Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Am J Physiol Cell Physiol ; 315(2): C164-C185, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29561660

RESUMO

Parkinson's disease is a neurodegenerative disease characterized by tremors, muscle stiffness, and muscle weakness. Molecular genetic analysis has confirmed that mutations in PARKIN and PINK1 genes, which play major roles in mitochondrial quality control and mitophagy, are frequently associated with Parkinson's disease. PARKIN is an E3 ubiquitin ligase that translocates to mitochondria during loss of mitochondrial membrane potential to increase mitophagy. Although muscle dysfunction is noted in Parkinson's disease, little is known about the involvement of PARKIN in the muscle phenotype of Parkinson's disease. In this study, we report that the mitochondrial uncoupler CCCP promotes PINK1/PARKIN-mediated mitophagy in myogenic C2C12 cells. As a result of this excess mitophagy, we show that CCCP treatment of myotubes leads to the development of myotube atrophy in vitro. Surprisingly, we also found that siRNA-mediated knockdown of Parkin results in impaired mitochondrial turnover. In addition, knockdown of Parkin led to myotubular atrophy in vitro. Consistent with these in vitro results, Parkin knockout muscles showed impaired mitochondrial function and smaller myofiber area, suggesting that Parkin function is required for post-natal skeletal muscle growth and development.


Assuntos
Mitocôndrias/metabolismo , Atrofia Muscular/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células Cultivadas , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Mitofagia/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteínas Quinases/metabolismo
2.
J Biol Chem ; 289(11): 7654-70, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24451368

RESUMO

To date a plethora of evidence has clearly demonstrated that continued high calorie intake leads to insulin resistance and type-2 diabetes with or without obesity. However, the necessary signals that initiate insulin resistance during high calorie intake remain largely unknown. Our results here show that in response to a regimen of high fat or high glucose diets, Mstn levels were induced in muscle and liver of mice. High glucose- or fat-mediated induction of Mstn was controlled at the level of transcription, as highly conserved carbohydrate response and sterol-responsive (E-box) elements were present in the Mstn promoter and were revealed to be critical for ChREBP (carbohydrate-responsive element-binding protein) or SREBP1c (sterol regulatory element-binding protein 1c) regulation of Mstn expression. Further molecular analysis suggested that the increased Mstn levels (due to high glucose or fatty acid loading) resulted in increased expression of Cblb in a Smad3-dependent manner. Casitas B-lineage lymphoma b (Cblb) is an ubiquitin E3 ligase that has been shown to specifically degrade insulin receptor substrate 1 (IRS1) protein. Consistent with this, our results revealed that elevated Mstn levels specifically up-regulated Cblb, resulting in enhanced ubiquitin proteasome-mediated degradation of IRS1. In addition, over expression or knock down of Cblb had a major impact on IRS1 and pAkt levels in the presence or absence of insulin. Collectively, these observations strongly suggest that increased glucose levels and high fat diet, both, result in increased circulatory Mstn levels. The increased Mstn in turn is a potent inducer of insulin resistance by degrading IRS1 protein via the E3 ligase, Cblb, in a Smad3-dependent manner.


Assuntos
Dieta/efeitos adversos , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Miostatina/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteína Smad3/metabolismo , Animais , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Gorduras na Dieta/administração & dosagem , Regulação da Expressão Gênica , Glucose/administração & dosagem , Glucose/metabolismo , Células Hep G2 , Humanos , Lentivirus/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Proteínas Nucleares/metabolismo , Palmitatos/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Fatores de Transcrição/metabolismo
3.
J Biol Chem ; 289(9): 5784-98, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24425880

RESUMO

One of the features of uncontrolled type 1 diabetes is oxidative stress that induces DNA damage and cell death. Skeletal muscle atrophy is also considerable in type 1 diabetes, however, the signaling mechanisms that induce oxidative stress culminating in muscle atrophy are not fully known. Here, we show that in Streptozotocin-induced diabetic wild type mice, hypo-phosphorylation of Akt, resulted in activation of Foxa2 transcription factor in the muscle. Foxa2 transcriptionally up-regulated Myostatin, contributing to exaggerated oxidative stress leading to DNA damage via p63/REDD1 pathway in skeletal muscle of Streptozotocin-treated wild type mice. In Myostatin(-/-) mice however, Streptozotocin treatment did not reduce Akt phosphorylation despite reduced IRS-1 signaling. Moreover, Foxa2 levels remained unaltered in Myostatin(-/-) mice, while levels of p63/REDD1 were higher compared with wild type mice. Consistent with these results, relatively less DNA damage and muscle atrophy was observed in Myostatin(-/-) muscle in response to Streptozotocin treatment. Taken together, our results for the first time show the role of Foxa2 in Myostatin regulation in skeletal muscle in diabetic mice. Altogether, these results demonstrate the mechanism by which Myostatin contributes to DNA damage in skeletal muscle of the diabetic mice that would lead to myofiber degeneration.


Assuntos
Dano ao DNA , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miostatina/metabolismo , Estresse Oxidativo , Animais , Linhagem Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/patologia , Miostatina/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Am J Physiol Endocrinol Metab ; 309(2): E122-31, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25921579

RESUMO

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a ubiquitously expressed gene with higher levels observed in skeletal muscle. Recently, our laboratory showed (Bonala S, Lokireddy S, Arigela H, Teng S, Wahli W, Sharma M, McFarlane C, Kambadur R. J Biol Chem 287: 12935-12951, 2012) that PPARß/δ modulates myostatin activity to induce myogenesis in skeletal muscle. In the present study, we show that PPARß/δ-null mice display reduced body weight, skeletal muscle weight, and myofiber atrophy during postnatal development. In addition, a significant reduction in satellite cell number was observed in PPARß/δ-null mice, suggesting a role for PPARß/δ in muscle regeneration. To investigate this, tibialis anterior muscles were injured with notexin, and muscle regeneration was monitored on days 3, 5, 7, and 28 postinjury. Immunohistochemical analysis revealed an increased inflammatory response and reduced myoblast proliferation in regenerating muscle from PPARß/δ-null mice. Histological analysis confirmed that the regenerated muscle fibers of PPARß/δ-null mice maintained an atrophy phenotype with reduced numbers of centrally placed nuclei. Even though satellite cell numbers were reduced before injury, satellite cell self-renewal was found to be unaffected in PPARß/δ-null mice after regeneration. Previously, our laboratory had showed (Bonala S, Lokireddy S, Arigela H, Teng S, Wahli W, Sharma M, McFarlane C, Kambadur R. J Biol Chem 287: 12935-12951, 2012) that inactivation of PPARß/δ increases myostatin signaling and inhibits myogenesis. Our results here indeed confirm that inactivation of myostatin signaling rescues the atrophy phenotype and improves muscle fiber cross-sectional area in both uninjured and regenerated tibialis anterior muscle from PPARß/δ-null mice. Taken together, these data suggest that absence of PPARß/δ leads to loss of satellite cells, impaired skeletal muscle regeneration, and postnatal myogenesis. Furthermore, our results also demonstrate that functional antagonism of myostatin has utility in rescuing these effects.


Assuntos
Desenvolvimento Muscular/genética , PPAR delta/genética , PPAR beta/genética , Células Satélites de Músculo Esquelético/metabolismo , Animais , Regulação para Baixo/genética , Inativação Gênica , Crescimento e Desenvolvimento/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/patologia , Músculo Esquelético/fisiologia , Atrofia Muscular/genética
5.
IUBMB Life ; 67(8): 589-600, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26305594

RESUMO

Myostatin is a secreted growth and differentiation factor that belongs to the TGF-ß superfamily. Myostatin is predominantly synthesized and expressed in skeletal muscle and thus exerts a huge impact on muscle growth and function. In keeping with its negative role in myogenesis, myostatin expression is tightly regulated at several levels including epigenetic, transcriptional, post-transcriptional, and post-translational. New revelations regarding myostatin regulation also offer mechanisms that could be exploited for developing myostatin antagonists. Increasingly, it is becoming clearer that besides its conventional role in muscle, myostatin plays a critical role in metabolism. Hence, molecular mechanisms by which myostatin regulates several key metabolic processes need to be further explored.


Assuntos
Diferenciação Celular/genética , Desenvolvimento Muscular/genética , Miostatina/genética , Fator de Crescimento Transformador beta/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Miostatina/biossíntese , Miostatina/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional
6.
J Biol Chem ; 288(9): 6663-78, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23297411

RESUMO

High doses of dexamethasone (Dex) or myostatin (Mstn) induce severe atrophy of skeletal muscle. Here we show a novel microRNA1 (miR1)-mediated mechanism through which Dex promotes skeletal muscle atrophy. Using both C2C12 myotubes and mouse models of Dex-induced atrophy we show that Dex induces miR1 expression through glucocorticoid receptor (GR). We further show that Mstn treatment facilitates GR nuclear translocation and thereby induces miR1 expression. Inhibition of miR1 in C2C12 myotubes attenuated the Dex-induced increase in atrophy-related proteins confirming a role for miR1 in atrophy. Analysis of miR1 targets revealed that HSP70 is regulated by miR1 during atrophy. Our results demonstrate that increased miR1 during atrophy reduced HSP70 levels, which resulted in decreased phosphorylation of AKT, as HSP70 binds to and protects phosphorylation of AKT. We further show that loss of pAKT leads to decreased phosphorylation, and thus, enhanced activation of FOXO3, up-regulation of MuRF1 and Atrogin-1, and progression of skeletal muscle atrophy. Based on these results, we propose a model whereby Dex- and Mstn-mediated atrophic signals are integrated through miR1, which then either directly or indirectly, inhibits the proteins involved in providing protection against atrophy.


Assuntos
Anti-Inflamatórios/efeitos adversos , Dexametasona/efeitos adversos , Proteínas de Choque Térmico HSP70/metabolismo , MicroRNAs/metabolismo , Modelos Biológicos , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Células CHO , Cricetinae , Cricetulus , Dexametasona/farmacologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Choque Térmico HSP70/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Atrofia Muscular/genética , Atrofia Muscular/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Ligases SKP Culina F-Box/biossíntese , Proteínas Ligases SKP Culina F-Box/genética , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
7.
J Cell Biochem ; 115(11): 1908-17, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24909401

RESUMO

Conversion of skin fibroblasts into myoblasts by transducing the cells with myogenic master regulator MyoD has been in practice for more than two decades. The purpose of such conversion is due to scarcity of muscle biopsies during muscle wasting, hence conversion of fibroblasts to myogenic lineage from various genetic backgrounds offers a great alternative for cell therapies. Here, we have investigated if eliminating Myostatin, a potent negative regulator of myogenesis, could improve the myogenic conversion of fibroblasts. In the present study, we have isolated primary muscle fibroblasts from the skeletal muscles of wild-type (WT) and myostatin null (Mstn(-/-)) mice and transduced the muscle fibroblasts with MyoD using adenoviral, lentiviral transduction, and electroporation methods. In contrast to what we predicted, it is only in WT muscle fibroblasts we detected significant ectopic expression of MyoD, and myogenic conversion. Muscle fibroblasts from Mstn(-/-) genotype failed to take up as much MyoD using the three methods and, therefore, failed to form myotubes. The aforesaid condition of greater MyoD uptake by WT muscle fibroblasts was attributed to the presence of adenoviral receptors, which facilitated adenoviral transduction. However, in Mstn(-/-) fibroblasts we detected negligible levels of adenovirus receptors. Moreover, we also detected significantly higher levels of MyoD antagonists, c-Fos, c-Jun, and cyclin D1 in Mstn(-/-) muscle fibroblasts. Taken together, our results demonstrate that lack of myostatin reduces myogenic potential of muscle fibroblasts by inhibiting MyoD function.


Assuntos
Fibroblastos/citologia , Desenvolvimento Muscular , Músculo Esquelético/citologia , Proteína MyoD/genética , Miostatina/deficiência , Animais , Diferenciação Celular , Células Cultivadas , Ciclina D1/metabolismo , Terapia Genética , Camundongos , Camundongos Transgênicos , Proteína MyoD/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução Genética
8.
J Biol Chem ; 287(16): 12935-51, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22362769

RESUMO

Classically, peroxisome proliferator-activated receptor ß/δ (PPARß/δ) function was thought to be restricted to enhancing adipocyte differentiation and development of adipose-like cells from other lineages. However, recent studies have revealed a critical role for PPARß/δ during skeletal muscle growth and regeneration. Although PPARß/δ has been implicated in regulating myogenesis, little is presently known about the role and, for that matter, the mechanism(s) of action of PPARß/δ in regulating postnatal myogenesis. Here we report for the first time, using a PPARß/δ-specific ligand (L165041) and the PPARß/δ-null mouse model, that PPARß/δ enhances postnatal myogenesis through increasing both myoblast proliferation and differentiation. In addition, we have identified Gasp-1 (growth and differentiation factor-associated serum protein-1) as a novel downstream target of PPARß/δ in skeletal muscle. In agreement, reduced Gasp-1 expression was detected in PPARß/δ-null mice muscle tissue. We further report that a functional PPAR-responsive element within the 1.5-kb proximal Gasp-1 promoter region is critical for PPARß/δ regulation of Gasp-1. Gasp-1 has been reported to bind to and inhibit the activity of myostatin; consistent with this, we found that enhanced secretion of Gasp-1, increased Gasp-1 myostatin interaction and significantly reduced myostatin activity upon L165041-mediated activation of PPARß/δ. Moreover, we analyzed the ability of hGASP-1 to regulate myogenesis independently of PPARß/δ activation. The results revealed that hGASP-1 protein treatment enhances myoblast proliferation and differentiation, whereas silencing of hGASP-1 results in defective myogenesis. Taken together these data revealed that PPARß/δ is a positive regulator of skeletal muscle myogenesis, which functions through negatively modulating myostatin activity via a mechanism involving Gasp-1.


Assuntos
Desenvolvimento Muscular/fisiologia , Mioblastos/metabolismo , Miostatina/metabolismo , PPAR delta/metabolismo , PPAR beta/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Mioblastos/citologia , Miostatina/genética , PPAR delta/agonistas , PPAR beta/agonistas , Fenoxiacetatos/farmacologia , Transdução de Sinais/fisiologia
9.
Biochem J ; 446(1): 23-36, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22621320

RESUMO

Humoral and tumoral factors collectively promote cancer-induced skeletal muscle wasting by increasing protein degradation. Although several humoral proteins, namely TNFα (tumour necrosis factor α) and IL (interleukin)-6, have been shown to induce skeletal muscle wasting, there is a lack of information regarding the tumoral factors that contribute to the atrophy of muscle during cancer cachexia. Therefore, in the present study, we have characterized the secretome of C26 colon cancer cells to identify the tumoral factors involved in cancer-induced skeletal muscle wasting. In the present study, we show that myostatin, a procachectic TGFß (transforming growth factor ß) superfamily member, is abundantly secreted by C26 cells. Consistent with myostatin signalling during cachexia, treating differentiated C2C12 myotubes with C26 CM (conditioned medium) resulted in myotubular atrophy due to the up-regulation of muscle-specific E3 ligases, atrogin-1 and MuRF1 (muscle RING-finger protein 1), and enhanced activity of the ubiquitin-proteasome pathway. Furthermore, the C26 CM also activated ActRIIB (activin receptor type II B)/Smad and NF-κB (nuclear factor κB) signalling, and reduced the activity of the IGF-I (insulin-like growth factor 1)/PI3K (phosphoinositide 3-kinase)/Akt pathway, three salient molecular features of myostatin action in skeletal muscles. Antagonists to myostatin prevented C26 CM-induced wasting in muscle cell cultures, further confirming that tumoral myostatin may be a key contributor in the pathogenesis of cancer cachexia. Finally, we show that treatment with C26 CM induced the autophagy-lysosome pathway and reduced the number of mitochondria in myotubes. These two previously unreported observations were recapitulated in skeletal muscles collected from C26 tumour-bearing mice.


Assuntos
Caquexia/fisiopatologia , Neoplasias do Colo/fisiopatologia , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Receptores de Activinas Tipo II/metabolismo , Ativinas/metabolismo , Animais , Autofagia , Caquexia/etiologia , Caquexia/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Meios de Cultivo Condicionados/farmacologia , Humanos , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
10.
Am J Physiol Cell Physiol ; 303(5): C512-29, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22673621

RESUMO

Atrogin-1, a muscle-specific E3 ligase, targets MyoD for degradation through the ubiquitin-proteasome-mediated system. Myostatin, a member of the transforming growth factor-ß superfamily, potently inhibits myogenesis by lowering MyoD levels. While atrogin-1 is upregulated by myostatin, it is currently unknown whether atrogin-1 plays a role in mediating myostatin signaling to regulate myogenesis. In this report, we have confirmed that atrogin-1 increasingly interacts with MyoD upon recombinant human myostatin (hMstn) treatment. The absence of atrogin-1, however, led to elevated MyoD levels and permitted the differentiation of atrogin-1(-/-) primary myoblast cultures despite the presence of exogenous myostatin. Furthermore, inactivation of atrogin-1 rescued myoblasts from growth inhibition by hMstn. Therefore, these results highlight the central role of atrogin-1 in regulating myostatin signaling during myogenesis. Currently, there are only two known targets of atrogin-1. Thus, we next characterized the associated proteins of atrogin-1 in control and hMstn-treated C2C12 cell cultures by stably expressing tagged atrogin-1 in myoblasts and myotubes, and sequencing the coimmunoprecipitated proteome. We found that atrogin-1 putatively interacts with sarcomeric proteins, transcriptional factors, metabolic enzymes, components of translation, and spliceosome formation. In addition, we also identified that desmin and vimentin, two components of the intermediate filament in muscle, directly interacted with and were degraded by atrogin-1 in response to hMstn. In summary, the muscle wasting effects of the myostatin-atrogin-1 axis are not only limited to the degradation of MyoD and eukaryotic translation initiation factor 3 subunit f, but also encompass several proteins that are involved in a wide variety of cellular activities in the muscle.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Mioblastos Esqueléticos/metabolismo , Miostatina/farmacologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Proliferação de Células , Fator de Iniciação 3 em Eucariotos/genética , Fator de Iniciação 3 em Eucariotos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Proteínas Musculares/genética , Proteína MyoD/genética , Proteína MyoD/metabolismo , Ligação Proteica , Subunidades Proteicas , Proteínas Ligases SKP Culina F-Box/genética , Sarcômeros/metabolismo , Transcriptoma
11.
Am J Physiol Cell Physiol ; 302(8): C1213-25, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22277753

RESUMO

Myostatin (Mstn) is a secreted growth and differentiation factor that belongs to the transforming growth factor-ß (TGF-ß) superfamily. Mstn has been well characterized as a regulator of myogenesis and has been shown to play a critical role in postnatal muscle regeneration. Herein, we report for the first time that Mstn is expressed in both epidermis and dermis of murine and human skin and that Mstn-null mice exhibited delayed skin wound healing attributable to a combination of effects resulting from delayed epidermal reepithelialization and dermal contraction. In epidermis, reduced keratinocyte migration and protracted keratinocyte proliferation were observed, which subsequently led to delayed recovery of epidermal thickness and slower reepithelialization. Furthermore, primary keratinocytes derived from Mstn-null mice displayed reduced migration capacity and increased proliferation rate as assessed through in vitro migration and adhesion assays, as well as bromodeoxyuridine incorporation and Western blot analysis. Moreover, in dermis, both fibroblast-to-myofibroblast transformation and collagen deposition were concomitantly reduced, resulting in a delayed dermal wound contraction. These decreases are due to the inhibition of TGF-ß signaling. In agreement, the expression of decorin, a naturally occurring TGF-ß suppressor, was elevated in Mstn-null mice; moreover, topical treatment with TGF-ß1 protein rescued the impaired skin wound healing observed in Mstn-null mice. These observations highlight the interplay between TGF-ß and Mstn signaling pathways, specifically through Mstn regulation of decorin levels during the skin wound healing process. Thus we propose that Mstn agonists might be beneficial for skin wound repair.


Assuntos
Decorina/metabolismo , Miostatina/deficiência , Pele/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Cicatrização/fisiologia , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Decorina/biossíntese , Decorina/genética , Feminino , Folículo Piloso/metabolismo , Humanos , Queratinócitos/metabolismo , Queratinócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Miofibroblastos/metabolismo , Miofibroblastos/fisiologia , Miostatina/genética , Miostatina/metabolismo , Transdução de Sinais , Pele/citologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Cicatrização/genética
14.
Am J Physiol Endocrinol Metab ; 303(1): E90-102, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22535746

RESUMO

Smad3 is a key intracellular signaling mediator for both transforming growth factor-ß and myostatin, two major regulators of skeletal muscle growth. Previous published work has revealed pronounced muscle atrophy together with impaired satellite cell functionality in Smad3-null muscles. In the present study, we have further validated a role for Smad3 signaling in skeletal muscle regeneration. Here, we show that Smad3-null mice had incomplete recovery of muscle weight and myofiber size after muscle injury. Histological/immunohistochemical analysis suggested impaired inflammatory response and reduced number of activated myoblasts during the early stages of muscle regeneration in the tibialis anterior muscle of Smad3-null mice. Nascent myofibers formed after muscle injury were also reduced in number. Moreover, Smad3-null regenerated muscle had decreased oxidative enzyme activity and impaired mitochondrial biogenesis, evident by the downregulation of the gene encoding mitochondrial transcription factor A, a master regulator of mitochondrial biogenesis. Consistent with known Smad3 function, reduced fibrotic tissue formation was also seen in regenerated Smad3-null muscle. In conclusion, Smad3 deficiency leads to impaired muscle regeneration, which underscores an essential role of Smad3 in postnatal myogenesis. Given the negative role of myostatin during muscle regeneration, the increased expression of myostatin observed in Smad3-null muscle may contribute to the regeneration defects.


Assuntos
Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fibrose , Regulação da Expressão Gênica , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias Musculares/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Atrofia Muscular/imunologia , Atrofia Muscular/patologia , Mioblastos Esqueléticos/enzimologia , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Miostatina/genética , Miostatina/metabolismo , Necrose , Infiltração de Neutrófilos , RNA Mensageiro/metabolismo , Células Satélites de Músculo Esquelético/enzimologia , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia , Proteína Smad3/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
NPJ Parkinsons Dis ; 8(1): 159, 2022 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396647

RESUMO

Parkinson's Disease (PD) is a chronic and progressive neurodegenerative disease manifesting itself with tremors, muscle stiffness, bradykinesia, dementia, and depression. Mutations of mitochondrial E3 ligase, PARKIN, have been associated with juvenile PD. Previous studies have characterized muscle atrophy and motor deficits upon loss of functional Parkin in fly and rodent models. However, the mechanisms behind pathophysiology of Parkin deficient muscle remains to be elusive. Here, results suggested that knock down of Parkin significantly increases proteolytic activities in skeletal muscle cell line, the C2C12 myotubes. However, the atrogene levels increase moderately in Parkin deficient cell line. To further investigate the role of Parkin in skeletal muscle atrophy, Parkin knock out (KO) and wild type mice were subjected to 48 h starvation. After 48 h fasting, a greater reduction in skeletal muscle weights was observed in Parkin KO mice as compared to age matched wild type control, suggesting elevated proteolytic activity in the absence of Parkin. Subsequent microarray analyses revealed further enhanced expression of FOXO and ubiquitin pathway in fasted Parkin KO mice. Furthermore, a greater reduction in the expression of cytoskeleton genes was observed in Parkin KO mice following 48 h fasting. Collectively, these results suggest that Parkin deficiency exacerbates fasting-induced skeletal muscle wasting, through upregulating genes involved in catabolic activities in skeletal muscle.

16.
Am J Physiol Cell Physiol ; 301(6): C1316-24, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21900687

RESUMO

Myostatin is a negative regulator of skeletal muscle growth and in fact acts as a potent inducer of "cachectic-like" muscle wasting in mice. The mechanism of action of myostatin in promoting muscle wasting has been predominantly studied in murine models. Despite numerous reports linking elevated levels of myostatin to human skeletal muscle wasting conditions, little is currently known about the signaling mechanism(s) through which myostatin promotes human skeletal muscle wasting. Therefore, in this present study we describe in further detail the mechanisms behind myostatin regulation of human skeletal muscle wasting using an in vitro human primary myotube atrophy model. Treatment of human myotube populations with myostatin promoted dramatic myotubular atrophy. Mechanistically, myostatin-induced myotube atrophy resulted in reduced p-AKT concomitant with the accumulation of active dephosphorylated Forkhead Box-O (FOXO1) and FOXO3. We further show that addition of myostatin results in enhanced activation of atrogin-1 and muscle-specific RING finger protein 1 (MURF1) and reduced expression of both myosin light chain (MYL) and myosin heavy chain (MYH). In addition, we found that myostatin-induced loss of MYL and MYH proteins is dependent on the activity of the proteasome and mediated via SMAD3-dependent regulation of FOXO1 and atrogin-1. Therefore, these data suggest that the mechanism through which myostatin promotes muscle wasting is very well conserved between species, and that myostatin-induced human myotube atrophy is mediated through inhibition of insulin-like growth factor (IGF)/phosphoinositide 3-kinase (PI3-K)/AKT signaling and enhanced activation of the ubiquitin-proteasome pathway and elevated protein degradation.


Assuntos
Proteínas Musculares/metabolismo , Mioblastos/metabolismo , Miostatina/metabolismo , Transdução de Sinais/fisiologia , Síndrome de Emaciação/metabolismo , Western Blotting , Células Cultivadas , Humanos , Imunoprecipitação , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Mioblastos/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Interferente Pequeno/genética , Radioimunoensaio , Sarcômeros/metabolismo , Sarcômeros/patologia , Ubiquitina/metabolismo , Síndrome de Emaciação/patologia
17.
Am J Physiol Cell Physiol ; 301(1): C195-203, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21508334

RESUMO

Myostatin, a member of the transforming growth factor-ß superfamily, has been implicated in the potent negative regulation of myogenesis in murine models. However, little is known about the mechanism(s) through which human myostatin negatively regulates human skeletal muscle growth. Using human primary myoblasts and recombinant human myostatin protein, we show here that myostatin blocks human myoblast proliferation by regulating cell cycle progression through targeted upregulation of p21. We further show that myostatin regulates myogenic differentiation through the inhibition of key myogenic regulatory factors including MyoD, via canonical Smad signaling. In addition, we have for the first time demonstrated the capability of myostatin to regulate the Notch signaling pathway during inhibition of human myoblast differentiation. Treatment with myostatin results in the upregulation of Hes1, Hes5, and Hey1 expression during differentiation; moreover, when we interfere with Notch signaling, through treatment with the γ-secretase inhibitor L-685,458, we find enhanced myotube formation despite the presence of excess myostatin. Therefore, blockade of the Notch pathway relieves myostatin repression of differentiation, and myostatin upregulates Notch downstream target genes. Immunoprecipitation studies demonstrate that myostatin treatment of myoblasts results in enhanced association of Notch1-intracellular domain with Smad3, providing an additional mechanism through which myostatin targets and represses the activity of the myogenic regulatory factor MyoD. On the basis of these results, we suggest that myostatin function and mechanism of action are very well conserved between species, and that myostatin regulation of postnatal myogenesis involves interactions with numerous downstream signaling mediators, including the Notch pathway.


Assuntos
Diferenciação Celular , Músculo Esquelético/crescimento & desenvolvimento , Mioblastos/citologia , Mioblastos/metabolismo , Miostatina/metabolismo , Adolescente , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carbamatos/farmacologia , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Criança , Proteínas de Ligação a DNA/metabolismo , Dipeptídeos/farmacologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imunoprecipitação , Camundongos , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Proteína MyoD/metabolismo , Fatores de Regulação Miogênica/metabolismo , Reação em Cadeia da Polimerase , Receptores Notch/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fatores de Transcrição HES-1 , Fator de Crescimento Transformador beta , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
19.
BMC Genomics ; 12: 509, 2011 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-21999700

RESUMO

BACKGROUND: Although an adverse early-life environment has been linked to an increased risk of developing the metabolic syndrome, the molecular mechanisms underlying altered disease susceptibility as well as their relevance to humans are largely unknown. Importantly, emerging evidence suggests that these effects operate within the normal range of birth weights and involve mechanisms of developmental palsticity rather than pathology. METHOD: To explore this further, we utilised a non-human primate model Macaca fascicularis (Cynomolgus macaque) which shares with humans the same progressive history of the metabolic syndrome. Using microarray we compared tissues from neonates in the average birth weight (50-75th centile) to those of lower birth weight (5-25th centile) and studied the effect of different growth trajectories within the normal range on gene expression levels in the umbilical cord, neonatal liver and skeletal muscle. RESULTS: We identified 1973 genes which were differentially expressed in the three tissue types between average and low birth weight animals (P < 0.05). Gene ontology analysis identified that these genes were involved in metabolic processes including cellular lipid metabolism, cellular biosynthesis, cellular macromolecule synthesis, cellular nitrogen metabolism, cellular carbohydrate metabolism, cellular catabolism, nucleotide and nucleic acid metabolism, regulation of molecular functions, biological adhesion and development. CONCLUSION: These differences in gene expression levels between animals in the upper and lower percentiles of the normal birth weight range may point towards early life metabolic adaptations that in later life result in differences in disease risk.


Assuntos
Peso ao Nascer/genética , Perfilação da Expressão Gênica , Macaca fascicularis/genética , Animais , Animais Recém-Nascidos , Feminino , Fígado/metabolismo , Músculo Esquelético/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Cordão Umbilical/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa