Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 308(7): R576-89, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25608750

RESUMO

Activation of muscle progenitor cell myogenesis and endothelial cell angiogenesis is critical for the recovery of skeletal muscle from injury. Angiopoietin-1 (Ang-1), a ligand of Tie-2 receptors, enhances angiogenesis and skeletal muscle satellite cell survival; however, its role in skeletal muscle regeneration after injury is unknown. We assessed the effects of Ang-1 on fiber regeneration, myogenesis, and angiogenesis in injured skeletal muscle (tibialis anterior, TA) in mice. We also assessed endogenous Ang-1 levels and localization in intact and injured TA muscles. TA fiber injury was triggered by cardiotoxin injection. Endogenous Ang-1 mRNA levels immediately decreased in response to cardiotoxin then increased during the 2 wk. Ang-1 protein was expressed in satellite cells, both in noninjured and recovering TA muscles. Positive Ang-1 staining was present in blood vessels but not in nerve fibers. Four days after the initiation of injury, injection of adenoviral Ang-1 into injured muscles resulted in significant increases in in situ TA muscle contractility, muscle fiber regeneration, and capillary density. In cultured human skeletal myoblasts, recombinant Ang-1 protein increased survival, proliferation, migration, and differentiation into myotubes. The latter effect was associated with significant upregulation of the expression of the myogenic regulatory factors MyoD and Myogenin and certain genes involved in cell cycle regulation. We conclude that Ang-1 strongly enhances skeletal muscle regeneration in response to fiber injury and that this effect is mediated through induction of the myogenesis program in muscle progenitor cells and the angiogenesis program in endothelial cells.


Assuntos
Angiopoietina-1/metabolismo , Terapia Genética/métodos , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Doenças Musculares/terapia , Regeneração , Adenoviridae/genética , Adulto , Angiopoietina-1/genética , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Cardiotoxinas , Diferenciação Celular , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Vetores Genéticos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Doenças Musculares/induzido quimicamente , Doenças Musculares/genética , Doenças Musculares/patologia , Doenças Musculares/fisiopatologia , Mioblastos/metabolismo , Mioblastos/patologia , Necrose , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo
2.
Hum Mol Genet ; 21(10): 2277-87, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22343142

RESUMO

Nutrient-deprivation autophagy factor-1 (NAF-1) was identified as an endoplasmic reticulum (ER) BCL-2-interacting protein, which functions to mediate the ability of ER BCL-2 to antagonize Beclin 1-dependent autophagy and depress ER calcium stores. In humans, a point mutation in Naf-1 (synonyms: Cisd2, Eris, Miner1 and Noxp70) is responsible for the neurodegenerative disorder Wolfram Syndrome 2. Here, we describe the generation and characterization of the Naf-1 gene deletion in mice. Naf-1 null mice display discernable clinical signs of degeneration at 2-3 months of age, with early evidence of significant defects in the structure and performance of skeletal muscle. Skeletal muscles from Naf-1 knockout mice demonstrate a significant shift towards slow-twitch (type I) fibers and greater resistance to muscle fatigue. Force-generating capacity is dramatically reduced in Naf-1(-/-) muscle. Consistent with its role in ER BCL-2-mediated regulation of autophagy and calcium flux, these physiological deficiencies were accompanied by augmented autophagy and dysregulated calcium homeostasis. In contrast, this also included adaptive enlargement of mitochondria with extensive cristae structures. Thus, NAF-1, a BCL-2-associated autophagy regulator, is required for homeostatic maintenance of skeletal muscle. Our findings uncover a novel pathway that is required for normal muscle maintenance, which may ultimately provide a novel therapeutic target for treating certain muscle pathologies.


Assuntos
Autofagia , Proteínas de Transporte/genética , Músculo Esquelético/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas/metabolismo , Ribonucleoproteínas/genética , Animais , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2 , Ribonucleoproteínas/metabolismo
3.
Crit Care Med ; 42(7): e501-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24933061

RESUMO

OBJECTIVE: Diaphragmatic weakness and acute respiratory failure are common in sepsis. Nuclear factor-κB acts as a general coordinator of the systemic inflammatory response, but its role within the diaphragm itself during sepsis is unknown. We investigated the potential protective effect upon the diaphragm of inhibiting nuclear factor-κB only within muscle fibers during acute endotoxemia. DESIGN: Prospective study in experimental animals. SETTING: University research laboratory. INTERVENTIONS: Wild-type and transgenic (muscle-specific IκBα super-repressor) mice with skeletal muscle-specific inhibition of the classical nuclear factor-κB pathway were subjected to acute endotoxemia. Muscle-specific ubiquitin ligases (muscle RING-finger protein 1 and atrogin-1), caspase-3 activity, inhibitor of apoptosis proteins, proinflammatory cytokines (interleukin-1ß, monocyte chemoattractant protein-1, and tumor necrosis factor-α), and diaphragmatic contractility were evaluated after 24 hours. MEASUREMENTS AND MAIN RESULTS: In wild-type mice, endotoxemia significantly increased proinflammatory cytokines (fold-change messenger RNA: interleukin-1ß = 7.6, monocyte chemoattractant protein-1 = 15.3, and tumor necrosis factor-α = 2.2) and proteolysis effectors (fold-change messenger RNA: muscle RING-finger protein 1 = 5.7, atrogin-1 = 2.8; caspase-3 activity elevated by 28%) in the diaphragm, while reducing its force-generating capacity by 38%. In nonendotoxemic muscle-specific IκBα super-repressor diaphragms, caspase-3 activity was unexpectedly increased by 40% above basal wild-type levels and inhibitors of apoptosis proteins were down-regulated, but force production remained normal. In muscle-specific IκBα super-repressor mice subjected to endotoxemia, proinflammatory cytokines, muscle RING-finger protein 1, and atrogin-1 were not significantly increased above their basal levels, and diaphragmatic weakness and further increases in caspase-3 activity were completely prevented. CONCLUSIONS: These results suggest that nuclear factor-κB signaling within skeletal muscle fibers is a key pathway leading to diaphragmatic weakness during acute endotoxemia, most likely via effects on multiple inflammatory mediators. In addition, inhibition of nuclear factor-κB signaling within diaphragm muscle fibers has complex effects on caspase-3 activation, which could have implications for the treatment of sepsis-induced diaphragmatic dysfunction.


Assuntos
Diafragma/metabolismo , Endotoxemia/fisiopatologia , Inflamação/metabolismo , NF-kappa B/antagonistas & inibidores , Animais , Apoptose/fisiologia , Caspase 3/biossíntese , Citocinas/biossíntese , Camundongos , Camundongos Transgênicos , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas , Ubiquitina-Proteína Ligases/metabolismo
4.
Am J Respir Crit Care Med ; 186(11): 1140-9, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23024021

RESUMO

RATIONALE: Mechanical ventilation (MV) is associated with adverse effects on the diaphragm, but the cellular basis for this phenomenon, referred to as ventilator-induced diaphragmatic dysfunction (VIDD), is poorly understood. OBJECTIVES: To determine whether mitochondrial function and cellular energy status are disrupted in human diaphragms after MV, and the role of mitochondria-derived oxidative stress in the development of VIDD. METHODS: Diaphragm and biceps specimens obtained from brain-dead organ donors who underwent MV (15-176 h) and age-matched control subjects were compared regarding mitochondrial enzymatic function, mitochondrial DNA integrity, lipid content, and metabolic gene and protein expression. In addition, diaphragmatic force and oxidative stress after exposure to MV for 6 hours were evaluated in mice under different conditions. MEASUREMENTS AND MAIN RESULTS: In human MV diaphragms, mitochondrial biogenesis and content were down-regulated, with a more specific defect of respiratory chain cytochrome-c oxidase. Laser capture microdissection of cytochrome-c oxidase-deficient fibers revealed mitochondrial DNA deletions, consistent with damage from oxidative stress. Diaphragmatic lipid accumulation and responses of master cellular metabolic sensors (AMP-activated protein kinase and sirtuins) were consistent with energy substrate excess as a possible stimulus for these changes. In mice, induction of hyperlipidemia worsened diaphragmatic oxidative stress during MV, whereas transgenic overexpression of a mitochondria-localized antioxidant (peroxiredoxin-3) was protective against VIDD. CONCLUSIONS: Our data suggest that mitochondrial dysfunction lies at the nexus between oxidative stress and the impaired diaphragmatic contractility that develops during MV. Energy substrate oversupply relative to demand, resulting from diaphragmatic inactivity during MV, could play an important role in this process.


Assuntos
Diafragma/metabolismo , Metabolismo dos Lipídeos/fisiologia , Mitofagia , Estresse Oxidativo/fisiologia , Respiração Artificial/efeitos adversos , Animais , Estudos de Casos e Controles , Diafragma/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Reação em Cadeia da Polimerase em Tempo Real , Valores de Referência , Estatísticas não Paramétricas , Técnicas de Cultura de Tecidos
5.
PLoS Genet ; 5(7): e1000586, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19649303

RESUMO

Cystic fibrosis (CF) patients often have reduced mass and strength of skeletal muscles, including the diaphragm, the primary muscle of respiration. Here we show that lack of the CF transmembrane conductance regulator (CFTR) plays an intrinsic role in skeletal muscle atrophy and dysfunction. In normal murine and human skeletal muscle, CFTR is expressed and co-localized with sarcoplasmic reticulum-associated proteins. CFTR-deficient myotubes exhibit augmented levels of intracellular calcium after KCl-induced depolarization, and exposure to an inflammatory milieu induces excessive NF-kB translocation and cytokine/chemokine gene upregulation. To determine the effects of an inflammatory environment in vivo, sustained pulmonary infection with Pseudomonas aeruginosa was produced, and under these conditions diaphragmatic force-generating capacity is selectively reduced in Cftr(-/-) mice. This is associated with exaggerated pro-inflammatory cytokine expression as well as upregulation of the E3 ubiquitin ligases (MuRF1 and atrogin-1) involved in muscle atrophy. We conclude that an intrinsic alteration of function is linked to the absence of CFTR from skeletal muscle, leading to dysregulated calcium homeostasis, augmented inflammatory/atrophic gene expression signatures, and increased diaphragmatic weakness during pulmonary infection. These findings reveal a previously unrecognized role for CFTR in skeletal muscle function that may have major implications for the pathogenesis of cachexia and respiratory muscle pump failure in CF patients.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Fibrose Cística/fisiopatologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Animais , Cálcio/metabolismo , Células Cultivadas , Fibrose Cística/genética , Fibrose Cística/imunologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Citocinas/imunologia , Diafragma/imunologia , Diafragma/metabolismo , Diafragma/patologia , Diafragma/fisiopatologia , Modelos Animais de Doenças , Expressão Gênica , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Debilidade Muscular , Músculo Esquelético/imunologia , Músculo Esquelético/patologia
6.
Am J Respir Crit Care Med ; 182(11): 1377-86, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20639440

RESUMO

RATIONALE: Controlled mechanical ventilation (CMV) results in atrophy of the human diaphragm. The autophagy-lysosome pathway (ALP) contributes to skeletal muscle proteolysis, but its contribution to diaphragmatic protein degradation in mechanically ventilated patients is unknown. OBJECTIVES: To evaluate the autophagy pathway responses to CMV in the diaphragm and limb muscles of humans and to identify the roles of FOXO transcription factors in these responses. METHODS: Muscle biopsies were obtained from nine control subjects and nine brain-dead organ donors. Subjects were mechanically ventilated for 2 to 4 hours and 15 to 276 hours, respectively. Activation of the ubiquitin-proteasome system was detected by measuring mRNA expressions of Atrogin-1, MURF1, and protein expressions of UBC2, UBC4, and the α subunits of the 20S proteasome (MCP231). Activation of the ALP was detected by electron microscopy and by measuring the expressions of several autophagy-related genes. Total carbonyl content and HNE-protein adduct formation were measured to assess oxidative stress. Total AKT, phosphorylated and total FOXO1, and FOXO3A protein levels were also measured. MEASUREMENTS AND MAIN RESULTS: Prolonged CMV triggered activation of the ALP as measured by the appearance of autophagosomes in the diaphragm and increased expressions of autophagy-related genes, as compared with controls. Induction of autophagy was associated with increased protein oxidation and enhanced expression of the FOXO1 gene, but not the FOXO3A gene. CMV also triggered the inhibition of both AKT expression and FOXO1 phosphorylation. CONCLUSIONS: We propose that prolonged CMV causes diaphragm disuse, which, in turn, leads to activation of the ALP through oxidative stress and the induction of the FOXO1 transcription factor.


Assuntos
Autofagia , Diafragma/fisiopatologia , Respiração Artificial/efeitos adversos , Idoso , Western Blotting , Diafragma/metabolismo , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/metabolismo , Atrofia Muscular/etiologia , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatologia , Estresse Oxidativo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
7.
FASEB Bioadv ; 3(8): 639-651, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34377959

RESUMO

Duchenne muscular dystrophy (DMD) is a genetic disorder that results in the absence of dystrophin, a cytoskeletal protein. Individuals with this disease experience progressive muscle destruction, which leads to muscle weakness. Studies have been conducted to find solutions for the relief of individuals with this disease, several of which have shown that utrophin, a protein closely related to dystrophin, when overexpressed in mdx neonatal mice (the murine model of DMD), is able to prevent the progressive muscle destruction observed in the absence of dystrophin. Furthermore, recent studies have shown that L-arginine induces utrophin upregulation in adult mdx mice. We hypothesized that L-arginine treatment also induces utrophin upregulation to prevent the development of muscle weakness in neonatal mdx mice. Hence, L-arginine should also prevent progressive muscle destruction via utrophin upregulation in mdx neonatal mice. Mdx neonatal mice were injected intraperitoneally daily with 800 mg/kg of L-arginine for 6 weeks, whereas control mice were injected with a physiological saline. The following experiments were performed on the tibialis anterior (TA) muscle: muscle contractility and resistance to mechanical stress; central nucleation and peripheral nucleation, utrophin, and creatine kinase quantification as well as a nitric oxide (NO) assay. Our findings show that early administration of L-arginine in mdx neonatal mice prevents the destruction of the tibialis anterior (TA) muscle. However, this improvement was related to nitric oxide (NO) production rather than the expected utrophin upregulation.

8.
Crit Care ; 14(5): R187, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20950459

RESUMO

INTRODUCTION: Respiratory muscle weakness is common in sepsis patients. Proinflammatory mediators produced during sepsis have been implicated in diaphragmatic contractile dysfunction, but the role of chemokines has not been explored. This study addressed the role of monocyte chemoattractant protein-1 (MCP-1, also known as CCL2), in the pathogenesis of diaphragmatic inflammation and weakness during endotoxemia. METHODS: Mice were treated as follows (n = 6 per group): (a) saline, (b) endotoxin (25 µg/g IP), (c) endotoxin + anti-MCP-1 antibody, and (d) endotoxin + isotype control antibody. Muscles were also exposed to recombinant MCP-1 in vivo and in vitro. Measurements were made of diaphragmatic force generation, leukocyte infiltration, and proinflammatory mediator (MCP-1, IL-1α, IL-1ß, IL-6, NF-κB) expression/activity. RESULTS: In vivo, endotoxin-treated mice showed a large decrease in diaphragmatic force, together with upregulation of MCP-1 and other cytokines, but without an increase in intramuscular leukocytes. Antibody neutralization of MCP-1 prevented the endotoxin-induced force loss and reduced expression of MCP-1, IL-1α, IL-1ß, and IL-6 in the diaphragm. MCP-1 treatment of nonseptic muscles also led to contractile weakness, and MCP-1 stimulated its own transcription independent of NF-κB activation in vitro. CONCLUSIONS: These results suggest that MCP-1 plays an important role in the pathogenesis of diaphragmatic weakness during sepsis by both direct and indirect mechanisms. We speculate that its immunomodulatory properties and ability to modify skeletal muscle function make MCP-1 a potential therapeutic target in critically ill patients with sepsis and associated respiratory muscle weakness.


Assuntos
Quimiocina CCL2/antagonistas & inibidores , Diafragma/patologia , Endotoxemia/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Contração Muscular , Animais , Células Cultivadas , Quimiocina CCL2/fisiologia , Diafragma/fisiopatologia , Endotoxemia/induzido quimicamente , Endotoxemia/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Inflamação/prevenção & controle , Mediadores da Inflamação/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/fisiologia , Debilidade Muscular/metabolismo , Debilidade Muscular/fisiopatologia , Debilidade Muscular/prevenção & controle
9.
Mediators Inflamm ; 2009: 860565, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19421418

RESUMO

Sepsis-induced diaphragmatic inflammation has been associated with respiratory failure, but the role of chemokines in this process has not been evaluated. Here we sought to study the local expression and molecular regulation of the chemokines, regulated upon activation normal T cell expressed and secreted (RANTES) and macrophage inflammatory protein (MIP)-1alpha, in the murine diaphragm during sepsis. Constitutive expression levels of RANTES and MIP-1alpha, as well as their receptors, CCR1 and CCR5, were significantly higher in diaphragm than limb muscle. Sepsis was induced by acute lipopolysaccharide (LPS) delivery or subacutely by intratracheal administration of live Pseudomonas aeruginosa bacteria. Both sepsis models triggered a marked upregulation of RANTES and MIP-1alpha in the diaphragm. In vitro, stimulation of diaphragmatic muscle cells with LPS also led to RANTES upregulation. Inhibition of the NF-kB pathway using pharmacologic or dominant negative genetic approaches blocked the LPS-induced RANTES upregulation, while free radical scavengers had no effect. We conclude that sepsis leads to greatly increased expression of RANTES, MIP-1alpha and their cognate receptors in the diaphragm. Manipulation of the NF-kB pathway and other regulators of chemokine expression in the diaphragm could represent a novel method for mitigating the skeletal muscle inflammatory response associated with sepsis-induced diaphragmatic dysfunction.


Assuntos
Diafragma/metabolismo , Endotoxemia/fisiopatologia , Pneumopatias/microbiologia , Pneumopatias/fisiopatologia , Pseudomonas aeruginosa/patogenicidade , Receptores de Quimiocinas/metabolismo , Regulação para Cima , Animais , Linhagem Celular , Células Cultivadas , Quimiocina CCL3/genética , Quimiocina CCL5/genética , Diafragma/efeitos dos fármacos , Diafragma/microbiologia , Diafragma/patologia , Endotoxemia/microbiologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Ensaios de Proteção de Nucleases , Pseudomonas aeruginosa/fisiologia , Receptores CCR1/genética , Receptores CCR5/genética , Sepse/induzido quimicamente , Sepse/microbiologia , Sepse/fisiopatologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transfecção
10.
Front Physiol ; 10: 372, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31024337

RESUMO

Obesity and ensuing disorders are increasingly prevalent worldwide. High-fat diets (HFD) and diet-induced obesity have been shown to induce oxidative stress and inflammation while altering metabolic homeostasis in many organs, including the skeletal muscle. We previously observed that 14 days of HFD impairs contractile functions of the soleus (SOL) oxidative skeletal muscle. However, the mechanisms underlying these effects are not clarified. In order to determine the effects of a short-term HFD on skeletal muscle glutathione metabolism, young male Wistar rats (100-125 g) were fed HFD or a regular chow diet (RCD) for 14 days. Reduced (GSH) and disulfide (GSSG) glutathione levels were measured in the SOL. The expression of genes involved in the regulation of glutathione metabolism, oxidative stress, antioxidant defense and inflammation were measured by RNA-Seq. We observed a significant 25% decrease of GSH levels in the SOL muscle. Levels of GSSG and the GSH:GSSG ratio were similar in both groups. Further, we observed a 4.5 fold increase in the expression of pro-inflammatory cytokine interleukin 6 (IL-6) but not of other cytokines or markers of inflammation and oxidative stress. We hereby demonstrate that a short-term HFD significantly lowers SOL muscle GSH levels. This effect could be mediated through the increased expression of IL-6. Further, the skeletal muscle antioxidant defense could be impaired under cellular stress. We surmise that these early alterations could contribute to HFD-induced insulin resistance observed in longer protocols.

11.
J Physiol ; 586(16): 3991-4004, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18566004

RESUMO

Chemokines have been implicated in the promotion of leucocyte trafficking to diseased muscle. The purpose of this study was to determine whether a subset of inflammatory chemokines are able to directly drive myoblast proliferation, an essential early component of muscle regeneration, in a manner which is entirely independent of leucocytes. Cultured myoblasts (C2C12) were exposed to monocyte chemoattractant protein-1 (MCP-1; CCL2), macrophage inflammatory protein-1alpha (MIP-1alpha; CCL3) or MIP-1beta (CCL4). All chemokines induced phosphorylation of extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein kinase (MAPK) and greatly increased myoblast proliferative responses. Chemokine-induced myoblast proliferation was abolished by pertussis toxin and the MEK1/2 inhibitor U0126, implicating both Galphai-coupled receptors and ERK1/2-dependent signalling. Myoblasts expressed receptors for all of the chemokines tested, and mitogenic responses were specifically inhibited by antibodies directed against CC family chemokine receptors 2 and 5 (CCR2 and CCR5). Within an in vitro myogenic wound healing assay devoid of leucocytes, all chemokines significantly accelerated the time course of myoblast wound closure after mechanical injury. Injections of MCP-1 into cardiotoxin-injured skeletal muscles in vivo also suppressed expression of the differentiation marker myogenin, consistent with a mitogenic effect. Taken together, our results indicate that CC chemokines have potent and direct effects on myoblast behaviour, thus indicating a novel role in muscle repair beyond leucocyte chemoattraction. Therefore, interventions aimed at modulating the balance between myoblast and leucocyte effects of CC chemokines in injured muscle could represent a novel strategy for the treatment of destructive muscle pathologies.


Assuntos
Quimiocinas CC/metabolismo , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Animais , Linhagem Celular , Camundongos , Músculo Esquelético/patologia , Mioblastos/patologia
12.
Front Physiol ; 9: 1327, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30356919

RESUMO

Obesity and ensuing disorders are increasingly prevalent in young populations. Prolonged exposure to high-fat diets (HFD) and excessive lipid accumulation were recently suggested to impair skeletal muscle functions in rodents. We aimed to determine the effects of a short-term HFD on skeletal muscle function in young rats. Young male Wistar rats (100-125 g) were fed HFD or a regular chow diet (RCD) for 14 days. Specific force, resistance to fatigue and recovery were tested in extensor digitorum longus (EDL; glycolytic) and soleus (SOL; oxidative) muscles using an ex vivo muscle contractility system. Muscle fiber typing and insulin signaling were analyzed while intramyocellular lipid droplets (LD) were characterized. Expression of key markers of lipid metabolism was also measured. Weight gain was similar for both groups. Specific force was decreased in SOL, but not in EDL of HFD rats. Muscle resistance to fatigue and force recovery were not altered in response to the diets. Similarly, muscle fiber type distribution and insulin signaling were not influenced by HFD. On the other hand, percent area and average size of intramyocellular LDs were significantly increased in the SOL of HFD rats. These effects were consistent with the increased expression of several mediators of lipid metabolism in the SOL muscle. A short-term HFD impairs specific force and alters lipid metabolism in SOL, but not EDL muscles of young rats. This indicates the importance of clarifying the early mechanisms through which lipid metabolism affects skeletal muscle functions in response to obesogenic diets in young populations.

13.
J Nutr Metab ; 2018: 1498150, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29805802

RESUMO

Information regarding the early effects of obesogenic diets on feeding patterns and behaviors is limited. To improve knowledge regarding the etiology of obesity, young male Wistar rats were submitted to high-fat (HFD) or regular chow diets (RCDs) for 14 days. Various metabolic parameters were continuously measured using metabolic chambers. Total weight gain was similar between groups, but heavier visceral fat depots and reduced weight of livers were found in HFD rats. Total calorie intake was increased while individual feeding bouts were shorter and of higher calorie intake in response to HFD. Ambulatory activity and sleep duration were decreased in HFD rats during passive and active phase, respectively. Acylated and unacylated ghrelin levels were unaltered by the increased calorie intake and the early changes in body composition. This indicates that at this early stage, the orexigenic signal did not adapt to the high-calorie content of HFD. We hereby demonstrate that, although total weight gain is not affected, a short-term obesogenic diet alters body composition, feeding patterns, satiation, ambulatory activity profiles, and behaviours in a young rat model. Moreover, this effect precedes changes in weight gain, obesity, and ensuing metabolic disorders.

14.
EMBO Mol Med ; 6(11): 1476-92, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25312642

RESUMO

Myofiber necrosis and fibrosis are hallmarks of Duchenne muscular dystrophy (DMD), leading to lethal weakness of the diaphragm. Macrophages (MPs) are required for successful muscle regeneration, but the role of inflammatory monocyte (MO)-derived MPs in either promoting or mitigating DMD is unclear. We show that DMD (mdx) mouse diaphragms exhibit greatly increased expression of CCR2 and its chemokine ligands, along with inflammatory (Ly6C(high)) MO recruitment and accumulation of CD11b(high) MO-derived MPs. Loss-of-function of CCR2 preferentially reduced this CD11b(high) MP population by impeding the release of Ly6C(high) MOs from the bone marrow but not the splenic reservoir. CCR2 deficiency also helped restore the MP polarization balance by preventing excessive skewing of MPs toward a proinflammatory phenotype. These effects were linked to amelioration of histopathological features and increased muscle strength in the diaphragm. Chronic inhibition of CCR2 signaling by mutated CCL2 secreted from implanted mesenchymal stem cells resulted in similar improvements. These data uncover a previously unrecognized role of inflammatory MOs in DMD pathogenesis and indicate that CCR2 inhibition could offer a novel strategy for DMD management.


Assuntos
Quimiocina CCL2/metabolismo , Imunoterapia/métodos , Monócitos/imunologia , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/fisiopatologia , Receptores CCR2/antagonistas & inibidores , Animais , Antígenos Ly/análise , Antígeno CD11b/análise , Diafragma/patologia , Feminino , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Monócitos/química , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Proteínas Mutantes/metabolismo , Receptores CCR2/biossíntese , Resultado do Tratamento
15.
J Biol Chem ; 283(50): 34720-7, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18945675

RESUMO

Duchenne muscular dystrophy is caused by a genetic defect in the dystrophin gene. The absence of dystrophin results in muscle fiber necrosis and regeneration, leading to progressive muscle fiber loss. Utrophin is a close analogue of dystrophin. A substantial, ectopic expression of utrophin in the extrasynaptic sarcolemma of dystrophin-deficient muscle fibers can prevent deleterious effects of dystrophin deficiency. An alternative approach for the extrasynaptic up-regulation of utrophin involves the augmentation of utrophin transcription via the endogenous utrophin A promoter using custom-designed transcriptional activator proteins with zinc finger (ZFP) motifs. We tested a panel of custom-designed ZFP for their ability to activate the utrophin A promoter. Expression of one such ZFP efficiently increased, in a time-dependent manner, utrophin transcript and protein levels both in vitro and in vivo. In dystrophic mouse (mdx) muscles, administration of adenoviral vectors expressing this ZFP led to significant enhancement of muscle function with decreased necrosis, restoration of the dystrophin-associated proteins, and improved resistance to eccentric contractions. These studies provide evidence that specifically designed ZFPs can act as strong transcriptional activators of the utrophin A promoter. These may thus serve as attractive therapeutic agents for dystrophin deficiency states such as Duchenne muscular dystrophy.


Assuntos
Regulação da Expressão Gênica , Músculos/patologia , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Regulação para Cima , Utrofina/genética , Animais , Humanos , Camundongos , Camundongos Transgênicos , Contração Muscular , Músculos/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Necrose , Fatores de Tempo
16.
Am J Respir Cell Mol Biol ; 36(4): 504-12, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17122368

RESUMO

Severe weakness of the respiratory muscles, with attendant respiratory failure and death, has been documented in sepsis. In this study, we show that during murine pulmonary infection with Pseudomonas aeruginosa, multiple proinflammatory genes are up-regulated not only within the lungs, but also within the diaphragm. Significant induction of TNF-alpha, IL-1alpha, IL-1beta, IL-6, and IL-18 gene expression occurred within the diaphragm in a bacterial dose-dependent manner. We determined whether the anti-inflammatory cytokine IL-10 could blunt proinflammatory gene expression within the diaphragm under these conditions. The IL-10 receptor was found to be expressed by the diaphragm in vivo as well as in primary diaphragmatic muscle cell cultures. Transduction of myoblasts with an adenoviral vector (Ad-IL-10) induced strong IL-10 expression, and intramuscular injection of the same vector in vivo produced significant increases in IL-10 serum levels. Ad-IL-10 treatment of mice infected with P. aeruginosa significantly inhibited the induction of proinflammatory cytokines within the diaphragm, but not in the infected lungs. Ad-IL-10 treatment also led to greatly improved diaphragmatic force production in infected mice. These results suggest that pulmonary infection triggers proinflammatory gene expression by the diaphragm along with diaphragmatic weakness. Shifting the balance between pro- and anti-inflammatory mediators in favor of the latter by IL-10 gene delivery was able to restore normal diaphragmatic force-generating capacity under these conditions, suggesting a possible avenue for therapeutic intervention.


Assuntos
Citocinas/metabolismo , Diafragma/imunologia , Interleucina-10/fisiologia , Pneumopatias/imunologia , Contração Muscular , Infecções por Pseudomonas/imunologia , Animais , Células Cultivadas , Diafragma/microbiologia , Diafragma/fisiopatologia , Inflamação/imunologia , Inflamação/microbiologia , Pneumopatias/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-10/metabolismo , Transdução Genética , Regulação para Cima
17.
Mol Ther ; 15(10): 1767-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17667948

RESUMO

Helper-dependent adenovirus vector (AdV)-mediated full-length dystrophin expression leads to significant mitigation of the dystrophic phenotype of the mdx mouse. However, dystrophin, as a neoantigen, elicits antibody formation. As an alternative approach, we evaluated gene transfer of full-length murine utrophin, a functional homologue of dystrophin that is normally present only at the neuromuscular junction. A single injection in the tibialis anterior (TA) muscle of the helper-dependent adenovirus vector encoding utrophin provided very good transduction, with 58% of fibers demonstrating sarcolemmal utrophin expression in the neonates, and 35% utrophin-positive (Utr(+)) fibers in adults. The presence of utrophin prevented extensive necrosis in the neonates, halted further necrosis in the adults, and led to restoration of sarcolemmal expression of dystrophin-associated proteins up to 1 year after injection. Marked physiological improvement was observed in both neonates and adults. Neither increased humoral responses nor cellular immune responses were evident. However, there was a time-related decline of the initial high utrophin expression. Although viral DNA persisted in animals that were injected in the neonatal stage, viral DNA levels decreased in muscles of adult mice. These results demonstrate that although utrophin gene transfer leads to amelioration of the dystrophic phenotype, the effects are not sustained upon loss of utrophin expression.


Assuntos
Adenoviridae/genética , Distrofina/genética , Utrofina/genética , Animais , Animais Recém-Nascidos , Formação de Anticorpos , DNA Viral/metabolismo , Imunidade Celular , Camundongos , Camundongos Mutantes , Músculo Esquelético/metabolismo , Reação em Cadeia da Polimerase , Transdução Genética , Utrofina/administração & dosagem , Utrofina/imunologia
18.
Am J Pathol ; 168(4): 1276-87; quiz 1404-5, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16565501

RESUMO

Dystrophin deficiency is the cause of Duchenne muscular dystrophy, but the precise physiological basis for muscle necrosis remains unclear. To determine whether dystrophin-deficient muscles are abnormally susceptible to oxidative and nitric oxide (NO)-driven tissue stress, a hindlimb ischemia/reperfusion (I/R) model was used. Dystrophic mdx mice exhibited abnormally high levels of lipid peroxidation and protein nitration, which were preceded by exaggerated NO production during ischemia. Visualization of NO with the fluorescent probe 4,5-diaminofluorescein diacetate suggested that excess NO production during ischemia occurred within a subset of mdx fibers. In mdx muscles only, prior exposure to I/R dramatically increased the level of sarcolemmal damage resulting from stretch-mediated mechanical stress, indicating greatly exacerbated hyperfragility of the dystrophic fiber membrane. Treatment with NO synthase inhibitors (l-N(G)-nitroarginine methyl ester hydrochloride or 7-nitroindazol) effectively blocked the synergistic interaction between I/R and mechanical stress-mediated sarcolemmal damage under these conditions. Taken together, our findings provide direct ex-perimental evidence that several prevailing hy-potheses regarding the cause of muscle fiber damage in dystrophin-deficient muscle can be integrated into a common pathophysiological framework involving interactions between oxidative stress, ab-normal NO regulation, and hyperfragility of the sarcolemma.


Assuntos
Distrofina/deficiência , Distrofina/fisiologia , Músculo Esquelético/fisiopatologia , Óxido Nítrico/fisiologia , Estresse Oxidativo , Sarcolema/fisiologia , Animais , Distrofina/genética , Membro Posterior , Indazóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/ultraestrutura , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Sarcolema/efeitos dos fármacos , Estresse Mecânico
19.
Am J Respir Crit Care Med ; 174(6): 646-53, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778157

RESUMO

BACKGROUND: Sepsis-induced diaphragmatic force loss and failure are associated with an increased exposure of the muscle to proinflammatory mediators. OBJECTIVES: Our objectives were to test the hypothesis that force-inhibiting mediators may arise in large part from the diaphragm itself and to evaluate the roles of mechanical stress, free radicals, and the nuclear factor (NF)-kappaB transcription factor pathway in endotoxin (LPS)-induced proinflammatory responses of the diaphragm. METHODS: Murine diaphragm and limb muscle cells were exposed to LPS in vitro and in vivo. Proinflammatory gene expression was measured using RNase protection assays (tumor necrosis factor [TNF]-alpha, TNF-alpha receptor p55, interleukin [IL]-1alpha, IL-1beta, IL-6, macrophage inflammatory peptide-2, intercellular adhesion molecule-1, Fas ligand, and inducible nitric oxide synthase) and ELISAs (TNF-alpha, IL-6, and macrophage inflammatory peptide-2). Cyclical muscle cell stretch and free-radical scavengers (N-acetylcysteine and catalase) were used to alter mechanical and oxidative stress levels, respectively. Pharmacologic (pyrrolidinedithiocarbamate) and dominant-negative transfection strategies were used to inhibit the NF-kappaB pathway. RESULTS: In primary diaphragm muscle cell cultures, modulation of mechanical stress levels or free-radical exposure did not alter responses to LPS stimulation. However, pharmacologic blockade of the NF-kappaB pathway and dominant-negative molecular inhibition of IKB kinase-beta strongly suppressed LPS-induced proinflammatory gene expression. In vivo, acute endotoxemia induced significantly greater mRNA and protein levels for proinflammatory mediators in the diaphragm as compared with limb muscle. Basal expression levels of proinflammatory genes were significantly higher in the diaphragm. CONCLUSIONS: Constitutive and LPS-induced proinflammatory gene expression are exaggerated in the diaphragm compared with limb muscles and are critically dependent on the NF-kappaB pathway. We suggest the diaphragm may be relatively predisposed to proinflammatory responses.


Assuntos
Citocinas/genética , Diafragma/metabolismo , Endotoxemia/metabolismo , NF-kappa B/metabolismo , RNA Mensageiro/genética , Regulação para Cima/efeitos dos fármacos , Animais , Células Cultivadas , Diafragma/efeitos dos fármacos , Diafragma/patologia , Endotoxemia/induzido quimicamente , Endotoxemia/patologia , Ensaio de Imunoadsorção Enzimática , Técnicas In Vitro , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo
20.
Am J Pathol ; 169(6): 2148-60, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17148677

RESUMO

The Coxsackie and adenovirus receptor (CAR), a cell adhesion molecule of the immunoglobulin superfamily, is usually confined to the sarcolemma at the neuromuscular junction in mature skeletal muscle fibers. Previously, we reported that adenovirus-mediated gene transfer is greatly facilitated in hemizygous transgenic mice with extrasynaptic CAR expression driven by a muscle-specific promoter. However, in the present study, when these mice were bred to homozygosity, they developed a severe myopathic phenotype and died prematurely. Large numbers of necrotic and regenerating fibers were present in the skeletal muscle of the homozygous CAR transgenics. The myopathy was further characterized by increased levels of caveolin-3 and beta-dystroglycan and decreased levels of dystrophin, dysferlin, and neuronal nitric-oxide synthase. Even the hemizygotes manifested a subtle phenotype, displaying deficits in isometric force generation and perturbed mitogen-activated protein kinase (MAPK-erk1/2) activation during contraction. There are few naturally occurring or engineered mouse lines showing as severe a skeletal myopathy as observed with ectopic expression of CAR in the homozygotes. Taken together, these findings suggest that substantial overexpression of CAR may lead to physiological dysfunction by disturbing sarcolemmal integrity (through dystrophin deficiency), impairing sarcolemmal repair (through dysferlin deficiency), and interfering with normal signaling (through alterations in caveolin-3 and neuronal nitric-oxide synthase levels).


Assuntos
Distrofina/deficiência , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Junção Neuromuscular/patologia , Animais , Caveolina 3/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Disferlina , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Doenças Musculares/genética , Doenças Musculares/patologia , Contração Miocárdica , Óxido Nítrico Sintase Tipo I/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA