Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 26(4): 997-1008, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18258721

RESUMO

Skeletal muscle is susceptible to injury following trauma, neurological dysfunction, and genetic diseases. Skeletal muscle homeostasis is maintained by a pronounced regenerative capacity, which includes the recruitment of stem cells. Chronic exposure to tumor necrosis factor-alpha (TNF) triggers a muscle wasting reminiscent of cachexia. To better understand the effects of TNF upon muscle homeostasis and stem cells, we exposed injured muscle to TNF at specific time points during regeneration. TNF exposure delayed the appearance of regenerating fibers, without exacerbating fiber death following the initial trauma. We observed modest cellular caspase activation during regeneration, which was markedly increased in response to TNF exposure concomitant with an inhibition in regeneration. Caspase activation did not lead to apoptosis and did not involve caspase-3. Inhibition of caspase activity improved muscle regeneration in either the absence or the presence of TNF, revealing a nonapoptotic role for this pathway in the myogenic program. Caspase activity was localized to the interstitial cells, which also express Sca-1, CD34, and PW1. Perturbation of PW1 activity blocked caspase activation and improved regeneration. The restricted localization of Sca-1+, CD34+, PW1+ cells to a subset of interstitial cells with caspase activity reveals a critical regulatory role for this population during myogenesis, which may directly contribute to resident muscle stem cells or indirectly regulate stem cells through cell-cell interactions.


Assuntos
Caspases/fisiologia , Músculo Esquelético/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Feminino , Camundongos , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Células-Tronco/citologia
2.
Cytometry A ; 71(10): 846-56, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17694560

RESUMO

Static magnetic field (SMF) interacts with mammal skeletal muscle; however, SMF effects on skeletal muscle cells are poorly investigated. The myogenic cell line L6, an in vitro model of muscle development, was used to investigate the effect of a 80 +/- mT SMF generated by a custom-made magnet. SMF promoted myogenic cell differentiation and hypertrophy, i.e., increased accumulation of actin and myosin and formation of large multinucleated myotubes. The elevated number of nuclei per myotube was derived from increased cell fusion efficiency, with no changes in cell proliferation upon SMF exposure. No alterations in myogenin expression, a modulator of myogenesis, occurred upon SMF exposure. SMF induced cells to align in parallel bundles, an orientation conserved throughout differentiation. SMF stimulated formation of actin stress-fiber like structures. SMF rescued muscle differentiation in the presence of TNF, a muscle differentiation inhibitor. We believe this is the first report showing that SMF promotes myogenic differentiation and cell alignment, in the absence of any invasive manipulation. SMF-enhanced parallel orientation of myotubes is relevant to tissue engineering of a highly organized tissue such as skeletal muscle. SMF rescue of muscle differentiation in the presence of TNF may have important therapeutic implications.


Assuntos
Diferenciação Celular , Magnetismo , Músculo Esquelético/citologia , Mioblastos/citologia , Actinas/metabolismo , Análise de Variância , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Polaridade Celular , Regulação da Expressão Gênica , Hipertrofia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Mioblastos/patologia , Cadeias Pesadas de Miosina/metabolismo , Fibras de Estresse/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
3.
FASEB J ; 21(7): 1393-402, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17264161

RESUMO

Muscle regeneration following injury is characterized by myonecrosis accompanied by local inflammation, activation of satellite cells, and repair of injured fibers. The resolution of the inflammatory response is necessary to proceed toward muscle repair, since persistence of inflammation often renders the damaged muscle incapable of sustaining efficient muscle regeneration. Here, we show that local expression of a muscle-restricted insulin-like growth factor (IGF)-1 (mIGF-1) transgene accelerates the regenerative process of injured skeletal muscle, modulating the inflammatory response, and limiting fibrosis. At the molecular level, mIGF-1 expression significantly down-regulated proinflammatory cytokines, such as tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta, and modulated the expression of CC chemokines involved in the recruitment of monocytes/macrophages. Analysis of the underlying molecular mechanisms revealed that mIGF-1 expression modulated key players of inflammatory response, such as macrophage migration inhibitory factor (MIF), high mobility group protein-1 (HMGB1), and transcription NF-kappaB. The rapid restoration of injured mIGF-1 transgenic muscle was also associated with connective tissue remodeling and a rapid recovery of functional properties. By modulating the inflammatory response and reducing fibrosis, supplemental mIGF-1 creates a qualitatively different environment for sustaining more efficient muscle regeneration and repair.


Assuntos
Quimiocinas/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Músculo Esquelético/fisiologia , Regeneração , Animais , Sequência de Bases , Imunofluorescência , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Genesis ; 43(3): 120-8, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16158413

RESUMO

Chronic disease states are associated with elevated levels of inflammatory cytokines that have been demonstrated to lead to severe muscle wasting. A mechanistic understanding of muscle wasting is hampered by limited in vivo cytokine models which can be applied to emerging mouse mutants as they are generated. We developed a simple and novel approach to induce adult mouse skeletal muscle wasting based on direct gene transfer of an expression vector encoding the secreted form of the murine tumor necrosis factor-alpha (mTNFalpha). This procedure results in the production of elevated levels of circulating mTNFalpha followed by body weight loss, upregulation of Atrogin1, and muscle atrophy, including muscles distant from the site of gene transfer. We also found that mTNFalpha gene transfer resulted in a significant inhibition of regeneration following muscle injury. We conclude that in addition to being a potent inducer of cachexia, TNFalpha is a potent inhibitor of myogenesis in vivo.


Assuntos
Técnicas de Transferência de Genes , Músculo Esquelético/fisiologia , Atrofia Muscular/genética , Regeneração/fisiologia , Fator de Necrose Tumoral alfa/genética , Animais , Eletroporação , Masculino , Camundongos , Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Mioblastos/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
5.
J Cell Biol ; 168(2): 193-9, 2005 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-15657392

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by a selective degeneration of motor neurons, atrophy, and paralysis of skeletal muscle. Although a significant proportion of familial ALS results from a toxic gain of function associated with dominant SOD1 mutations, the etiology of the disease and its specific cellular origins have remained difficult to define. Here, we show that muscle-restricted expression of a localized insulin-like growth factor (Igf) -1 isoform maintained muscle integrity and enhanced satellite cell activity in SOD1(G93A) transgenic mice, inducing calcineurin-mediated regenerative pathways. Muscle-specific expression of local Igf-1 (mIgf-1) isoform also stabilized neuromuscular junctions, reduced inflammation in the spinal cord, and enhanced motor neuronal survival in SOD1(G93A) mice, delaying the onset and progression of the disease. These studies establish skeletal muscle as a primary target for the dominant action of inherited SOD1 mutation and suggest that muscle fibers provide appropriate factors, such as mIgf-1, for neuron survival.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Fator de Crescimento Insulin-Like I/fisiologia , Neurônios Motores/metabolismo , Músculo Esquelético/metabolismo , Agrina/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/mortalidade , Animais , Astrócitos/metabolismo , Northern Blotting , Western Blotting , Calcineurina/genética , Calcineurina/metabolismo , Sistema Nervoso Central/química , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Desmina/metabolismo , Modelos Animais de Doenças , Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/genética , Camundongos , Camundongos Transgênicos , Neurônios Motores/patologia , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/química , Músculo Esquelético/patologia , Cadeias Pesadas de Miosina/metabolismo , Junção Neuromuscular/metabolismo , Fator de Transcrição PAX7 , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Células Satélites de Músculo Esquelético/química , Células Satélites de Músculo Esquelético/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Taxa de Sobrevida , Fator de Necrose Tumoral alfa/metabolismo , Caminhada
6.
Proc Natl Acad Sci U S A ; 101(5): 1206-10, 2004 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-14745025

RESUMO

We investigated the mechanism whereby expression of a transgene encoding a locally acting isoform of insulin-like growth factor 1 (mIGF-1) enhances repair of skeletal muscle damage. Increased recruitment of proliferating bone marrow cells to injured MLC/mIgf-1 transgenic muscles was accompanied by elevated bone marrow stem cell production in response to distal trauma. Regenerating MLC/mIgf-1 transgenic muscles contained increased cell populations expressing stem cell markers, exhibited accelerated myogenic differentiation, expressed markers of regeneration and readily converted cocultured bone marrow to muscle. These data implicate mIGF-1 as a powerful enhancer of the regeneration response, mediating the recruitment of bone marrow cells to sites of tissue damage and augmenting local repair mechanisms.


Assuntos
Fator de Crescimento Insulin-Like I/fisiologia , Músculos/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Animais , Células da Medula Óssea/fisiologia , Antígeno CD11b/análise , Movimento Celular , Antígenos Comuns de Leucócito/análise , Camundongos , Camundongos Transgênicos , Isoformas de Proteínas
7.
J Biol Chem ; 278(49): 49308-15, 2003 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-14506285

RESUMO

We have previously shown that myogenesis induction by Arg8-vasopressin (AVP) in L6 rat myoblasts involves a sustained stimulation of type 4 cAMP-phosphodiesterase. In this model, we observed that a transient cAMP generation occurs in the minutes following AVP addition. Evidence suggests that cAMP generation is due to the prostaglandins produced in response to AVP binding to V1a receptors and subsequent activation of phospholipase A2. The early cAMP increase was effective in activating cAMP-dependent protein kinase (PKA) and increasing phosphorylation of CREB transcription factor. Inhibition of PKA by compound H89 prior to AVP addition led to a significant reduction of expression of the differentiation marker creatine kinase, whereas H89 added 1-5 h after AVP had no significant effect. Furthermore, PKA inhibition 24 h after the beginning of AVP treatment potentiated differentiation. This shows that both an early activation and a later down-regulation of the cAMP pathway are required for AVP induction of myogenesis. Because phosphodiesterase PDE4D3 overexpressed in L6 cells lost its ability to potentiate AVP-induced differentiation when mutated and rendered insensitive to PKA phosphorylation and activation, we hypothesize that the early cAMP increase is required to trigger the down-regulation of cAMP pathway through stimulation of phosphodiesterase.


Assuntos
Diferenciação Celular , AMP Cíclico/metabolismo , Mioblastos/citologia , Sulfonamidas , Animais , Arginina Vasopressina/farmacologia , Sequência de Bases , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Primers do DNA , Inibidores Enzimáticos/farmacologia , Isoquinolinas/farmacologia , Mioblastos/enzimologia , Mioblastos/metabolismo , Ratos , Transdução de Sinais
8.
Am J Physiol Cell Physiol ; 284(4): C969-76, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12490436

RESUMO

Cytoplasmic Ca(2+) concentration ([Ca(2+)](i)) variation is a key event in myoblast differentiation, but the mechanism by which it occurs is still debated. Here we show that increases of extracellular Ca(2+) concentration ([Ca(2+)](o)) produced membrane hyperpolarization and a concentration-dependent increase of [Ca(2+)](i) due to Ca(2+) influx across the plasma membrane. Responses were not related to inositol phosphate turnover and Ca(2+)-sensing receptor. [Ca(2+)](o)-induced [Ca(2+)](i) increase was inhibited by Ca(2+) channel inhibitors and appeared to be modulated by several kinase activities. [Ca(2+)](i) increase was potentiated by depletion of intracellular Ca(2+) stores and depressed by inactivation of the Na(+)/Ca(2+) exchanger. The response to arginine vasopressin (AVP), which induces inositol 1,4,5-trisphosphate-dependent [Ca(2+)](i) increase in L6-C5 cells, was not modified by high [Ca(2+)](o). On the contrary, AVP potentiated the [Ca(2+)](i) increase in the presence of elevated [Ca(2+)](o). Other clones of the L6 line as well as the rhabdomyosarcoma RD cell line and the satellite cell-derived C2-C12 line expressed similar responses to high [Ca(2+)](o), and the amplitude of the responses was correlated with the myogenic potential of the cells.


Assuntos
Cálcio/metabolismo , Citosol/metabolismo , Espaço Extracelular/metabolismo , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/fisiologia , Animais , Arginina Vasopressina/farmacologia , Linhagem Celular , Membranas Intracelulares/metabolismo , Potenciais da Membrana/fisiologia , Músculo Esquelético/citologia , Mioblastos/fisiologia , Concentração Osmolar , Fosforilação , Ratos
9.
J Cell Sci ; 115(Pt 18): 3587-99, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12186945

RESUMO

We have previously suggested that PKCalpha has a role in 12-O-Tetradecanoylphorbol-13-acetate (TPA)-mediated growth arrest and myogenic differentiation in human embryonal rhabdomyosarcoma cells (RD). Here, by monitoring the signalling pathways triggered by TPA, we demonstrate that PKCalpha mediates these effects by inducing transient activation of c-Jun N-terminal protein kinases (JNKs) and sustained activation of both p38 kinase and extracellular signal-regulated kinases (ERKs) (all referred to as MAPKs). Activation of MAPKs following ectopic expression of constitutively active PKCalpha, but not its dominant-negative form, is also demonstrated. We investigated the selective contribution of MAPKs to growth arrest and myogenic differentiation by monitoring the activation of MAPK pathways, as well as by dissecting MAPK pathways using MEK1/2 inhibitor (UO126), p38 inhibitor (SB203580) and JNK and p38 agonist (anisomycin) treatments. Growth-arresting signals are triggered either by transient and sustained JNK activation (by TPA and anisomycin, respectively) or by preventing both ERK and JNK activation (UO126) and are maintained, rather than induced, by p38. We therefore suggest a key role for JNK in controlling ERK-mediated mitogenic activity. Notably, sarcomeric myosin expression is induced by both TPA and UO126 but is abrogated by the p38 inhibitor. This finding indicates a pivotal role for p38 in controlling the myogenic program. Anisomycin persistently activates p38 and JNKs but prevents myosin expression induced by TPA. In accordance with this negative role, reactivation of JNKs by anisomycin, in UO126-pre-treated cells, also prevents myosin expression. This indicates that, unlike the transient JNK activation that occurs in the TPA-mediated myogenic process, long-lasting JNK activation supports the growth-arrest state but antagonises p38-mediated myosin expression. Lastly, our results with the MEK inhibitor suggest a key role of the ERK pathway in regulating myogenic-related morphology in differentiated RD cells.


Assuntos
Diferenciação Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fibras Musculares Esqueléticas/enzimologia , Neoplasias Musculares/enzimologia , Proteína Quinase C/metabolismo , Rabdomiossarcoma/enzimologia , Células-Tronco/enzimologia , Anisomicina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Criança , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Musculares/genética , Proteína Quinase C/genética , Proteína Quinase C-alfa , Rabdomiossarcoma/genética , Ativador de Plasminogênio Tecidual/farmacologia , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA