Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anim Sci J ; 89(8): 1214-1219, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29900636

RESUMO

Myogenesis is precisely proceeded by myogenic regulatory factors. Myogenic stem cells are activated, proliferated and fused into a multinuclear myofiber. Pax7, paired box 7, one of the earliest markers during myogenesis. It has been reported that Pax7 regulates the muscle marker genes, Myf5 and MyoD toward differentiation. The possible roles of Pax7 in myogenic cells have been well researched. However, it has not yet been clarified if Pax7 itself is able to induce myogenic fate in nonmyogenic lineage cells. In this study, we performed experiments using stably expressed Pax7 in 3T3-L1 preadipocytes to elucidate if Pax7 inhibits adipogenesis. We found that Pax7 represses adipogenic markers and prevents differentiation. These cells showed decreased expression of PDGFRα, PPARγ and Fabp4 and inhibited forming lipid droplets.


Assuntos
Adipócitos/fisiologia , Adipogenia/genética , Diferenciação Celular/genética , Fator de Transcrição PAX7/fisiologia , Células 3T3-L1 , Animais , Regulação para Baixo/genética , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Expressão Gênica/genética , Gotículas Lipídicas , Camundongos , Desenvolvimento Muscular , Proteína MyoD/fisiologia , Fator Regulador Miogênico 5/fisiologia , PPAR gama/genética , PPAR gama/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
2.
Gene Expr Patterns ; 20(1): 1-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26547039

RESUMO

Myogenic regulatory factors (MRFs) are muscle-specific basic helix-loop-helix (bHLH) transcription factor that plays an essential role in regulating skeletal muscle development and growth. To investigate molecular characterization of Myf5 and compare the expressional patterns of the four MRFs, we cloned the Myf5 cDNA sequence and analyzed the MRFs expressional patterns using quantitative real-time polymerase chain reaction in Chinese perch (Siniperca chuatsi). Sequence analysis indicated that Chinese perch Myf5 and other MRFs shared a highly conserved bHLH domain with those of other vertebrates. Sequence alignment and phylogenetic tree showed that Chinese perch MRFs had the highest identity with the MRFs of Epinephelus coioides. Spatio-temporal expressional patterns revealed that the MRFs were primarily expressed in muscle, especially in white muscle. During embryonic development period, Myf5, MyoD and MyoG mRNAs had a steep increase at neurula stage, and their highest expressional level was predominantly observed at hatching period. Whereas the highest expressional level of the MRF4 was observed at the muscular effect stage. The expressional patterns of post-embryonic development showed that the Myf5, MyoD and MyoG mRNAs were highest at 90 days post-hatching (dph). Furthermore, starvation and refeeding results showed that the transcription of the MRFs in the fast skeletal muscle of Chinese perch responded quickly to a single meal after 7 days of fasting. It indicated that the MRFs might contribute to muscle recovery after refeeding in Chinese perch.


Assuntos
Peixes/genética , Perfilação da Expressão Gênica , Fator Regulador Miogênico 5/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar , Jejum/metabolismo , Humanos , Dados de Sequência Molecular , Fator Regulador Miogênico 5/fisiologia , Filogenia , Reação em Cadeia da Polimerase em Tempo Real , Alinhamento de Sequência
3.
Science ; 347(6223): 760-4, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25678660

RESUMO

A new Late Jurassic docodontan shows specializations for a subterranean lifestyle. It is similar to extant subterranean golden moles in having reduced digit segments as compared to the ancestral phalangeal pattern of mammaliaforms and extant mammals. The reduction of digit segments can occur in mammals by fusion of the proximal and intermediate phalangeal precursors, a developmental process for which a gene and signaling network have been characterized in mouse and human. Docodontans show a positional shift of thoracolumbar ribs, a developmental variation that is controlled by Hox9 and Myf5 genes in extant mammals. We argue that these morphogenetic mechanisms of modern mammals were operating before the rise of modern mammals, driving the morphological disparity in the earliest mammaliaform diversification.


Assuntos
Evolução Biológica , Falanges dos Dedos da Mão/anatomia & histologia , Falanges dos Dedos da Mão/crescimento & desenvolvimento , Mamíferos/anatomia & histologia , Mamíferos/crescimento & desenvolvimento , Morfogênese/fisiologia , Animais , China , Pé/anatomia & histologia , Pé/crescimento & desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Humanos , Mamíferos/genética , Camundongos , Morfogênese/genética , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/fisiologia
4.
J Dent Res ; 94(3): 421-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25576472

RESUMO

The tongue is a muscular organ that is essential in vertebrates for important functions, such as food intake and communication. Little is known about regulation of myogenic progenitors during tongue development when compared with the limb or trunk region. In this study, we investigated the relationship between different myogenic subpopulations and the function of canonical Wnt signaling in regulating these subpopulations. We found that Myf5- and MyoD-expressing myogenic subpopulations exist during embryonic tongue myogenesis. In the Myf5-expressing myogenic progenitors, there is a cell-autonomous requirement for canonical Wnt signaling for cell migration and differentiation. In contrast, the MyoD-expressing subpopulation does not require canonical Wnt signaling during tongue myogenesis. Taken together, our results demonstrate that canonical Wnt signaling differentially regulates the Myf5- and MyoD-expressing subpopulations during tongue myogenesis.


Assuntos
Desenvolvimento Muscular/fisiologia , Células-Tronco/fisiologia , Língua/embriologia , Via de Sinalização Wnt/fisiologia , Animais , Proteína Axina/análise , Proteína Axina/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Movimento Celular/fisiologia , Camundongos , Fibras Musculares Esqueléticas/citologia , Proteína MyoD/análise , Proteína MyoD/fisiologia , Fator Regulador Miogênico 5/análise , Fator Regulador Miogênico 5/fisiologia , RNA não Traduzido/análise , RNA não Traduzido/fisiologia , Língua/citologia , beta Catenina/análise , beta Catenina/fisiologia
5.
Am J Physiol Regul Integr Comp Physiol ; 306(11): R837-44, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24671243

RESUMO

Chronic alcohol abuse is associated with skeletal muscle myopathy. Previously, we demonstrated that chronic binge alcohol (CBA) consumption by rhesus macaques accentuates skeletal muscle wasting at end-stage of simian immunodeficiency virus (SIV) infection. A proinflammatory, prooxidative milieu and enhanced ubiquitin proteasome activity were identified as possible mechanisms leading to loss of skeletal muscle. The possibility that impaired regenerative capacity, as reflected by the ability of myoblasts derived from satellite cell (SCs) to differentiate into myotubes has not been examined. We hypothesized that the inflammation and oxidative stress in skeletal muscle from CBA animals impair the differentiation capacity of myoblasts to form new myofibers in in vitro assays. We isolated primary myoblasts from the quadriceps femoris of rhesus macaques that were administered CBA or isocaloric sucrose (SUC) for 19 mo. Proliferation and differentiation potential of cultured myoblasts were examined in vitro. Myoblasts from the CBA group had significantly reduced PAX7, MYOD1, MYOG, MYF5, and MEF2C expression. This was associated with decreased myotube formation as evidenced by Jenner-Giemsa staining and myonuclei fusion index. No significant difference in the proliferative ability, cell cycle distribution, or autophagy was detected between myoblasts isolated from CBA and SUC groups. Together, these results reflect marked dysregulation of myoblast myogenic gene expression and myotube formation, which we interpret as evidence of impaired skeletal muscle regenerative capacity in CBA-administered macaques. The contribution of this mechanism to alcoholic myopathy warrants further investigation.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Macaca mulatta/fisiologia , Proteínas Musculares/fisiologia , Mioblastos Esqueléticos/patologia , Animais , Proliferação de Células , Técnicas In Vitro , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Masculino , Modelos Animais , Proteínas Musculares/genética , Proteína MyoD/genética , Proteína MyoD/fisiologia , Mioblastos Esqueléticos/fisiologia , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/fisiologia , Miogenina/genética , Miogenina/fisiologia , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/fisiologia
6.
Zebrafish ; 10(4): 486-99, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23992145

RESUMO

Myf5, a myogenic regulatory factor, plays a key role in regulating muscle differentiation. However, it is not known if Myf5 has a regulatory role during early embryogenesis. Here, we used myf5-morpholino oligonucleotides [MO] to knock down myf5 expression and demonstrated a series of results pointing to the functional roles of Myf5 during early embryogenesis: (1) reduced head size resulting from abnormal morphology in the cranial skeleton; (2) decreased expressions of the cranial neural crest (CNC) markers foxd3, sox9a, dlx2, and col2a1; (3) defect in the chondrogenic neural crest similar to that of fgf3 morphants; (4) reduced fgf3/fgf8 transcripts in the cephalic mesoderm rescued by co-injection of myf5 wobble-mismatched mRNA together with myf5-MO1 during 12 h postfertilization; (5) abnormal patterns of axial and non-axial mesoderm causing expansion of the dorsal organizer, and (6) increased bmp4 gradient, but reduced fgf3/fgf8 marginal gradient, during gastrulation. Interestingly, overexpression of fgf3 could rescue the cranial cartilage defects caused by myf5-MO1, suggesting that Myf5 modulates craniofacial cartilage development through the fgf3 signaling pathway. Together, the loss of Myf5 function results in a cascade effect that begins with abnormal formation of the dorsal organizer during gastrulation, causing, in turn, defects in the CNC and cranial cartilage of myf5-knockdown embryos.


Assuntos
Região Branquial/embriologia , Cartilagem Articular/crescimento & desenvolvimento , Desenvolvimento Embrionário , Fator Regulador Miogênico 5/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Condrogênese , Técnicas de Silenciamento de Genes , Cabeça/embriologia , Músculos/embriologia , Fator Regulador Miogênico 5/deficiência , Crista Neural/embriologia , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/deficiência
7.
Dev Biol ; 376(2): 236-44, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23384562

RESUMO

The Myf5 gene plays an important role in myogenic determination during mouse embryo development. Multiple genomic regions of the Mrf4-Myf5 locus have been characterised as enhancer sequences responsible for the complex spatiotemporal expression of the Myf5 gene at the onset of myogenesis. These include an enhancer sequence, located at -111 kb upstream of the Myf5 transcription start site, which is responsible of Myf5 activation in ventral somitic domains (Ribas et al., 2011. Dev. Biol. 355, 372-380). We show that the -111 kb-Myf5 enhancer also directs transgene expression in some limb muscles, and is active at foetal as well as embryonic stages. We have carried out further characterisation of the regulation of this enhancer and show that the paired-box Pax3 transcription factor binds to it in vitro as in vivo, and that Pax binding sites are essential for its activity. This requirement is independent of the previously reported regulation by TEAD transcription factors. Six1/4 which, like Pax3, are important upstream regulators of myogenesis, also bind in vivo to sites in the -111 kb-Myf5 enhancer and modulate its activity. The -111 kb-Myf5 enhancer therefore shares common functional characteristics with another Myf5 regulatory sequence, the hypaxial and limb 145 bp-Myf5 enhancer, both being directly regulated in vivo by Pax3 and Six1/4 proteins. However, in the case of the -111 kb-Myf5 enhancer, Six has less effect and we conclude that Pax regulation plays a major role in controlling this aspect of the Myf5 gene expression at the onset of myogenesis in the embryo.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Desenvolvimento Muscular , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/fisiologia , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/fisiologia , Transativadores/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Fator II de Transcrição COUP/metabolismo , Elementos Facilitadores Genéticos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Modelos Biológicos , Modelos Genéticos , Dados de Sequência Molecular , Fator de Transcrição PAX3 , Plasmídeos/metabolismo , Homologia de Sequência do Ácido Nucleico
8.
Dev Biol ; 350(2): 464-75, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21147088

RESUMO

Differentiation often requires conversion of analogue signals to a stable binary output through positive feedback. Hedgehog (Hh) signalling promotes myogenesis in the vertebrate somite, in part by raising the activity of muscle regulatory factors (MRFs) of the Myod family above a threshold. Hh is known to enhance MRF expression. Here we show that Hh is also essential at a second step that increases Myod protein activity, permitting it to promote Myogenin expression. Hh acts by inducing expression of cdkn1c (p57(Kip2)) in slow muscle precursor cells, but neither Hh nor Cdkn1c is required for their cell cycle exit. Cdkn1c co-operates with Myod to drive differentiation of several early zebrafish muscle fibre types. Myod in turn up-regulates cdkn1c, thereby providing a positive feedback loop that switches myogenic cells to terminal differentiation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/fisiologia , Desenvolvimento Muscular , Proteína MyoD/fisiologia , Animais , Diferenciação Celular , Retroalimentação Fisiológica , Proteínas Hedgehog/fisiologia , Fator Regulador Miogênico 5/fisiologia , Miogenina/genética , Transdução de Sinais , Peixe-Zebra
9.
Dev Cell ; 18(4): 655-61, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20412779

RESUMO

Hox genes are essential for the patterning of the axial skeleton. Hox group 10 has been shown to specify the lumbar domain by setting a rib-inhibiting program in the presomitic mesoderm (PSM). We have now produced mice with ribs in every vertebra by ectopically expressing Hox group 6 in the PSM, indicating that Hox genes are also able to specify the thoracic domain. We show that the information provided by Hox genes to specify rib-containing and rib-less areas is first interpreted in the myotome through the regional-specific control of Myf5 and Myf6 expression. This information is then transmitted to the sclerotome by a system that includes FGF and PDGF signaling to produce vertebrae with or without ribs at different axial levels. Our findings offer a new perspective of how Hox genes produce global patterns in the axial skeleton and support a redundant nonmyogenic role of Myf5 and Myf6 in rib formation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Fator Regulador Miogênico 5/fisiologia , Fatores de Regulação Miogênica/fisiologia , Animais , Desenvolvimento Ósseo , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Homeobox A10 , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Modelos Genéticos , Desenvolvimento Muscular , Fenótipo , Fator de Crescimento Derivado de Plaquetas/metabolismo
10.
Biochem J ; 428(2): 213-21, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20307260

RESUMO

Recurrent injuries eventually exhaust the capacity of skeletal muscle to fully restore or regenerate its cellular architecture. Therefore a comprehensive understanding of the muscle regeneration programme is needed to provide a platform for new therapies for devastating diseases such as Duchenne muscular dystrophy. To begin to decipher the molecular programme that directs muscle regeneration, we undertook an unbiased strategy using microarray analysis of cardiotoxin-injured skeletal muscle at defined time periods in the adult mouse. Using this strategy, we identified Tceal7 [transcription elongation factor A (SII)-like 7], which was dynamically regulated during muscle regeneration. Our studies revealed that Tceal7 was restricted to the skeletal muscle lineage during embryogenesis. Using transgenic technologies and transcriptional assays, we defined an upstream 0.7 kb fragment of the Tceal7 gene that directed the LacZ reporter to the developing skeletal muscle lineage. Analysis of the Tceal7 promoter revealed evolutionarily conserved E-box motifs within the 0.7 kb upstream fragment that were essential for promoter activity, as mutation of the E-box motifs resulted in the loss of reporter expression in the somites of transgenic embryos. Furthermore, we demonstrated that MRFs (myogenic regulatory factors) were Tceal7 upstream transactivators using transcriptional assays, EMSAs (electrophoretic mobility-shift assays), and ChIP (chromatin immunoprecipitation) assays. Overexpression of Tceal7 in C2C12 myoblasts decreased cellular proliferation and enhanced differentiation. Further studies revealed that p27 expression was up-regulated following Tceal7 overexpression. These studies support the hypothesis that MRFs transactivate Tceal7 gene expression and promote muscle differentiation during muscle development and regeneration.


Assuntos
Diferenciação Celular/fisiologia , Músculo Esquelético/metabolismo , Fatores de Regulação Miogênica/fisiologia , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células , Imunoprecipitação da Cromatina , Elementos E-Box/genética , Elementos E-Box/fisiologia , Ensaio de Desvio de Mobilidade Eletroforética , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/citologia , Proteína MyoD/genética , Proteína MyoD/fisiologia , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/fisiologia , Fatores de Regulação Miogênica/genética , Miogenina/genética , Miogenina/fisiologia , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
11.
Mech Dev ; 126(10): 852-62, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19643178

RESUMO

Klhl31 is an orthologue of Drosophila Kelch and belongs to a family of Kelch-like proteins in vertebrates. Members of this family contain multiple protein domains, including an amino-terminal broad complex/tram-track/bric-a-brac (BTB) or poxvirus and zinc finger (POZ) domain, carboxy-terminal Kelch repeats and a central linker region. We show that Klhl31 is highly expressed in the developing heart, the somite myotome and later in differentiated skeletal muscle and the myocardium. In developing somites expression of Klhl31 was initiated in the epaxial domain of the myotome, shortly after the skeletal muscle specific bHLH transcription factor, MyoD, was first expressed. Klhl31 remained expressed in skeletal muscle throughout embryonic and fetal development. Tissue ablations and rescue experiments that regulate myogenesis also govern expression of Klhl31 expression in somites. In particular, axial tissues, neural tube, floor plate and notochord, and surface ectoderm, provide combinatorial cues for myogenesis and the appropriate expression of Klhl31. We show that a combination of myogenic signals, Shh and either Wnt-1 or Wnt-6, are sufficient for Klhl31 expression in the dorsal somite. Furthermore, ectopic expression of Myf-5 led to expression of Klhl31 in the developing neural tube, indicating that Klhl31 is a novel and integral part of vertebrate myogenesis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/embriologia , Fator Regulador Miogênico 5/fisiologia , Proteínas Repressoras/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Primers do DNA , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Eletroporação , Glicoproteínas/fisiologia , Proteínas Hedgehog/fisiologia , Hibridização In Situ , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Filogenia , Ratos , Proteínas Repressoras/genética , Proteínas Wnt/fisiologia , Proteína Wnt1/fisiologia
12.
J Orthop Res ; 27(4): 504-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18932240

RESUMO

Chronic rotator cuff tendon tears lead to fatty infiltration and muscle atrophy with impaired physiological functions of the affected muscles. However, the cellular and molecular mechanisms of corresponding pathophysiological processes remain unknown. The purpose of this study was to characterize the expression pattern of adipogenic (PPARgamma, C/EBPbeta) and myogenic (myostatin, myogenin, Myf-5) transcription factors in infraspinatus muscle of sheep after tenotomy, implantation of a tension device, refixation of the tendon, and rehabilitation, reflecting a model of chronic rotator cuff tears. In contrast to human patients, the presented sheep model allows a temporal evaluation of the expression of a given marker in the same individual over time. Semiquantitative RT/PCR analysis of PPARgammaã, myostatin, myogenin, Myf-5, and C/EBPbeta transcript levels was carried out with sheep muscle biopsy-derived total RNA. We found a significantly increased expression of Myf-5 and PPARgamma after tenotomy and a significant change for Myf-5 and C/EBPbeta after continuous traction and refixation. This experimental sheep model allows the molecular analysis of pathomechanisms of muscular changes after rotator cuff tear. The results point to a crucial role of the transcription factors PPARgamma, C/EBPbeta, and Myf-5 in impairment and regeneration of rotator cuff muscles after tendon tears in sheep.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/genética , Fator Regulador Miogênico 5/genética , Miogenina/genética , Miostatina/genética , PPAR gama/genética , Lesões do Manguito Rotador , Manguito Rotador/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Modelos Animais de Doenças , Expressão Gênica , Fator Regulador Miogênico 5/fisiologia , PPAR gama/fisiologia , RNA Mensageiro/análise , Regeneração , Ovinos , Traumatismos dos Tendões/metabolismo
13.
Mol Biol Rep ; 36(6): 1497-504, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18752038

RESUMO

Myogenic Regulatory Factors (MRFs), a family of basic helix-loop-helix (bHLH) transcription factors, play important roles in regulating skeletal muscle development and growth. Myf5, the primary factor of MRFs, initiates myogenesis. Its expression pattern during somitomyogenesis in some fish has been revealed. To further study its effect on fish muscle during postembryonic growth, characterization and function analysis of myf5 cDNA were carried out in largemouth bass. The 1,093 bp cDNA sequence was identified by RT-PCR and 3'RACE, then the ORF of Myf5 cDNA was cloned into the expression vector pcDNA3.1(-)/mycHisB. The recombinant plasmid pcDNA3.1(-)/mycHisB-Myf5 was injected into the dorsal muscle of tilapias. RT-PCR and histochemical results showed that the exogenous gene was transcribed and translated in vivo. Its effect on muscle growth focused on myofiber hypertrophy in white muscle 60 days post injection. This indicated that overexpression of Myf5 can promote myogenesis during the fish muscle postembryonic growth period.


Assuntos
Bass/genética , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/fisiologia , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/administração & dosagem , DNA Complementar/genética , DNA Complementar/farmacologia , Hipertrofia/induzido quimicamente , Músculo Esquelético/efeitos dos fármacos , Miofibrilas , Fator Regulador Miogênico 5/administração & dosagem
14.
Am J Physiol Endocrinol Metab ; 295(2): E297-304, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18492772

RESUMO

Recent studies have shown that administration of peroxisome proliferator-activated receptor-beta (PPARbeta) agonists enhances fatty acid oxidation in rodent and human skeletal muscle and that muscle-restricted PPARbeta overexpression affects muscle metabolic profile by increasing oxidative myofiber number, which raises the possibility that PPARbeta agonists alter muscle morphology in adult animals. This possibility was examined in this study in which adult mice were treated with a PPARbeta agonist, and the resulting changes in myofiber metabolic phenotype and angiogenesis were quantified in tibialis anterior muscles. The findings indicate a muscle remodeling that is completed within 2 days and is characterized by a 1.63-fold increase in oxidative fiber number and by a 1.55-fold increase in capillary number. These changes were associated with a quick and transient upregulation of myogenic and angiogenic markers. Both myogenic and angiogenic responses were dependent on the calcineurin pathway, as they were blunted by cyclosporine A administration. In conclusion, the data indicate that PPARbeta activation is associated with a calcineurin-dependent effect on muscle morphology that enhances the oxidative phenotype.


Assuntos
Calcineurina/fisiologia , Músculo Esquelético/fisiologia , PPAR beta/agonistas , Condicionamento Físico Animal/fisiologia , Tiazóis/farmacologia , Animais , Inibidores de Calcineurina , Ciclosporina/farmacologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Proteína MyoD/fisiologia , Fator Regulador Miogênico 5/fisiologia , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Succinato Desidrogenase/metabolismo
15.
Cell Stem Cell ; 2(1): 22-31, 2008 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-18371418

RESUMO

Muscle satellite cells have been shown to be a heterogeneous population of committed myogenic progenitors and noncommitted stem cells. This hierarchical composition of differentiating progenitors and self-renewable stem cells assures the extraordinary regenerative capacity of skeletal muscles. Recent studies have revealed a role for asymmetric division in satellite cell maintenance and offer novel insights into the regulation of satellite cell function by the niche. A thorough understanding of the molecular regulation and cell fate determination of satellite cells and other potential stem cells resident in muscle is essential for successful stem cell-based therapies to treat muscular diseases.


Assuntos
Diferenciação Celular , Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Comunicação Celular , Proteínas de Ciclo Celular/fisiologia , Regulação da Expressão Gênica , Humanos , Camundongos , Distrofias Musculares/terapia , Fator Regulador Miogênico 5/fisiologia , Fator de Transcrição PAX7/fisiologia , Regeneração , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais , Nicho de Células-Tronco/citologia , Transplante de Células-Tronco
16.
Endocrinology ; 149(7): 3698-707, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18372333

RESUMO

Transcription factors regulate tissue-specific differentiation of pluripotent mesenchyme to osteoblast (OB), myoblast (MB), and other lineages. Osterix (Osx) is an essential transcription factor for bone development because knockout results in lack of a mineralized skeleton. The proximal Osx promoter contains numerous binding sequences for MyoD and 14 repeats of a binding sequence for Myf5. These basic helix-loop-helix (bHLH) transcription factors have a critical role in MB differentiation and muscle development. We tested the hypothesis that bHLH transcription factors also support OB differentiation through regulation of Osx. Transfection of a MyoD expression vector into two primitive mesenchymal cell lines, C3H/10T1/2 and C2C12, stimulated a 1.2-kb Osx promoter-luciferase reporter 70-fold. Myf5 stimulated the Osx promoter 6-fold. Deletion analysis of the promoter revealed that one of three proximal bHLH sites is essential for MyoD activity. The Myf5 repeat conferred 60% of Myf5 activity with additional upstream sequence required for full activity. MyoD bound the active bHLH sequence and its 3'-flanking region, as shown by EMSA and chromatin immunoprecipitation assays. Real-time PCR revealed that primitive C2C12 and C3H/10T1/2 cells, pre-osteoblastic MC3T3 cells, and undifferentiated primary marrow stromal cells express the muscle transcription factors. C2C12 cells, which differentiate to MB spontaneously and form myotubules, were treated with bone morphogenetic protein 2 (BMP-2) to induce OB differentiation. BMP-2 stimulated expression of Osx and the differentiation marker alkaline phosphatase and blocked myotubule development. BMP-2 suppressed the muscle transcription factor myogenin, but expression of MyoD and Myf5 persisted. Silencing of MyoD inhibited BMP-2 stimulation of Osx and blocked the later appearance of bone alkaline phosphatase. MyoD support of Osx transcription contributes to early OB differentiation.


Assuntos
Diferenciação Celular , Proteína MyoD/fisiologia , Osteoblastos/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Células 3T3 , Animais , Sequência de Bases , Western Blotting , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Sequências Hélice-Alça-Hélice/genética , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/metabolismo , Fator Regulador Miogênico 5/fisiologia , Osteoblastos/citologia , Ligação Proteica , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp7
17.
Development ; 135(9): 1597-604, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18367555

RESUMO

The development of myogenic cells is mainly determined by expression of two myogenic factors, Myf5 and Myod1 (MyoD), which genetically compensate for each other during embryogenesis. Here, we demonstrate by conditional cell ablation in mice that Myf5 determines a distinct myogenic cell population, which also contains some Myod1-positive cells. Ablation of this lineage uncovers the presence of a second autonomous myogenic lineage, which superseded Myf5-dependent myogenic cells and expressed Myod1. By contrast, ablation of myogenin-expressing cells erased virtually all differentiated muscle cells, indicating that some aspects of the myogenic program are shared by most skeletal muscle cells. We conclude that Myf5 and Myod1 define different cell lineages with distinct contributions to muscle precursor cells and differentiated myotubes. Individual myogenic cell lineages seem to substitute for each other within the developing embryo.


Assuntos
Linhagem da Célula/fisiologia , Desenvolvimento Muscular/fisiologia , Mioblastos Esqueléticos/citologia , Fator Regulador Miogênico 5/fisiologia , Animais , Animais Recém-Nascidos , Osso e Ossos/anormalidades , Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Músculo Esquelético/anormalidades , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Mutação , Proteína MyoD/genética , Proteína MyoD/fisiologia , Mioblastos Esqueléticos/fisiologia , Fator Regulador Miogênico 5/genética
18.
Artigo em Inglês | MEDLINE | ID: mdl-19329572

RESUMO

Muscle satellite cells are responsible for the postnatal growth and robust regeneration capacity of adult skeletal muscle. A subset of satellite cells purified from adult skeletal muscle is capable of repopulating the satellite cell pool, suggesting that it has direct therapeutic potential for treating degenerative muscle disease. Satellite cells uniformly express the transcription factor Pax7, and Pax7 is required for satellite cell viability and to give rise to myogenic precursors that express the basic helix-loop-helic (bHLH) transcription factors Myf5 and MyoD. Pax7 activates expression of target genes such as Myf5 and MyoD through recruitment of the Wdr5/Ash2L/MLL2 histone methyltransferase complex. Extensive genetic analysis has revealed that Myf5 and MyoD are required for myogenic determination, whereas myogenin and MRF4 have roles in terminal differentiation. Using a Myf5-Cre knockin allele and an R26R-YFP Cre reporter, we observed that in vivo about 10% of satellite cells only express Pax7 and have never expressed Myf5. Moreover, we found that Pax7(+)/Myf5(-) satellite cells give rise to Pax7(+)/Myf5(+) satellite cells through basal-apical asymmetric cell divisions. Therefore, satellite cells in skeletal muscle are a heterogeneous population composed of satellite stem cells (Pax7(+)/Myf5(-)) and satellite myogenic cells (Pax7(+)/Myf5(+)). Evidence is accumulating that indicates that satellite stem cells represent a true stem cell reservoir, and targeting mechanisms that regulate their function represents an important therapeutic strategy for the treatment of neuromuscular disease.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Desenvolvimento Muscular , Doenças Musculares/terapia , Proteína MyoD/genética , Proteína MyoD/fisiologia , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/fisiologia , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/fisiologia , Regeneração
19.
Dev Biol ; 310(1): 140-53, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17727833

RESUMO

Craniofacial development involves cranial neural crest (CNC) and mesoderm-derived cells. TGF-beta signaling plays a critical role in instructing CNC cells to form the craniofacial skeleton. However, it is not known how TGF-beta signaling regulates the fate of mesoderm-derived cells during craniofacial development. In this study, we show that occipital somites contribute to the caudal region of mammalian skull development. Conditional inactivation of Tgfbr2 in mesoderm-derived cells results in defects of the supraoccipital bone with meningoencephalocele and discontinuity of the neural arch of the C1 vertebra. At the cellular level, loss of TGF-beta signaling causes decreased chondrocyte proliferation and premature differentiation of cartilage to bone. Expression of Msx2, a critical factor in the formation of the dorsoventral axis, is diminished in the Tgfbr2 mutant. Significantly, overexpression of Msx2 in Myf5-Cre;Tgfbr2flox/flox mice partially rescues supraoccipital bone development. These results suggest that the TGF-beta/Msx2 signaling cascade is critical for development of the caudal region of the skull.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Homeodomínio/fisiologia , Crânio/embriologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Condrogênese , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Morfogênese , Fator Regulador Miogênico 5/fisiologia , Crista Neural/embriologia , Transdução de Sinais , Crânio/fisiologia , Somitos/embriologia
20.
Cell ; 129(5): 859-61, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17540166

RESUMO

Satellite cells are crucial for maintaining muscle homeostasis and for regeneration following injury. In this issue, Kuang et al. (2007) reveal that muscle satellite cells are a heterogeneous mixture of stem cells and committed myogenic progenitors. They show that asymmetric division of stem cells in the satellite cell niche is a mechanism for generating these two populations.


Assuntos
Células Satélites de Músculo Esquelético/citologia , Animais , Divisão Celular , Camundongos , Fator Regulador Miogênico 5/fisiologia , Fator de Transcrição PAX7/fisiologia , Células Satélites de Músculo Esquelético/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...