peixe-Zebra/metabolismo , Animais , Sequência Conservada , Embrião não Mamífero/metabolismo , Evolução Molecular , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição MEF2/metabolismo , Camundongos , Modelos Biológicos , Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética
4.
Cell Rep ; 14(9): 2238-2249, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26923583

RESUMO

Nfix belongs to a family of four highly conserved proteins that act as transcriptional activators and/or repressors of cellular and viral genes. We previously showed a pivotal role for Nfix in regulating the transcriptional switch from embryonic to fetal myogenesis. Here, we show that Nfix directly represses the Myostatin promoter, thus controlling the proper timing of satellite cell differentiation and muscle regeneration. Nfix-null mice display delayed regeneration after injury, and this deficit is reversed upon in vivo Myostatin silencing. Conditional deletion of Nfix in satellite cells results in a similar delay in regeneration, confirming the functional requirement for Nfix in satellite cells. Moreover, mice lacking Nfix show reduced myofiber cross sectional area and a predominant slow twitching phenotype. These data define a role for Nfix in postnatal skeletal muscle and unveil a mechanism for Myostatin regulation, thus providing insights into the modulation of its complex signaling pathway.


Assuntos
Fibras Musculares Esqueléticas/fisiologia , Miostatina/genética , Fatores de Transcrição NFI/fisiologia , Regeneração , Animais , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Inativação Gênica , Camundongos Transgênicos , Mioblastos/fisiologia , Miostatina/metabolismo
5.
Development ; 140(7): 1528-36, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23482488

RESUMO

During mouse skeletal muscle development, the Nfix gene has a pivotal role in regulating fetal-specific transcription. Zebrafish and mice share related programs for muscle development, although zebrafish develops at a much faster rate. In fact, although mouse fetal muscle fibers form after 15 days of development, in fish secondary muscle fibers form by 48 hours post-fertilization in a process that until now has been poorly characterized mechanically. In this work, we studied the zebrafish ortholog Nfix (nfixa) and its role in the proper switch to the secondary myogenic wave. This allowed us to highlight evolutionarily conserved and divergent functions of Nfix. In fact, the knock down of nfixa in zebrafish blocks secondary myogenesis, as in mouse, but also alters primary slow muscle fiber formation. Moreover, whereas Nfix mutant mice are motile, nfixa knockdown zebrafish display impaired motility that probably depends upon disruption of the sarcoplasmic reticulum. We conclude that, during vertebrate evolution, the transcription factor Nfix lost some specific functions, probably as a consequence of the different environment in which teleosts and mammals develop.


Assuntos
Evolução Molecular , Músculo Esquelético/embriologia , Fatores de Transcrição NFI/fisiologia , Vertebrados/genética , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Sequência Conservada/fisiologia , Embrião de Mamíferos , Embrião não Mamífero , Especiação Genética , Camundongos , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Fatores de Transcrição NFI/genética , Filogenia , Vertebrados/embriologia , Vertebrados/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
6.
Cell ; 140(4): 554-66, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20178747

RESUMO

Skeletal myogenesis, like hematopoiesis, occurs in successive developmental stages that involve different cell populations and expression of different genes. We show here that the transcription factor nuclear factor one X (Nfix), whose expression is activated by Pax7 in fetal muscle, in turn activates the transcription of fetal specific genes such as MCK and beta-enolase while repressing embryonic genes such as slow myosin. In the case of the MCK promoter, Nfix forms a complex with PKC theta that binds, phosphorylates, and activates MEF2A. Premature expression of Nfix activates fetal and suppresses embryonic genes in embryonic muscle, whereas muscle-specific ablation of Nfix prevents fetal and maintains embryonic gene expression in the fetus. Therefore, Nfix acts as a transcriptional switch from embryonic to fetal myogenesis.


Assuntos
Desenvolvimento Muscular , Músculo Esquelético/embriologia , Fatores de Transcrição NFI/metabolismo , Transcrição Gênica , Animais , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Isoenzimas/metabolismo , Fatores de Transcrição MEF2 , Camundongos , Fatores de Regulação Miogênica/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fator de Transcrição PAX7/metabolismo , Fosfopiruvato Hidratase , Proteína Quinase C/metabolismo , Proteína Quinase C-theta
7.
Stem Cells ; 27(1): 157-64, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18845762

RESUMO

Mesoangioblasts have been characterized as a population of vessel-associated stem cells able to differentiate into several mesodermal cell types, including skeletal muscle. Here, we report that the paired box transcription factor Pax3 plays a crucial role in directing mouse mesoangioblasts toward skeletal myogenesis in vitro and in vivo. Mesoangioblasts isolated from the aorta of Pax3 null embryos are severely impaired in skeletal muscle differentiation, whereas most other differentiation programs are not affected by the absence of Pax3. Moreover, Pax3(-/-) null mesoangioblasts failed to rescue the myopathic phenotype of the alpha-sarcoglycan mutant mouse. In contrast, mesoangioblasts from Pax3 gain of function, Pax3(PAX3-FKHR/+), mice display enhanced myogenesis in vitro and are more efficient in regenerating new muscle fibers in this model of muscular dystrophy. These data demonstrate that Pax3 is required for the differentiation of mesoangioblast stem cells into skeletal muscle, in keeping with its role in orchestrating entry into the myogenic program.


Assuntos
Vasos Sanguíneos/citologia , Diferenciação Celular , Embrião de Mamíferos/citologia , Mesoderma/citologia , Músculo Esquelético/citologia , Fatores de Transcrição Box Pareados/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Vasos Sanguíneos/enzimologia , Osso e Ossos/citologia , Proliferação de Células , Forma Celular , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Camundongos , Desenvolvimento Muscular , Distrofia Muscular Animal/metabolismo , Miócitos de Músculo Liso/citologia , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/deficiência , Fenótipo , Sarcoglicanas/biossíntese
8.
Dev Biol ; 304(2): 633-51, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17292343

RESUMO

Skeletal muscle development occurs asynchronously and it has been proposed to be dependent upon the generation of temporally distinct populations of myogenic cells. This long-held hypothesis has not been tested directly due to the inability to isolate and analyze purified populations of myoblasts derived from specific stages of prenatal development. Using a mouse strain with the GFP reporter gene targeted into the Myf5 locus, a cell-sorting method was developed for isolating embryonic and fetal myoblasts. The two types of myoblasts show an intrinsic difference in fusion ability, proliferation, differentiation and response to TGFbeta, TPA and BMP-4 in vitro. Microarray and quantitative PCR were used to identify differentially expressed genes both before and after differentiation, thus allowing a precise phenotypic analysis of the two populations. Embryonic and fetal myoblasts differ in the expression of a number of transcription factors and surface molecules, which may control different developmental programs. For example, only embryonic myoblasts express a Hox code along the antero-posterior axis, indicating that they possess direct positional information. Taken together, the data presented here demonstrate that embryonic and fetal myoblasts represent intrinsically different myogenic lineages and provide important information for the understanding of the molecular mechanisms governing skeletal muscle development.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/citologia , Feto/citologia , Músculo Esquelético/citologia , Mioblastos/citologia , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/farmacologia , Linhagem da Célula , Células Cultivadas , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Feto/efeitos dos fármacos , Feto/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Músculo Esquelético/embriologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Acetato de Tetradecanoilforbol/farmacologia , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA