Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 27(5): 1990-2000, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23388382

RESUMO

Skeletal muscle remodeling in response to muscle disuse and unloading is known to be associated with so-called ER stress, which, in turn, activates autophagy and contributes to muscle atrophy. Different molecules are involved in ER stress-induced autophagy, among which PKCθ has recently been described. In this study, we dissected both in vitro and in vivo ER stress-induced autophagy pathways in muscle. Using C2C12 muscle cells in culture, we demonstrated that PKC activation induced autophagy in the absence of ER stress. We further demonstrated that PKCθ was strongly activated in cultured myoblasts and myotubes during ER stress induced by different stimuli, such as TG or TN treatment, and that it localized into Lc3-positive autophagic dots upon TG treatment. Neither Akt dephosphorylation nor Foxo or GSK3ß activation was observed in these conditions. Moreover, PKCθ inhibition in myoblasts and myotubes prevented ER stress-induced Lc3 activation and autophagic dot formation, but not ER stress. In vivo, lack of PKCθ prevented both food deprivation- and immobilization-induced autophagy and muscle atrophy, irrespective of Akt pathway inhibition. Taken together, these results demonstrate that PKCθ functions as an ER stress sensor in skeletal muscle, required for ER-stress-dependent autophagy activation, and can be proposed as a novel molecular target to maintain muscle homeostasis in response to external stimuli, such as disuse and unloading, still allowing intracellular clearance.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Transdução de Sinais/fisiologia , Animais , Autofagia/fisiologia , Linhagem Celular , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Privação de Alimentos/fisiologia , Isoenzimas/metabolismo , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Atrofia Muscular/fisiopatologia , Mioblastos/metabolismo , Mioblastos/patologia , Proteína Quinase C/metabolismo , Proteína Quinase C-theta , Proteínas Proto-Oncogênicas c-akt/fisiologia , Tapsigargina/farmacologia
2.
Neurol Res ; 30(2): 207-14, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18397614

RESUMO

The production of engineered three-dimensional (3D) skeletal muscle grafts holds promise for treatment of several diseases. An important factor in the development of such approach involves the capability of preserving myogenicity and regenerative potential during ex vivo culturing. We have previously shown that electrical stimulation of myogenic cells grown in monolayer could improve the differentiation process. Here we investigated the effect of exogenous electrical field, specifically designed to mimic part of the neuronal activity, on muscle precursor cells (MPCs) cultured within 3D collagen scaffolds. Our data showed that electric stimulation did not affect cell viability and increased by 65.6% the release rate of NO(x), an early molecular activator of satellite cells in vivo. NO(x) release rate was decreased by an inhibitor of NO synthase, both in stimulated and non-stimulated cultures, confirming the endocrine origin of the measured NO(x). Importantly, electrical stimulation also increased the expression of two myogenic markers, MyoD and desmin. We also carried out some preliminary experiments aimed at determining the biocompatibility of our seeded collagen scaffolds, implanting them in the tibialis anterior muscles of syngeneic mice. Ten days after transplantation, we could observe the formation of new myofibers both inside the scaffold and at the scaffold/muscle interface. Altogether, our findings indicate that electrical stimulation could be a new strategy for the effective 3D expansion of muscle precursor cells in vitro without losing myogenic potential and that 3D collagen matrices could be a promising tool for delivering myogenic cells in recipient muscles.


Assuntos
Colágeno/fisiologia , Músculo Esquelético/fisiologia , Mioblastos/fisiologia , Engenharia Tecidual , Análise de Variância , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Desmina/metabolismo , Estimulação Elétrica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína MyoD/metabolismo , Mioblastos/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/metabolismo , Transplante de Tecidos/métodos
3.
Autophagy ; 10(11): 1883-94, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483961

RESUMO

Physical activity has been recently documented to play a fundamental physiological role in the regulation of autophagy in several tissues. It has also been reported that autophagy is required for exercise itself and for training-induced adaptations in glucose homeostasis. These autophagy-mediated metabolic improvements are thought to be largely dependent on the activation of the metabolic sensor PRKAA1/AMPK. However, it is unknown whether these important benefits stem from systemic adaptations or are due solely to alterations in skeletal muscle metabolism. To address this we utilized inducible, muscle-specific, atg7 knockout mice that we have recently generated. Our findings indicate that acute inhibition of autophagy in skeletal muscle just prior to exercise does not have an impact on physical performance, PRKAA1 activation, or glucose homeostasis. However, we reveal that autophagy is critical for the preservation of mitochondrial function during damaging muscle contraction. This effect appears to be gender specific affecting primarily females. We also establish that basal oxidative stress plays a crucial role in mitochondrial maintenance during normal physical activity. Therefore, autophagy is an adaptive response to exercise that ensures effective mitochondrial quality control during damaging physical activity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/fisiologia , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/fisiologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Antioxidantes/química , Proteína 7 Relacionada à Autofagia , Feminino , Glucose/metabolismo , Homeostase , Masculino , Camundongos , Camundongos Knockout , Contração Muscular , Estresse Oxidativo , Condicionamento Físico Animal
4.
Autophagy ; 10(1): 123-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24220501

RESUMO

Removal of ubiquitinated targets by autophagosomes can be mediated by receptor molecules, like SQSTM1, in a mechanism referred to as selective autophagy. While cytoplasmic protein aggregates, mitochondria, and bacteria are the best-known targets of selective autophagy, their role in the turnover of membrane receptors is scarce. We here showed that fasting-induced wasting of skeletal muscle involves remodeling of the neuromuscular junction (NMJ) by increasing the turnover of muscle-type CHRN (cholinergic receptor, nicotinic/nicotinic acetylcholine receptor) in a TRIM63-dependent manner. Notably, this process implied enhanced production of endo/lysosomal carriers of CHRN, which also contained the membrane remodeler SH3GLB1, the E3 ubiquitin ligase, TRIM63, and the selective autophagy receptor SQSTM1. Furthermore, these vesicles were surrounded by the autophagic marker MAP1LC3A in an ATG7-dependent fashion, and some of them were also positive for the lysosomal marker, LAMP1. While the amount of vesicles containing endocytosed CHRN strongly augmented in the absence of ATG7 as well as upon denervation as a model for long-term atrophy, denervation-induced increase in autophagic CHRN vesicles was completely blunted in the absence of TRIM63. On a similar note, in trim63(-/-) mice denervation-induced upregulation of SQSTM1 and LC3-II was abolished and endogenous SQSTM1 did not colocalize with CHRN vesicles as it did in the wild type. SQSTM1 and LC3-II coprecipitated with surface-labeled/endocytosed CHRN and SQSTM1 overexpression significantly induced CHRN vesicle formation. Taken together, our data suggested that selective autophagy regulates the basal and atrophy-induced turnover of the pentameric transmembrane protein, CHRN, and that TRIM63, together with SH3GLB1 and SQSTM1 regulate this process.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Proteínas de Choque Térmico/metabolismo , Proteínas Musculares/metabolismo , Receptores Nicotínicos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Aminoácidos/deficiência , Animais , Biomarcadores/metabolismo , Endocitose , Endossomos/metabolismo , Jejum , Imunofluorescência , Marcação por Isótopo , Lisossomos/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Denervação Muscular , Músculos/inervação , Músculos/metabolismo , Músculos/patologia , Junção Neuromuscular/metabolismo , Fagossomos/metabolismo , Estabilidade Proteica , Proteína Sequestossoma-1 , Sinapses/metabolismo , Proteínas com Motivo Tripartido , Regulação para Cima
5.
Cell Rep ; 8(5): 1509-21, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25176656

RESUMO

The cellular basis of age-related tissue deterioration remains largely obscure. The ability to activate compensatory mechanisms in response to environmental stress is an important factor for survival and maintenance of cellular functions. Autophagy is activated both under short and prolonged stress and is required to clear the cell of dysfunctional organelles and altered proteins. We report that specific autophagy inhibition in muscle has a major impact on neuromuscular synaptic function and, consequently, on muscle strength, ultimately affecting the lifespan of animals. Inhibition of autophagy also exacerbates aging phenotypes in muscle, such as mitochondrial dysfunction, oxidative stress, and profound weakness. Mitochondrial dysfunction and oxidative stress directly affect acto-myosin interaction and force generation but show a limited effect on stability of neuromuscular synapses. These results demonstrate that age-related deterioration of synaptic structure and function is exacerbated by defective autophagy.


Assuntos
Envelhecimento , Autofagia , Músculo Esquelético/metabolismo , Junção Neuromuscular/metabolismo , Actinas/metabolismo , Animais , Proteína 7 Relacionada à Autofagia , Linhagem Celular , Humanos , Longevidade , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias Musculares/metabolismo , Força Muscular , Músculo Esquelético/fisiologia , Miosinas/metabolismo , Junção Neuromuscular/ultraestrutura , Estresse Oxidativo
6.
Skelet Muscle ; 4(1): 22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25530838

RESUMO

BACKGROUND: Nitric oxide (NO), generated in skeletal muscle mostly by the neuronal NO synthases (nNOSµ), has profound effects on both mitochondrial bioenergetics and muscle development and function. The importance of NO for muscle repair emerges from the observation that nNOS signalling is defective in many genetically diverse skeletal muscle diseases in which muscle repair is dysregulated. How the effects of NO/nNOSµ on mitochondria impact on muscle function, however, has not been investigated yet. METHODS: In this study we have examined the relationship between the NO system, mitochondrial structure/activity and skeletal muscle phenotype/growth/functions using a mouse model in which nNOSµ is absent. Also, NO-induced effects and the NO pathway were dissected in myogenic precursor cells. RESULTS: We show that nNOSµ deficiency in mouse skeletal muscle leads to altered mitochondrial bioenergetics and network remodelling, and increased mitochondrial unfolded protein response (UPR(mt)) and autophagy. The absence of nNOSµ is also accompanied by an altered mitochondrial homeostasis in myogenic precursor cells with a decrease in the number of myonuclei per fibre and impaired muscle development at early stages of perinatal growth. No alterations were observed, however, in the overall resting muscle structure, apart from a reduced specific muscle mass and cross sectional areas of the myofibres. Investigating the molecular mechanisms we found that nNOSµ deficiency was associated with an inhibition of the Akt-mammalian target of rapamycin pathway. Concomitantly, the Akt-FoxO3-mitochondrial E3 ubiquitin protein ligase 1 (Mul-1) axis was also dysregulated. In particular, inhibition of nNOS/NO/cyclic guanosine monophosphate (cGMP)/cGMP-dependent-protein kinases induced the transcriptional activity of FoxO3 and increased Mul-1 expression. nNOSµ deficiency was also accompanied by functional changes in muscle with reduced muscle force, decreased resistance to fatigue and increased degeneration/damage post-exercise. CONCLUSIONS: Our results indicate that nNOSµ/NO is required to regulate key homeostatic mechanisms in skeletal muscle, namely mitochondrial bioenergetics and network remodelling, UPR(mt) and autophagy. These events are likely associated with nNOSµ-dependent impairments of muscle fibre growth resulting in a deficit of muscle performance.

7.
J Tissue Eng Regen Med ; 5(1): 1-10, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20607681

RESUMO

Duchenne muscular dystrophy (DMD) is caused by the lack of dystrophin; affected muscles are characterized by continuous bouts of muscle degeneration, eventually leading to the exhaustion of the endogenous satellite cell pool. At present, only palliative treatments are available, although several gene and cell therapy-based approaches are being studied. In this study we proposed to overcome the limitations hampering intramuscular cell injection by using a biomaterial-based strategy. In particular, we used a three-dimensional (3D) collagen porous scaffold to deliver myogenic precursor cells (MPCs) in vivo in the mdx murine model of DMD. MPCs, derived from single fibres of wild-type donors, were expanded in vitro, seeded onto collagen scaffolds and implanted into the tibialis anterior muscles of normal and mdx mice. As a control, cells were delivered via direct intramuscular cell injection in the contralateral muscles. Scaffold-delivered MPCs displayed lower apoptosis and higher proliferation than injected cells; in terms of dystrophin restoration, collagen scaffolds yielded better results than direct injections. Importantly, time-course experiments indicated that the scaffolds acted as a cell reservoir, although cell migration was mostly contained within 400 µm from the scaffold-host tissue interface.


Assuntos
Distrofia Muscular Animal/terapia , Transplante de Células-Tronco/métodos , Alicerces Teciduais/química , Animais , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/farmacologia , Distrofina/metabolismo , Imuno-Histoquímica , Implantes Experimentais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Porosidade/efeitos dos fármacos , Fatores de Tempo
8.
J Cell Biol ; 191(1): 101-13, 2010 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-20921137

RESUMO

The size of skeletal muscle cells is precisely regulated by intracellular signaling networks that determine the balance between overall rates of protein synthesis and degradation. Myofiber growth and protein synthesis are stimulated by the IGF-1/Akt/mammalian target of rapamycin (mTOR) pathway. In this study, we show that the transcription factor JunB is also a major determinant of whether adult muscles grow or atrophy. We found that in atrophying myotubes, JunB is excluded from the nucleus and that decreasing JunB expression by RNA interference in adult muscles causes atrophy. Furthermore, JunB overexpression induces hypertrophy without affecting satellite cell proliferation and stimulated protein synthesis independently of the Akt/mTOR pathway. When JunB is transfected into denervated muscles, fiber atrophy is prevented. JunB blocks FoxO3 binding to atrogin-1 and MuRF-1 promoters and thus reduces protein breakdown. Therefore, JunB is important not only in dividing populations but also in adult muscle, where it is required for the maintenance of muscle size and can induce rapid hypertrophy and block atrophy.


Assuntos
Músculo Esquelético/citologia , Proteínas Proto-Oncogênicas c-jun/fisiologia , Animais , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Hipertrofia , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Atrofia Muscular/metabolismo , Regiões Promotoras Genéticas , Biossíntese de Proteínas , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA