Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Intervalo de ano de publicação
2.
Artigo em Inglês | MEDLINE | ID: mdl-19022756

RESUMO

Pax genes have important roles in the regulation of stem cell behavior, leading to tissue differentiation. In the case of skeletal muscle, Pax3 and Pax7 perform this function both during development and on regeneration in the adult. The myogenic determination gene Myf5 is directly activated by Pax3, leading to the formation of skeletal muscle. Fgfr4 is also a direct Pax3 target and Sprouty1, which encodes an intracellular inhibitor of fibroblast growth factor (FGF) signaling, is under Pax3 control. Orchestration of FGF signaling, through Fgfr4/Sprouty1, modulates the entry of cells into the myogenic program, thus controling the balance between stem cell self-renewal and tissue differentiation. This and other aspects of Pax3/7 function in regulating the behavior of skeletal muscle stem cells are discussed.


Assuntos
Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Fator de Transcrição PAX7/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Desenvolvimento Muscular , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/metabolismo , Fator de Transcrição PAX3 , Fator de Transcrição PAX7/genética , Fatores de Transcrição Box Pareados/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
4.
J Biol Chem ; 276(50): 46961-7, 2001 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-11598109

RESUMO

Transforming growth factor-beta (TGF-beta) and insulin-like growth factors (IGFs) play critical roles in the control of myogenesis. Insulin-like growth factor-binding protein-5 (IGFBP-5), by regulating the bioavailability of IGFs, is involved in controlling IGF-dependent differentiation. We investigated the effects of TGF-beta on the IGFBP-5 production induced by IGFs in mouse myoblasts. TGF-beta leads to a decrease in IGFBP-5 synthesis at both transcript and protein levels, and blocked muscle differentiation. The Smad proteins and the c-Jun N-terminal kinase (JNK) have been shown to be involved in TGF-beta signaling pathways. We provide evidence that the JNK pathway, rather than Smad proteins, is involved in the response of muscle cells to TGF-beta. This factor failed to stimulate the GAL4-Smad 2/3 transcriptional activities of the constructs used to transfect myoblasts. Moreover, stable expression of the antagonistic Smad7 did not abolish the inhibitory effect of TGF-beta on IGFBP-5 production whereas expression of a dominant-negative version of MKK4, an upstream activator of JNK, did. We also showed, using a specific inhibitor, that the p38 mitogen-activated protein kinase (p38 MAPK) was not involved in the inhibition of IGFBP-5 production. Thus, TGF-beta-mediated IGFBP-5 inhibition is independent of Smads and requires activation of the JNK signaling pathway.


Assuntos
Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , MAP Quinase Quinase 4 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Esquelético/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose , Northern Blotting , Diferenciação Celular , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Genes Dominantes , Insulina/metabolismo , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno , Luciferases/metabolismo , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Plasmídeos/metabolismo , Ligação Proteica , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína Smad2 , Proteína Smad3 , Proteína Smad7 , Transativadores/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção , Troponina T/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
5.
FEBS Lett ; 488(3): 174-8, 2001 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-11163767

RESUMO

We have derived skeletal muscle cell lines from wild-type (wt) and insulin receptor (IR) knockout mice to unravel the metabolic potential of IGF-1 receptor (IGF-1R). Both wt and IR(-/-) myoblasts differentiated into myotubes with similar patterns of expression of muscle-specific genes such as MyoD, myogenin and MLC1A indicating that IR is not required for this process. Binding of 125I-IGF-1 on wt and IR(-/-) myotubes was similar showing that IGF-1R was not upregulated in the absence of IR. Stimulation of IR(-/-) myotubes with IGF-1 (10(-10) to 10(-7) M) increased glucose uptake and incorporation into glycogen, induced IRS-1 phosphorylation and activated PI 3-kinase and MAP kinase, two enzymes of major signaling pathways. These effects were comparable to those obtained with wt myotubes using insulin or IGF-1 or with IR(-/-) myotubes using insulin at higher concentrations. This study provides a direct evidence that IGF-1R can represent an alternative receptor for metabolic signaling in muscle cells.


Assuntos
Deleção de Genes , Músculo Esquelético/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/deficiência , Animais , Animais Recém-Nascidos , Sítios de Ligação , Biomarcadores , Células Cultivadas , Meios de Cultura Livres de Soro , Desoxiglucose/metabolismo , Glucose/metabolismo , Glicogênio/metabolismo , Insulina/farmacologia , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , RNA Mensageiro/análise , RNA Mensageiro/genética , Receptor de Insulina/genética , Transdução de Sinais/efeitos dos fármacos
6.
Dev Biol ; 240(2): 574-84, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11784084

RESUMO

Transcription factors Myf5 and MyoD are critical for myoblast determination. Myogenin is a direct transcriptional target of these factors and its expression is associated with commitment to terminal differentiation. Here, we have used myogenic derivatives of human U20S cells expressing Myf5 or MyoD under control of a tetracycline-sensitive promoter to study expression of endogenous myogenin (myf4). We find that Myf5-mediated induction of myogenin shows striking dependence on cell density. At high cell density, Myf5 is a potent inducer of myogenin expression. At low cell density, Myf5 (unlike MyoD) is a poor inducer of myogenin expression, whilst retaining the capacity to direct expression of other muscle-specific genes. The permissive influence of high cell density on myogenin induction by Myf5 is not a consequence of serum depletion or cell cycle arrest, but is mimicked by a disruption adjacent to the basic region of Myf5 (Myf5/mt) which reduces its DNA binding affinity for E-boxes without compromising its ability to transactivate a reporter gene driven by the myogenin promoter. Coculture of cells expressing wild-type Myf5 and Myf5/mt leads to reduced myogenin induction in Myf5/mt cells. We propose that at low cell density Myf5 inhibits induction of myogenin.


Assuntos
Proteínas de Ligação a DNA , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Fatores de Regulação Miogênica/biossíntese , Fatores de Regulação Miogênica/genética , Miogenina/biossíntese , Miogenina/genética , Animais , Contagem de Células , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Genes Reporter , Humanos , Camundongos , Fator Regulador Miogênico 5 , Transativadores/genética , Transativadores/metabolismo , Transfecção
7.
Mol Cell Biol ; 20(23): 8923-32, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11073992

RESUMO

Transcription factors Myf5 and MyoD play critical roles in controlling myoblast identity and differentiation. In the myogenic cell line C2, we have found that Myf5 expression, unlike that of MyoD, is restricted to cycling cells and regulated by proteolysis at mitosis. In the present study, we have examined Myf5 proteolysis through stable transfection of myogenically convertible U20S cells with Myf5 derivatives under the control of a tetracycline-sensitive promoter. A motif within the basic helix-loop-helix domain of Myf5 (R93 to Q101) resembles the "destruction box" characteristic of substrates of mitotic proteolysis and thought to be recognized by the anaphase-promoting complex or cyclosome (APC). Mutation of this motif in Myf5 stabilizes the protein at mitosis but does not affect its constitutive turnover. Conversely, mutation of a serine residue (S158) stabilizes Myf5 in nonsynchronized cultures but not at mitosis. Thus, at least two proteolytic pathways control Myf5 levels in cycling cells. The mitotic proteolysis of Myf5 is unlike that which has been described for other destruction box-dependent substrates: down-regulation of Myf5 at mitosis appears to precede that of known targets of the APC and is not affected by a dominant-negative version of the ubiquitin carrier protein UbcH10, implicated in the APC-mediated pathway. Finally, we find that induction of Myf5 perturbs the passage of cells through mitosis, suggesting that regulation of Myf5 levels at mitosis may influence cell cycle progression of Myf5-expressing muscle precursor cells.


Assuntos
Proteínas de Ligação a DNA , Sequências Hélice-Alça-Hélice , Mitose/fisiologia , Proteínas Musculares/metabolismo , Músculos/citologia , Fatores de Regulação Miogênica/metabolismo , Transativadores , Complexos Ubiquitina-Proteína Ligase , Ciclossomo-Complexo Promotor de Anáfase , Diferenciação Celular , Ligases , Fator Regulador Miogênico 5 , Células-Tronco , Ubiquitina-Proteína Ligases
8.
Biol Cell ; 92(8-9): 565-72, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11374435

RESUMO

Myf-5 and MyoD are the two muscle regulatory factors expressed from the myoblast stage to maintain the identity and to promote the subsequent differentiation of muscle precursor cells. To get insight into their role we have studied the capacity to proliferate and to differentiate of myf-5 and myoD null myoblasts in primary cultures and in the subsequent passages. Our results indicate that myf-5 null myoblasts differ from wild type (wt) myoblasts in that they undergo precocious differentiation: they become myogenin- and troponin T-positive and fail to incorporate bromodeoxyuridine (BrdU) under culture conditions and at a time when wt cells are not yet differentiated and continue to proliferate. In primary cultures of myoD null cells, up to 60% of the cells were scored as myoblasts on the basis of the expression of myf-5. These myoD-deficient myoblasts, unlike myoD-expressing cells, were poorly differentiating and displayed a severe growth defect that led to their elimination from the cultures: within a few passages myoblasts were absent from myoD-deficient cultures, which mostly consisted of senescent cells. That a null mutation in either gene reduces the proliferative potential of cultured myoblasts raises the possibility that Myf-5 and MyoD serve proliferation of muscle precursor cells.


Assuntos
Diferenciação Celular/genética , Divisão Celular/genética , Linhagem da Célula/genética , Proteínas de Ligação a DNA , Proteínas Musculares/deficiência , Músculo Esquelético/embriologia , Proteína MyoD/metabolismo , Células-Tronco/metabolismo , Transativadores , Fatores Etários , Animais , Técnicas de Cultura de Células , Tamanho Celular/genética , Células Cultivadas/citologia , Células Cultivadas/metabolismo , Senescência Celular/fisiologia , Genes Reporter/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Proteína MyoD/genética , Fator Regulador Miogênico 5 , Miogenina/metabolismo , Fenótipo , Regeneração/genética , Células-Tronco/citologia , Troponina T/metabolismo
9.
Oncogene ; 18(36): 5063-75, 1999 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-10490843

RESUMO

One of the most frequent genetic abnormalities associated with non Hodgkin lymphoma is the structural alteration of the 5' non coding/regulatory region of the BCL6 (LAZ3) protooncogene. BCL6 encodes a POZ/Zn finger protein, a structure similar to that of many Drosophila developmental regulators and to another protein involved in a human hematopoietic malignancy, PLZF. BCL6 is a sequence specific transcriptional repressor controlling germinal center formation and T cell dependent immune response. Although the expression of BCL6 negatively correlates with cellular proliferation in different cell types, the influence of BCL6 on cell growth and survival is currently unknown so that the way its deregulation may contribute to cancer remains elusive. To directly address this issue, we used a tetracycline-regulated system in human U2OS osteosarcoma cells and thus found that BCL6 mediates growth suppression associated with impaired S phase progression and apoptosis. Interestingly, overexpressed BCL6 can colocalize with sites of ongoing DNA synthesis, suggesting that it may directly interfere with S phase initiation and/or progression. In contrast, the isolated Zn finger region of BCL6, which binds BCL6 target sequence but lacks transcriptional repression activity, slows, but does not suppress, U2OS cell growth, is less efficient at delaying S phase progression, and does not trigger apoptosis. Thus, for a large part, the effects of BCL6 overexpression on cell growth and survival depend on its ability to engage protein/protein interactions with itself and/or its transcriptional corepressors. That BCL6 restricts cell growth suggests that its deregulation upon structural alterations may alleviate negative controls on the cell cycle and cell survival.


Assuntos
Apoptose/fisiologia , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fase S/fisiologia , Fatores de Transcrição/fisiologia , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Humanos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-6 , Fatores de Transcrição/genética , Células Tumorais Cultivadas
10.
Mol Cell Biol ; 19(6): 4047-55, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10330145

RESUMO

Calpain 3 is a nonlysosomal cysteine protease whose biological functions remain unknown. We previously demonstrated that this protease is altered in limb girdle muscular dystrophy type 2A patients. Preliminary observations suggested that its gene is subjected to alternative splicing. In this paper, we characterize transcriptional and posttranscriptional events leading to alterations involving the NS, IS1, and IS2 regions and/or the calcium binding domains of the mouse calpain 3 gene (capn3). These events can be divided into three groups: (i) splicing of exons that preserve the translation frame, (ii) inclusion of two distinct intronic sequences between exons 16 and 17 that disrupt the frame and would lead, if translated, to a truncated protein lacking domain IV, and (iii) use of an alternative first exon specific to lens tissue. In addition, expression of these isoforms seems to be regulated. Investigation of the proteolytic activities and titin binding abilities of the translation products of some of these isoforms clearly indicated that removal of these different protein segments affects differentially the biochemical properties examined. In particular, removal of exon 6 impaired the autolytic but not fodrinolytic activity and loss of exon 16 led to an increased titin binding and a loss of fodrinolytic activity. These results are likely to impact our understanding of the pathophysiology of calpainopathies and the development of therapeutic strategies.


Assuntos
Calpaína/genética , Calpaína/metabolismo , Isoenzimas , Processamento Pós-Transcricional do RNA , Transcrição Gênica , Processamento Alternativo , Animais , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Células Cultivadas , Clonagem Molecular , Conectina , Primers do DNA , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/metabolismo , Humanos , Hibridização In Situ , Íntrons , Cristalino/anatomia & histologia , Cristalino/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/metabolismo , Modelos Genéticos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Liso/metabolismo , Miocárdio/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Quinases/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Distribuição Tecidual
11.
Hum Mol Genet ; 7(13): 2121-8, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9817930

RESUMO

The fragile X syndrome results from transcriptional silencing of the FMR1 gene and the absence of its encoded FMRP protein. Two autosomal homologues of the FMR1 gene, FXR1 and FXR2, have been identified and the overall structures of the corresponding proteins are very similar to that of FMRP. Using antibodies raised against FXR1P, we observed that two major protein isoforms of relative MW of 78 and 70 kDa are expressed in different mammalian cell lines and in the majority of mouse tissues. In mammalian cells grown in culture as well as in brain extracts, both P78and P70isoforms are associated with mRNPs within translating polyribosomes, similarly to their closely related FMRP homologues. In muscle tissues as well as in murine myoblastic cell lines induced to differentiate into myotubes, FXR1P78and P70isoforms are replaced by novel unpredicted isoforms of 81-84 kDa and a novel FXR1 exon splice variant was detected in muscle RNA. While P81-84isoforms expressed after fusion into myotubes in murine myoblast cell lines grown in culture are associated with polyribosomes, this is not the case when isolated from muscle tissues since they sediment with lower S values. Immunohistochemical studies showed coexpression of FMRP and FXR1P70and P78in the cytoplasm of brain neurons, while in muscle no FMRP was detected and FXR1P81-84were mainly localized to structures within the muscle contractile bands. The complex expression pattern of FXR1P suggests tissue-specific expression for the various isoforms of FXR1 and the differential expression of FMRP and FXR1Ps suggests that in certain types of cells and tissues, complementary functions may be fulfilled by the various FMRP family members.


Assuntos
Músculos/metabolismo , Proteínas de Ligação a RNA/genética , Células 3T3 , Processamento Alternativo , Animais , Sequência de Bases , Encéfalo/metabolismo , Células COS , Linhagem Celular , Fracionamento Químico , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células HeLa , Humanos , Immunoblotting , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Desenvolvimento Muscular , Músculos/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Polirribossomos/metabolismo , RNA/genética , Proteínas de Ligação a RNA/análise , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Homologia de Sequência de Aminoácidos
13.
Endocrinology ; 139(4): 1487-93, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528925

RESUMO

Skeletal myoblast differentiation is stimulated by insulin-like growth factors (IGFs). The autocrine action of IGFs is mediated through the type-1 IGF receptor (IGFR-1) and modulated by IGF binding proteins (IGFBPs) secreted by the cells. The mouse C2 myoblast cell line stably transfected with a vector producing IGF-II antisense RNA was used to show that specific IGFBP expression changes with the state of the cells: high levels of IGFBP-2 messenger RNA (mRNA) were found only in proliferating myoblasts, whereas IGFBP-3 mRNA was induced in quiescent cells. Secretion of IGFBP5 was strongly stimulated during differentiation. Insulin and IGF dose-response experiments showed that up-regulation of IGFBP-5 resulted from IGFR-1 activation. Drugs interfering with IGFR-1 signaling and inhibiting myoblast differentiation had different effects on IGFBP-5 up-regulation. Two phosphatidylinositol 3-kinase (PI 3-kinase) inhibitors, wortmaninn and LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one], failed to alter IGFBP-5 up-regulation, which persisted in the absence of differentiation. Rapamycin which indirectly prevents activation of the p70 ribosomal protein-S6 kinase (p70S6k), suppressed IGFBP-5 induction. Because the PI3-kinase inhibitors block p70S6k, neither kinase would be required for IGFR-1-dependent IGFBP-5 induction. In C2 anti-IGF-II myoblasts, IGFBP-5 induction is therefore rapamycin-sensitive and independent of differentiation.


Assuntos
Androstadienos/farmacologia , Diferenciação Celular , Cromonas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Morfolinas/farmacologia , Músculos/citologia , Polienos/farmacologia , Animais , Divisão Celular , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Insulina/farmacologia , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Camundongos , Inibidores de Fosfoinositídeo-3 Quinase , Biossíntese de Proteínas , Receptor IGF Tipo 1/fisiologia , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Sirolimo , Transfecção , Wortmanina
14.
J Cell Biol ; 140(1): 111-8, 1998 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-9425159

RESUMO

Myf5 is the earliest-known muscle-specific factor to be expressed in vivo and its expression is associated with determination of the myoblast lineage. In C2 cells, we show by immunocytolocalization that Myf5 disappears rapidly from cells in which the differentiation program has been initiated. In proliferating myoblasts, the levels of Myf5 and MyoD detected from cell to cell are very heterogeneous. We find that some of the heterogeneity of Myf5 expression arises from a posttranscriptional regulation of Myf5 by the cell cycle. Immunoblotting of extracts from synchronized cultures reveals that Myf5 undergoes periodic fluctuations during the cell cycle and is absent from cells blocked early in mitosis by use of nocodazole. The disappearance of Myf5 from mitotic cells involves proteolytic degradation of a phosphorylated form of Myf5 specific to this phase of the cell cycle. In contrast, MyoD levels are not depleted in mitotic C2 cells. The mitotic destruction of Myf5 is the first example of a transcription factor showing cell cycle-regulated degradation. These results may be significant in view of the possible role of Myf5 in maintaining the determination of proliferating cells and in timing the onset of differentiation.


Assuntos
Ciclo Celular/fisiologia , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Proteínas Musculares/biossíntese , Músculo Esquelético/citologia , Transativadores , Animais , Diferenciação Celular , Divisão Celular , Linhagem Celular , Técnica Indireta de Fluorescência para Anticorpo , Camundongos , Mitose , Músculo Esquelético/metabolismo , Fator Regulador Miogênico 5 , Fatores de Transcrição/biossíntese
15.
Differentiation ; 64(1): 33-44, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9921651

RESUMO

The structural alterations of the LAZ3 (BCL6) gene are one of the most frequent events found in non-Hodgkin lymphoma. LAZ3 encodes a transcriptional repressor with a POZ/zinc finger structure similar to several Drosophila development regulators and to the human promyelocytic leukemia-associated PLZF gene. Consistent with the origin of LAZ3-associated malignancies, LAZ3 is expressed in mature B-cells and required for germinal center formation. However, its ubiquitous expression, with predominant levels in skeletal muscle, suggests that it may act outside the lymphoid system. To study how LAZ3 could be involved in skeletal muscle differentiation, we examined its expression in the C2 muscle cells. We report here that LAZ3 is upregulated at both mRNA and protein levels during the differentiation of proliferating C2 myoblasts into post-mitotic myotubes. This rise in LAZ3 expression is both precocious and sustained, and is not reversed when myotubes are re-exposed to mitogen-rich medium, suggesting that irreversible evens occurring upon myogenic terminal differentiation contribute to lock LAZ3 upregulation. In addition, using two different models, we found that a "simple" growth-arrest upon serum starvation is not sufficient to induce LAZ3 upregulation which rather appears as a feature of myogenic commitment and/or differentiation. Finally, BrdU incorporation assays in C2 cells entering the differentiation pathway indicate that "high" LAZ3 expression strongly correlates with their exit from the cell cycle. Taken as a whole, these findings suggest that LAZ3 could play a role in muscle differentiation. Together with some results reported in other cell types, we propose that LAZ3 may contribute to events common to various differentiation processes, possibly the induction and stabilization of the withdrawal from the cell cycle.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/citologia , Proteínas Proto-Oncogênicas/biossíntese , Proto-Oncogenes , Fatores de Transcrição/biossíntese , Dedos de Zinco/genética , Animais , Diferenciação Celular , Divisão Celular , Células Cultivadas , Meios de Cultivo Condicionados , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Camundongos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-6 , RNA Mensageiro/biossíntese , Fatores de Transcrição/genética
16.
Cell Mol Biol (Noisy-le-grand) ; 43(6): 881-8, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9359635

RESUMO

Myotonic dystrophy (DM) is an autosomal dominant neuromuscular disorder characterized by a great variability in its clinical manifestations. The mutational basis underlying DM consists of an unstable (CTG)n trinucleotide repeat in the 3' untranslated region of the myotonic dystrophy protein kinase gene (DMPK). Conflicting results on DMPK gene expression in congenitally affected infants (CDM) have been published. Moreover, the prominence of satellite cells seen in muscle of CDM infants supports the notion that the congenital form is associated with an arrest in muscle development and suggests a role for the DMPK gene during differentiation and maturation of muscle. In order to clarify these findings, a comparative study of DMPK and myogenic factor mRNA levels was performed in developing mouse muscle tissues and cultured muscle cells at different developmental stages. Results show that DMPK gene expression is upregulated at a late stage of muscular development. This upregulation does not seem to depend on a given muscle specific bHLH factor.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Desenvolvimento Muscular , Músculo Esquelético/enzimologia , Músculo Esquelético/crescimento & desenvolvimento , Distrofia Miotônica/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Linhagem Celular , Embrião de Mamíferos , Camundongos , Camundongos Endogâmicos C3H , Músculo Esquelético/citologia , Distrofia Miotônica/enzimologia , Miotonina Proteína Quinase
17.
Exp Cell Res ; 233(1): 99-117, 1997 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-9184080

RESUMO

Prosomes constitute the multicatalytic proteinase (MCP) core of the 26S proteasomes, but were first observed as subcomplexes of untranslated mRNP; this suggests that they play a putative role in the control of protein biosynthesis in addition to their catabolic enzymatic function. In previous investigations it was shown that some prosomes colocalize with the intermediate filaments (IF) of the cytoskeleton, of the cytokeratin type in epithelial cells, and of the vimentin type in fibroblasts. Studies on adult rat muscle carried out with prosome-specific monoclonal antibodies (p-mAbs) have shown, surprisingly, that specific types of prosomes predominantly occupy a particular zone in between the M and the Z lines of the sarcomeric structure. The data presented here show that the subunit composition of prosomes changes when the dividing C2.7 myoblasts fuse into myotubes. We show furthermore that, in dividing C2.7 myoblasts, prosomes colocalize with the desmin network as well as with that of actin, in a distribution that changes with the subunit pattern of the prosomes investigated by individual p-mAbs. Surprisingly, when myogenic fusion is induced, specific types of prosomes move first to the nuclei; later on, they reappear in the cytoplasm. There, superimposing initially onto the reorganizing desmin filaments that run from one pole of the prefusion myoblast to the other, prosomes gradually colocalize with the actin fibers in the fusing myotubes, finally forming a "pearl on a string" pattern. These results are discussed in relation to parallel observations of prosome distribution between the actin and IF networks not only in epithelial cells but also in fusing muscle satellite cells, which made it possible to monitor the complete buildup of the sarcomeric structure.


Assuntos
Actinas/ultraestrutura , Cisteína Endopeptidases/ultraestrutura , Desmina/ultraestrutura , Filamentos Intermediários/ultraestrutura , Complexos Multienzimáticos/ultraestrutura , Músculos/ultraestrutura , Actinas/isolamento & purificação , Animais , Compartimento Celular , Diferenciação Celular , Fusão Celular , Células Cultivadas , Cisteína Endopeptidases/química , Desmina/isolamento & purificação , Técnica Indireta de Fluorescência para Anticorpo , Filamentos Intermediários/química , Camundongos , Complexos Multienzimáticos/química , Desenvolvimento Muscular , Complexo de Endopeptidases do Proteassoma , Células-Tronco/citologia , Frações Subcelulares/química
18.
C R Acad Sci III ; 320(5): 367-74, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9239322

RESUMO

Evidence has accumulated that suggests that insulin-like growth factors (IGFs) exert a positive influence on myoblast differentiation. We have undertaken to study the signalling events required for differentiation resulting from type-1 IGF receptor stimulation in C2 myoblasts, where autocrine production of IGF-II was abolished by means of antisense RNA. Exposure of the cells to IGFs leads to a rapid and sustained activation of phosphatidyl-inositol 3-kinase followed by the expression of Myod, myogenin and differentiation. The fungal metabolite, wortmannin, inhibits both PI 3-kinase and muscle differentiation with an IC 50 in the nanomolar range. IGFs are also known to cause a rapid activation of MAP kinase. However, the synthetic inhibitor of MEK, PD098059, which prevents MAP kinase activation, does not affect myoblast differentiation. These results provide evidence that PI 3-kinase, but not MAP kinase, is required for insulin-like growth factor receptor-dependent differentiation of muscle cells.


Assuntos
Androstadienos/farmacologia , Inibidores Enzimáticos/farmacologia , Músculos/citologia , Somatomedinas/farmacologia , Androstadienos/administração & dosagem , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Flavonoides/farmacologia , Camundongos , Músculos/enzimologia , Wortmanina
19.
J Cell Sci ; 110 ( Pt 22): 2771-9, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9427286

RESUMO

The muscle regulatory factor, myf5, is involved in the establishment of skeletal muscle precursor cells. Little is known, however, about the control of the expression of the gene encoding this basic helix-loop-helix (bHLH) factor. We have addressed this question in the mouse myogenic cell line, C2, and in a derivative of this cell line where the myf5 gene is the only muscle-specific bHLH factor to be expressed at the myoblast stage. We present evidence that the synthetic glucocorticoid dexamethasone, and the pharmacological agent anisomycin, act synergistically to rapidly up-regulate the levels of myf5 transcript and protein. The glucocorticoid antagonist RU 486 abolishes this synergy, demonstrating the involvement of the glucocorticoid receptor. The expression of a dominant negative mutant of c-jun which interferes with the transactivating properties of all AP-1 family members also blocks the induction of myf5 by anisomycin and dexamethasone. An activator of protein kinase C (PKCs), 12-O-tetradecanoyl phorbol 13-acetate (TPA), abolishes the up-regulation of myf5 gene expression by dexamethasone and anisomycin, and its effect is counteracted by an inhibitor of PKCs, GF 109203X. These results point to the possible involvement of PKCs in the negative control of myf5. Evidence that both positive and negative regulation of myf5 transcripts, described here, does not require the fresh synthesis of transcription factors suggests that myf5 may behave like an immediate early gene.


Assuntos
Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Receptores de Glucocorticoides/genética , Transativadores , Fator de Transcrição AP-1/genética , Animais , Linhagem Celular , Sequências Hélice-Alça-Hélice , Camundongos , Proteínas Musculares/metabolismo , Fator Regulador Miogênico 5 , Receptores de Glucocorticoides/metabolismo , Fator de Transcrição AP-1/metabolismo , Transfecção
20.
J Cell Sci ; 109 ( Pt 3): 551-60, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8907701

RESUMO

We have studied the contribution of the endogenous production of insulin-like growth factor II (IGFII) and of the muscle regulatory factor, MyoD, to the autonomy of differentiation in isolated skeletal myoblasts. Inhibition of MyoD and IGFII gene expression in myoblasts of the mouse myogenic cell line, C2, was achieved by transfection and selection of stably transfected cells (anti-MyoD and anti-IGFII cells) with vectors producing MyoD or IGFII antisense RNA. We observed that inhibiting either MyoD or IGFII has multiple and similar consequences. In addition to the inhibition of the target gene, expression of MyoD transcripts in anti-IGFII myoblasts and expression of IGFII in anti-MyoD myoblasts were also abolished, whereas accumulation of transcripts for the muscle regulatory factor, Myf5, was markedly increased in both cell types. However, despite this Myf5 up-regulation, both anti-IGFII and anti-MyoD myoblasts lost the ability to undergo autonomous differentiation (differentiation in the absence of added IGF), further indicating that Myf5 and MyoD are not strictly interchangeable. Additional evidence of a link between MyoD and IGFII was obtained: (1) forced expression of the MyoD cDNA stimulated IGFII gene expression, and (2) treatment of C2 myoblasts with fibroblast growth factor, not only diminished MyoD expression and compromised differentiation as previously shown by others, but also abolished IGFII expression. These experiments showing loss or gain of function argue in favor of a mutual positive control between IGFII and MyoD operating as early as the myoblast stage.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator de Crescimento Insulin-Like II/fisiologia , Músculos/citologia , Proteína MyoD/fisiologia , RNA Antissenso , Animais , Sequência de Bases , Diferenciação Celular/fisiologia , Linhagem Celular , Regulação para Baixo , Desenvolvimento Embrionário e Fetal/fisiologia , Camundongos , Dados de Sequência Molecular , Proteína MyoD/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...