Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 288(11): 7676-7687, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23364797

RESUMO

Facilitates chromatin transcription (FACT) functions to reorganize nucleosomes by acting as a histone chaperone that destabilizes and restores nucleosomal structure. The FACT complex is composed of two subunits: SSRP1 and SPT16. We have discovered that myogenin interacts with the FACT complex. Transfection of FACT subunits with myogenin is highly stimulatory for endogenous muscle gene expression in 10T1/2 cells. We have also found that FACT subunits do not associate with differentiation-specific genes while C2C12 cells are proliferating but are recruited to muscle-specific genes as differentiation initiates and then dissociate as differentiation proceeds. The recruitment is dependent on myogenin, as knockdowns of myogenin show no recruitment of the FACT complex. These data suggest that FACT is involved in the early steps of gene activation through its histone chaperone activities that serve to open the chromatin structure and facilitate transcription. Consistent with this hypothesis, we find that nucleosomes are depleted at muscle-specific promoters upon differentiation and that this activity is dependent on the presence of FACT. Our results show that the FACT complex promotes myogenin-dependent transcription and suggest that FACT plays an important role in the establishment of the appropriate transcription profile in a differentiated muscle cell.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Histonas/química , Miogenina/fisiologia , Nucleossomos/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Montagem e Desmontagem da Cromatina , Reparo do DNA , Células HEK293 , Histonas/metabolismo , Humanos , Imuno-Histoquímica/métodos , Camundongos , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Músculos/metabolismo , Proteína MyoD/metabolismo , Miogenina/metabolismo , Regiões Promotoras Genéticas , Transfecção
2.
J Cell Sci ; 125(Pt 6): 1440-54, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22328527

RESUMO

Expression of the paired-box 7 (PAX7) transcription factor is regulated during both myoblast proliferation and differentiation: high levels of PAX7 compromise myogenic differentiation because of excess and prolonged proliferation, whereas low levels of PAX7 result in precocious differentiation. We showed that myogenin repressed Pax7 transcription in differentiating myoblasts by binding to specific recognition sites in the Pax7 promoter, and that high-mobility group box 1 (HMGB1)-receptor for advanced glycation end-products (RAGE) signaling was required for myogenin induction and myogenin-dependent repression of Pax7 transcription. In addition, PAX7 negatively and myogenin positively regulated RAGE expression. RAGE, a multiligand receptor of the immunoglobulin superfamily, was not expressed in adult skeletal muscles, and was transiently expressed in activated, proliferating and differentiating satellite cells (SCs) in injured muscles. Compared with wild-type muscles, Rage(-/-) muscles exhibited increased numbers of basal SCs that were further increased in injured Rage(-/-) muscles following elevated myoblast asymmetric division; complete regeneration of injured Rage(-/-) muscles was found to be delayed by ~1 week. Thus, RAGE signaling physiologically repressed Pax7 transcription in SCs by upregulating myogenin, thereby accelerating muscle regeneration and limiting SC self-renewal.


Assuntos
Proteína HMGB1/fisiologia , Homeostase/fisiologia , Miogenina/fisiologia , Fator de Transcrição PAX7/genética , Receptores Imunológicos/genética , Células Satélites de Músculo Esquelético/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mioblastos/citologia , Mioblastos/metabolismo , Fator de Transcrição PAX7/biossíntese , Cultura Primária de Células , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/deficiência , Elementos Reguladores de Transcrição/fisiologia , Proteínas Repressoras/fisiologia , Células Satélites de Músculo Esquelético/citologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
3.
Biochem Biophys Res Commun ; 450(4): 1631-7, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25044114

RESUMO

MEGF10 is known to function as a myogenic regulator of satellite cells in skeletal muscle. Mutations in MEGF10 gene cause a congenital myopathy called early onset myopathy, areflexia, respiratory distress and dysphagia (EMARDD). Despite its biological importance in muscle physiology, transcriptional regulation of the MEGF10 gene is unknown. Here, we characterized the 5' flanking region of the human MEGF10 gene and showed that the role of myogenic basic helix-loop-helix factor (bHLH) myogenin in MEGF10 transcription in muscle cells. Myogenin was found to share a similar expression pattern with MEGF10 during muscle regeneration and to increase the promoter activity of the MEGF10 gene in C2C12 cells. Overexpression of myogenin led to upregulation of MEGF10 mRNA in C2C12 cells. Site-directed mutagenesis assays revealed that the conserved E-box element at the region -114/-108 serves as a myogenin-binding motif. Promoter enzyme immunoassays and chromatin immunoprecipitation analysis showed direct interaction between myogenin and the myogenin-binding motif in the MEGF10 promoter. Taken together, these results indicate that myogenin is a positive regulator in transcriptional regulation of MEGF10 in skeletal muscle.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/genética , Músculo Esquelético/metabolismo , Miogenina/fisiologia , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Primers do DNA , Humanos , Masculino , Camundongos , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real
4.
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
5.
Proc Natl Acad Sci U S A ; 107(15): 6918-23, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20351273

RESUMO

MyTH/FERM (myosin tail homology 4/band 4.1, ezrin, radixin, and moesin) myosins have roles in cellular adhesion, extension of actin-filled projections such as filopodia and stereocilia, and directional migration. The amoeba Dictyostelium discoideum expresses a simple complement of MyTH/FERM myosins, a class VII (M7) myosin required for cell-substrate adhesion and a unique myosin named MyoG. Mutants lacking MyoG exhibit a wide range of normal actin-based behaviors, including chemotaxis to folic acid, but have a striking defect in polarization and chemotaxis to cAMP. Although the myoG mutants respond to cAMP stimulation by increasing persistence and weakly increasing levels of cortical F-actin, they do not polarize; instead, they maintain a round shape and move slowly and randomly when exposed to a chemotactic gradient. The mutants also fail to activate and localize PI3K to the membrane closest to the source of chemoattractant. These data reveal a role for a MyTH/FERM myosin in mediating early chemotactic signaling and suggest that MyTH/FERM proteins have conserved roles in signaling and the generation of cell polarity.


Assuntos
Dictyostelium/metabolismo , Miogenina/fisiologia , Miosinas/fisiologia , Actinas/química , Actinas/metabolismo , Animais , Movimento Celular , Quimiotaxia , Citoesqueleto/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Modelos Biológicos , Miogenina/genética , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Estrutura Terciária de Proteína , Transdução de Sinais
6.
J Cell Biol ; 177(5): 769-79, 2007 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-17548510

RESUMO

Postnatal growth and regeneration of skeletal muscle requires a population of resident myogenic precursors named satellite cells. The transcription factor Pax7 is critical for satellite cell biogenesis and survival and has been also implicated in satellite cell self-renewal; however, the underlying molecular mechanisms remain unclear. Previously, we showed that Pax7 overexpression in adult primary myoblasts down-regulates MyoD and prevents myogenin induction, inhibiting myogenesis. We show that Pax7 prevents muscle differentiation independently of its transcriptional activity, affecting MyoD function. Conversely, myogenin directly affects Pax7 expression and may be critical for Pax7 down-regulation in differentiating cells. Our results provide evidence for a cross-inhibitory interaction between Pax7 and members of the muscle regulatory factor family. This could represent an additional mechanism for the control of satellite cell fate decisions resulting in proliferation, differentiation, and self-renewal, necessary for skeletal muscle maintenance and repair.


Assuntos
Diferenciação Celular/fisiologia , Proteína MyoD/fisiologia , Fator de Transcrição PAX7/fisiologia , Células Satélites de Músculo Esquelético/citologia , Animais , Linhagem Celular , Proliferação de Células , Regulação da Expressão Gênica , Camundongos , Desenvolvimento Muscular/fisiologia , Proteína MyoD/antagonistas & inibidores , Miogenina/fisiologia , Fator de Transcrição PAX7/antagonistas & inibidores , Fator de Transcrição PAX7/química , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína
7.
Curr Opin Cell Biol ; 5(6): 1057-64, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8129944

RESUMO

Embryological studies have identified axial signalling processes that determine somite cells to skeletal myogenesis and control the spatial patterning of muscle differentiation in vertebrate embryos. Gene knockout studies provide evidence that the muscle-specific myoD genes have essential, although partially redundant functions in vivo as regulators of muscle differentiation. However, continuous cell-cell signalling processes also appear to be required to control and maintain the myogenic potential of embryonic progenitor cells, even after activation of myoD genes. The implications of these findings are discussed in relation to cellular mechanisms of muscle regeneration and the use of myoblast transfer as a muscle regeneration therapy.


Assuntos
Proteínas de Ligação a DNA , Indução Embrionária , Músculos/embriologia , Transativadores , Anfíbios/embriologia , Animais , Cartilagem/citologia , Cartilagem/embriologia , Comunicação Celular , Diferenciação Celular , Embrião de Galinha , Coturnix/embriologia , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Músculos/citologia , Proteína MyoD/genética , Proteína MyoD/fisiologia , Fator Regulador Miogênico 5 , Miogenina/genética , Miogenina/fisiologia , Transdução de Sinais/fisiologia , Pele/citologia , Pele/embriologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
8.
Biochem J ; 428(2): 223-33, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20334626

RESUMO

M- and N-cadherin are members of the Ca(2+)-dependent cell-cell adhesion molecule family. M-cadherin is expressed predominantly in developing skeletal muscles and has been implicated in terminal myogenic differentiation, particularly in myoblast fusion. N-cadherin-mediated cell-cell adhesion also plays an important role in skeletal myogenesis. In the present study, we found that both genes were differentially expressed in C2C12 and Sol8 myoblasts during myogenic differentiation and that the expression of M-cadherin was preferentially enhanced in slow-twitch muscle. Interestingly, most MRFs (myogenic regulatory factors) significantly activated the promoter of M-cadherin, but not that of N-cadherin. In line with this, overexpression of MyoD in C3H10T1/2 fibroblasts strongly induced endogenous M-cadherin expression. Promoter analysis in silico and in vitro identified an E-box (from -2 to +4) abutting the transcription initiation site within the M-cadherin promoter that is bound and differentially activated by different MRFs. The activation of the M-cadherin promoter by MRFs was also modulated by Bhlhe40 (basic helix-loop-helix family member e40). Finally, chromatin immunoprecipitation proved that MyoD as well as myogenin binds to the M-cadherin promoter in vivo. Taken together, these observations identify a molecular mechanism by which MRFs regulate M-cadherin expression directly to ensure the terminal differentiation of myoblasts.


Assuntos
Caderinas/genética , Fatores de Regulação Miogênica/fisiologia , Regiões Promotoras Genéticas/genética , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Elementos E-Box/genética , Ensaio de Desvio de Mobilidade Eletroforética , Fatores de Transcrição MEF2 , Camundongos , Proteína MyoD/genética , Proteína MyoD/metabolismo , Proteína MyoD/fisiologia , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Miogenina/genética , Miogenina/metabolismo , Miogenina/fisiologia , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
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
10.
Dev Biol ; 331(2): 152-66, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19409884

RESUMO

Transcription factor Six1a plays important roles in morphogenesis, organogenesis, and cell differentiation. However, the role of Six1a during zebrafish cranial muscle development is still unclear. Here, we demonstrated that Six1a was required for sternohyoideus, medial rectus, inferior rectus, and all pharyngeal arch muscle development. Although Six1a was also necessary for myod and myogenin expression in head muscles, it did not affect myf5 expression in cranial muscles that originate from head mesoderm. Overexpression of myod enabled embryos to rescue all the defects in cranial muscles induced by injection of six1a-morpholino (MO), suggesting that myod is directly downstream of six1a in controlling craniofacial myogenesis. However, overexpression of six1a was unable to rescue arch muscle defects in the tbx1- and myf5-morphants, suggesting that six1a is only involved in myogenic maintenance, not its initiation, during arch muscle myogenesis. Although the craniofacial muscle defects caused by pax3-MO phenocopied those induced by six1a-MO, injection of six1a, myod or myf5 mRNA did not rescue the cranial muscle defects in pax3 morphants, suggesting that six1a and pax3 do not function in the same regulatory network. Therefore, we proposed four putative regulatory pathways to understand how six1a distinctly interacts with either myf5 or myod during zebrafish craniofacial muscle development.


Assuntos
Músculos Faciais/embriologia , Desenvolvimento Muscular/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Embrião não Mamífero/metabolismo , Músculos Faciais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteína MyoD/fisiologia , Miogenina/fisiologia , Peixe-Zebra/metabolismo
11.
Elife ; 92020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33001028

RESUMO

Growth and maintenance of skeletal muscle fibres depend on coordinated activation and return to quiescence of resident muscle stem cells (MuSCs). The transcription factor Myogenin (Myog) regulates myocyte fusion during development, but its role in adult myogenesis remains unclear. In contrast to mice, myog-/-zebrafish are viable, but have hypotrophic muscles. By isolating adult myofibres with associated MuSCs, we found that myog-/- myofibres have severely reduced nuclear number, but increased myonuclear domain size. Expression of fusogenic genes is decreased, Pax7 upregulated, MuSCs are fivefold more numerous and mis-positioned throughout the length of myog-/-myofibres instead of localising at myofibre ends as in wild-type. Loss of Myog dysregulates mTORC1 signalling, resulting in an 'alerted' state of MuSCs, which display precocious activation and faster cell cycle entry ex vivo, concomitant with myod upregulation. Thus, beyond controlling myocyte fusion, Myog influences the MuSC:niche relationship, demonstrating a multi-level contribution to muscle homeostasis throughout life.


Assuntos
Músculo Esquelético/crescimento & desenvolvimento , Miofibrilas/fisiologia , Miogenina/fisiologia , Células-Tronco/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Técnicas de Inativação de Genes , Homeostase , Músculo Esquelético/metabolismo , Miofibrilas/metabolismo , Miogenina/metabolismo , Células-Tronco/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
12.
Dev Biol ; 322(2): 406-14, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18721801

RESUMO

In contrast to the detailed understanding we have for the regulation of skeletal muscle gene expression in embryos, similar insights into postnatal muscle growth and regeneration are largely inferential or do not directly address gene regulatory mechanisms. Muscle stem cells (satellite cells) are chiefly responsible for providing new muscle during postnatal and adult life. The purpose of this study was to determine the role that the myogenic basic helix-loop-helix regulatory factor myogenin has in postnatal muscle growth and adult muscle stem cell gene expression. We found that myogenin is absolutely required for skeletal muscle development and survival until birth, but it is dispensable for postnatal life. However, Myog deletion after birth led to reduced body size implying a role for myogenin in regulating body homeostasis. Despite a lack of skeletal muscle defects in Myog-deleted mice during postnatal life and the efficient differentiation of cultured Myog-deleted adult muscle stem cells, the loss of myogenin profoundly altered the pattern of gene expression in cultured muscle stem cells and adult skeletal muscle. Remarkably, these changes in gene expression were distinct from those found in Myog-null embryonic skeletal muscle, indicating that myogenin has separate functions during postnatal life.


Assuntos
Células-Tronco Adultas/metabolismo , Diferenciação Celular/fisiologia , Músculo Esquelético/metabolismo , Miogenina/fisiologia , Células-Tronco Adultas/citologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Desenvolvimento Muscular , Músculo Esquelético/embriologia , Músculo Esquelético/crescimento & desenvolvimento , Fatores de Regulação Miogênica/fisiologia , Miogenina/genética , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez
13.
J Cell Biol ; 166(6): 865-76, 2004 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-15364961

RESUMO

To investigate the requirement for pRb in myogenic differentiation, a floxed Rb allele was deleted either in proliferating myoblasts or after differentiation. Myf5-Cre mice, lacking pRb in myoblasts, died immediately at birth and exhibited high numbers of apoptotic nuclei and an almost complete absence of myofibers. In contrast, MCK-Cre mice, lacking pRb in differentiated fibers, were viable and exhibited a normal muscle phenotype and ability to regenerate. Induction of differentiation of Rb-deficient primary myoblasts resulted in high rates of apoptosis and a total inability to form multinucleated myotubes. Upon induction of differentiation, Rb-deficient myoblasts up-regulated myogenin, an immediate early marker of differentiation, but failed to down-regulate Pax7 and exhibited growth in low serum conditions. Primary myoblasts in which Rb was deleted after expression of differentiated MCK-Cre formed normal multinucleated myotubes that did not enter S-phase in response to serum stimulation. Therefore, Rb plays a crucial role in the switch from proliferation to differentiation rather than maintenance of the terminally differentiated state.


Assuntos
Diferenciação Celular , Fibras Musculares Esqueléticas/fisiologia , Mioblastos/fisiologia , Proteína do Retinoblastoma/fisiologia , Adenoviridae/genética , Alelos , Animais , Apoptose/genética , Divisão Celular , Células Cultivadas , Meios de Cultura Livres de Soro , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Camundongos Transgênicos , Músculo Esquelético/citologia , Mioblastos/citologia , Miogenina/fisiologia , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Regulação para Cima
14.
J Cell Biol ; 123(2): 467-75, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8408226

RESUMO

Human carcinoembryonic antigen (CEA), a widely used tumor marker, is a member of a family of cell surface glycoproteins that are overexpressed in many carcinomas. CEA has been shown to function in vitro as a homotypic intercellular adhesion molecule. This correlation of overproduction of an adhesion molecule with neoplastic transformation provoked a test of the effect of CEA on cell differentiation. Using stable CEA transfectants of the rat L6 myoblast cell line as a model system of differentiation, we show that fusion into myotubes and, in fact, the entire molecular program of differentiation, including creatine phosphokinase upregulation, myogenin upregulation, and beta-actin downregulation are completely abrogated by the ectopic expression of CEA. The blocking of the upregulation of myogenin, a transcriptional regulator responsible for the execution of the entire myogenic differentiation program, indicates that CEA expression intercepts the process at a very early stage. The adhesion function of CEA is essential for this effect since an adhesion-defective N domain deletion mutant of CEA was ineffective in blocking fusion and CEA transfectants treated with adhesion-blocking peptides fused normally. Furthermore, CEA transfectants maintain their high division potential, whereas control transfectants lose division potential with differentiation similarly to the parental cell line. Thus the expression of functional CEA on the surface of cells can block terminal differentiation and maintain proliferative potential.


Assuntos
Antígeno Carcinoembrionário/fisiologia , Músculos/citologia , Músculos/embriologia , Actinas/análise , Actinas/genética , Actinas/fisiologia , Animais , Sequência de Bases , Northern Blotting , Antígeno Carcinoembrionário/análise , Antígeno Carcinoembrionário/genética , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Fusão Celular/fisiologia , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Creatina Quinase/análise , Creatina Quinase/genética , Creatina Quinase/fisiologia , DNA/genética , Regulação para Baixo , Modelos Biológicos , Dados de Sequência Molecular , Músculos/química , Miogenina/análise , Miogenina/genética , Miogenina/fisiologia , Ratos , Transfecção , Regulação para Cima
15.
Science ; 267(5200): 1024-7, 1995 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-7863329

RESUMO

Terminal differentiation is coupled to withdrawal from the cell cycle. The cyclin-dependent kinase inhibitor (CKI) p21Cip1 is transcriptionally regulated by p53 and can induce growth arrest. CKIs are therefore potential mediators of developmental control of cell proliferation. The expression pattern of mouse p21 correlated with terminal differentiation of multiple cell lineages including skeletal muscle, cartilage, skin, and nasal epithelium in a p53-independent manner. Although the muscle-specific transcription factor MyoD is sufficient to activate p21 expression in 10T1/2 cells, p21 was expressed in myogenic cells of mice lacking the genes encoding MyoD and myogenin, demonstrating that p21 expression does not require these transcription factors. The p21 protein may function during development as an inducible growth inhibitor that contributes to cell cycle exit and differentiation.


Assuntos
Diferenciação Celular , Ciclinas/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/citologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Ciclo Celular , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Embrião de Mamíferos/metabolismo , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Proteína MyoD/genética , Proteína MyoD/fisiologia , Miogenina/genética , Miogenina/fisiologia
16.
Endocrinology ; 148(3): 1108-17, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17122077

RESUMO

Our main goal in this study was to investigate the role of phospholipase C (PLC) beta(1) and PLCgamma(1) in skeletal muscle differentiation and the existence of potential downstream targets of their signaling activity. To examine whether PLC signaling can modulate the expression of cyclin D3, a target of PLCbeta(1) in erythroleukemia cells, we transfected C2C12 cells with expression vectors containing PLCbeta(1) or PLCgamma(1) cDNA and with small interfering RNAs from regions of the PLCbeta(1) or PLCgamma(1) gene and followed myogenic differentiation in this well-established cell system. Intriguingly, overexpressed PLCbeta(1) and PLCgamma(1) were able to mimic insulin induction of both cyclin D3 and muscle differentiation. By knocking down PLCbeta(1) or PLCgamma(1) expression, C2C12 cells almost completely lost the increase in cyclin D3, and the differentiation program was down-regulated. To explore the induction of the cyclin D3 gene promoter during this process, we used a series of 5'-deletions of the 1.68-kb promoter linked to a reporter gene and noted a 5-fold augmentation of promoter activity upon insulin stimulation. These constructs were also cotransfected with PLCbeta(1) or PLCgamma(1) cDNAs and small interfering RNAs, respectively. Our data indicate that PLCbeta(1) or PLCgamma(1) signaling is capable of acting like insulin in regard to both the myogenic differentiation program and cyclin D3 up-regulation. Taken together, this is the first study that hints at cyclin D3 as a target of PLCbeta(1) and PLCgamma(1) during myogenic differentiation in vitro and implies that up-regulation of these enzymes is sufficient to mimic the actions of insulin in this process.


Assuntos
Ciclinas/genética , Insulina/fisiologia , Isoenzimas/fisiologia , Desenvolvimento Muscular , Músculo Esquelético/enzimologia , Fosfolipases Tipo C/fisiologia , Animais , Células Cultivadas , Ciclina D3 , Ciclinas/metabolismo , Ciclinas/fisiologia , Imuno-Histoquímica , Isoenzimas/metabolismo , Camundongos , Desenvolvimento Muscular/efeitos dos fármacos , Miogenina/fisiologia , Fosfolipase C beta , Fosfolipase C gama/metabolismo , Fosfolipase C gama/fisiologia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Fosfolipases Tipo C/metabolismo
17.
Curr Biol ; 4(1): 61-3, 1994 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-7922315
18.
Mol Cell Biol ; 14(8): 5474-86, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8035824

RESUMO

Members of the MyoD family of gene-regulatory proteins (MyoD, myogenin, myf5, and MRF4) have all been shown not only to regulate the transcription of numerous muscle-specific genes but also to positively autoregulate and cross activate each other's transcription. In the case of muscle-specific genes, this transcriptional regulation can often be correlated with the presence of a DNA consensus in the regulatory region CANNTG, known as an E box. Little is known about the regulatory interactions of the myogenic factors themselves; however, these interactions are thought to be important for the activation and maintenance of the muscle phenotype. We have identified the minimal region in the chicken MyoD (CMD1) promoter necessary for muscle-specific transcription in primary cultures of embryonic chicken skeletal muscle. The CMD1 promoter is silent in primary chick fibroblast cultures and in muscle cell cultures treated with the thymidine analog bromodeoxyuridine. However, CMD1 and chicken myogenin, as well as, to a lesser degree, chicken Myf5 and MRF4, expressed in trans can activate transcription from the minimal CMD1 promoter in these primary fibroblast cultures. Here we show that the CMD1 promoter contains numerous E-box binding sites for CMD1 and the other myogenic factors, as well as a MEF-2 binding site. Surprisingly, neither muscle-specific and the other myogenic factors, as well as a MEF-2 binding site. Surprisingly, neither muscle-specific expression, autoregulation, or cross activation depends upon the presence of of these E-box or MEF-2 binding sites in the CMD1 promoter. These results demonstrate that the autoregulation and cross activation of the chicken MyoD promoter through the putative direct binding of the myogenic basic helix-loop-helix regulatory factors is mediated through an indirect pathway that involves unidentified regulatory elements and/or ancillary factors.


Assuntos
Regulação da Expressão Gênica , Músculos/fisiologia , Proteína MyoD/genética , Regiões Promotoras Genéticas , Transativadores , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Galinhas , Clonagem Molecular , Proteínas de Ligação a DNA/metabolismo , Genes , Sequências Hélice-Alça-Hélice , Fatores de Transcrição MEF2 , Dados de Sequência Molecular , Proteínas Musculares/fisiologia , Fator Regulador Miogênico 5 , Fatores de Regulação Miogênica/fisiologia , Miogenina/fisiologia , Fatores de Transcrição/metabolismo
19.
Mol Cell Biol ; 16(8): 4014-23, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8754799

RESUMO

The alternative exon 5 of the striated muscle-specific cardiac troponin T (cTNT) gene is included in mRNA from embryonic skeletal and cardiac muscle and excluded in mRNA from the adult. The embryonic splicing pattern is reproduced in primary skeletal muscle cultures for both the endogenous gene and transiently transfected minigenes, whereas in nonmuscle cell lines, minigenes express a default exon skipping pattern. Using this experimental system, we previously showed that a purine-rich splicing enhancer in the alternative exon functions as a constitutive splicing element but not as a target for factors regulating cell-specific splicing. In this study, we identify four intron elements, one located upstream,and three located downstream of the alternative exon, which act in a positive manner to mediate the embryonic splicing pattern of exon inclusion. Synergistic interactions between at least three of the four elements are necessary and sufficient to regulate splicing of a heterologous alternative exon and heterologous splice sites. Mutations in these elements prevent activation of exon inclusion in muscle cells but do not affect the default level of exon inclusion in nonmuscle cells. Therefore, these elements function as muscle-specific splicing enhancers (MSEs) and are the first muscle-specific positive-acting splicing elements to be described. One MSE located downstream from the alternative exon is conserved in the rat and chicken cTNT genes. A related sequence is found in a third muscle-specific gene, that encoding skeletal troponin T, downstream from an alternative exon with a developmental pattern of alternative splicing similar to that of rat and chicken cTNT. Therefore, the MSEs identified in the cTNT gene may play a role in developmentally regulated alternative splicing in a number of different genes.


Assuntos
Processamento Alternativo , Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/fisiologia , Troponina/genética , Animais , Sequência de Bases , Células Cultivadas , Galinhas , Coturnix , Primers do DNA/química , Éxons , Íntrons , Dados de Sequência Molecular , Músculo Esquelético/embriologia , Proteína MyoD/fisiologia , Miogenina/fisiologia , RNA Mensageiro/genética , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Troponina T
20.
Endocrinology ; 147(6): 3093-106, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16527841

RESUMO

Peroxisomal proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha), a transcriptional coactivator, is selectively expressed in slow-twitch fibers in skeletal muscle. Ectopic expression of the PGC-1alpha gene in either a cell or an animal has been shown to promote fast to slow fiber-type switch. The expression of PGC-1alpha in muscle is regulated by myocyte enhancer factor 2 and Forkhead in rhabdomyosarcoma, two transcription factors implicated in terminal muscle differentiation. In this study we found that PGC-1alpha expression was activated during terminal muscle differentiation in both C2C12 and Sol8 myoblasts. Using retrovirus-mediated MyoD overexpression in C3H10T1/2 cells, we also demonstrated that MyoD, the master regulator of terminal differentiation, could activate PGC-1alpha expression in vivo. Our transient transfection results also show that myogenic basic helix-loop-helix (bHLH) proteins, especially MyoD, can activate PGC-1alpha expression by targeting its promoter. Myogenic bHLH protein target sites on PGC-1alpha promoter were localized to a short region (-49 to approximately +2) adjacent to the transcription start site, which contains two putative E boxes. Mutation of either site significantly reduced MyoD-mediated transactivation in the cells, suggesting that both sites are required for myogenic bHLH protein-mediated activation. However, only one site, the E2 box, was directly bound by glutathione-S-transferase-MyoD protein in EMSAs. Our results indicate that myogenic bHLH proteins not only are involved in lineage determination and terminal differentiation, but also are directly implicated in activation of the key fiber-type and metabolic switch gene, PGC-1alpha.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Diferenciação Celular , Regulação da Expressão Gênica , Proteínas de Choque Térmico/genética , Mioblastos Esqueléticos/citologia , Fatores de Transcrição/genética , Animais , Sequência de Bases , Linhagem da Célula , Células Cultivadas , Proteínas de Choque Térmico/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Proteína MyoD/fisiologia , Fatores de Regulação Miogênica/metabolismo , Miogenina/fisiologia , Proteínas Nucleares/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Regiões Promotoras Genéticas , Fatores de Transcrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA