Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Rev ; 103(4): 2679-2757, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37382939

RESUMO

Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.


Assuntos
Músculo Esquelético , Transdução de Sinais , Humanos , Animais , Cães , Músculo Esquelético/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Biossíntese de Proteínas , Hipertrofia/metabolismo , Mamíferos/metabolismo
2.
BMC Biol ; 21(1): 287, 2023 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-38066609

RESUMO

Physical activity and several pharmacological approaches individually combat age-associated conditions and extend healthy longevity in model systems. It is tantalizing to extrapolate that combining geroprotector drugs with exercise could extend healthy longevity beyond any individual treatment. However, the current dogma suggests that taking leading geroprotector drugs on the same day as exercise may limit several health benefits. Here, we review leading candidate geroprotector drugs and their interactions with exercise and highlight salient gaps in knowledge that need to be addressed to identify if geroprotector drugs can have a harmonious relationship with exercise.


Assuntos
Longevidade , Senoterapia , Humanos , Exercício Físico , Envelhecimento
3.
J Physiol ; 599(11): 2887-2906, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33873245

RESUMO

KEY POINTS: A decrease in protein synthesis plays a major role in the loss of muscle mass that occurs in response to immobilization. In mice, immobilization leads to a rapid (within 6 h) and progressive decrease in the rate of protein synthesis and this effect is mediated by a decrease in translational efficiency. Deep proteomic and phosphoproteomic analyses of mouse skeletal muscles revealed that the rapid immobilization-induced decrease in protein synthesis cannot be explained by changes in the abundance or phosphorylation state of proteins that have been implicated in the regulation of translation. ABSTRACT: The disuse of skeletal muscle, such as that which occurs during immobilization, can lead to the rapid loss of muscle mass, and a decrease in the rate of protein synthesis plays a major role in this process. Indeed, current dogma contends that the decrease in protein synthesis is mediated by changes in the activity of protein kinases (e.g. mTOR); however, the validity of this model has not been established. Therefore, to address this, we first subjected mice to 6, 24 or 72 h of unilateral immobilization and then used the SUnSET technique to measure changes in the relative rate of protein synthesis. The result of our initial experiments revealed that immobilization leads to a rapid (within 6 h) and progressive decrease in the rate of protein synthesis and that this effect is mediated by a decrease in translational efficiency. We then performed a deep mass spectrometry-based analysis to determine whether this effect could be explained by changes in the expression and/or phosphorylation state of proteins that regulate translation. From this analysis, we were able to quantify 4320 proteins and 15,020 unique phosphorylation sites, and surprisingly, the outcomes revealed that the rapid immobilization-induced decrease in protein synthesis could not be explained by changes in either the abundance, or phosphorylation state, of proteins. The results of our work not only challenge the current dogma in the field, but also provide an expansive resource of information for future studies that are aimed at defining how disuse leads to loss of muscle mass.


Assuntos
Atrofia Muscular , Proteômica , Animais , Imobilização , Camundongos , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Fosforilação
4.
Cell Physiol Biochem ; 55(1): 91-116, 2021 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-33543862

RESUMO

BACKGROUND/AIMS: Signaling and metabolic perturbations contribute to dysregulated skeletal muscle protein homeostasis and secondary sarcopenia in response to a number of cellular stressors including ethanol exposure. Using an innovative multiomics-based curating of unbiased data, we identified molecular and metabolic therapeutic targets and experimentally validated restoration of protein homeostasis in an ethanol-fed mouse model of liver disease. METHODS: Studies were performed in ethanol-treated differentiated C2C12 myotubes and physiological relevance established in an ethanol-fed mouse model of alcohol-related liver disease (mALD) or pair-fed control C57BL/6 mice. Transcriptome and proteome from ethanol treated-myotubes and gastrocnemius muscle from mALD and pair-fed mice were analyzed to identify target pathways and molecules. Readouts including signaling responses and autophagy markers by immunoblots, mitochondrial oxidative function and free radical generation, and metabolic studies by gas chromatography-mass spectrometry and sarcopenic phenotype by imaging. RESULTS: Multiomics analyses showed that ethanol impaired skeletal muscle mTORC1 signaling, mitochondrial oxidative pathways, including intermediary metabolite regulatory genes, interleukin-6, and amino acid degradation pathways are ß-hydroxymethyl-butyrate targets. Ethanol decreased mTORC1 signaling, increased autophagy flux, impaired mitochondrial oxidative function with decreased tricarboxylic acid cycle intermediary metabolites, ATP synthesis, protein synthesis and myotube diameter that were reversed by HMB. Consistently, skeletal muscle from mALD had decreased mTORC1 signaling, reduced fractional and total muscle protein synthesis rates, increased autophagy markers, lower intermediary metabolite concentrations, and lower muscle mass and fiber diameter that were reversed by ß-hydroxymethyl-butyrate treatment. CONCLUSION: An innovative multiomics approach followed by experimental validation showed that ß-hydroxymethyl-butyrate restores muscle protein homeostasis in liver disease.


Assuntos
Etanol/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Hidroxibutiratos/farmacologia , Hepatopatias Alcoólicas , Deficiências na Proteostase , Sarcopenia , Transdução de Sinais/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Etanol/farmacologia , Feminino , Genômica , Hepatopatias Alcoólicas/complicações , Hepatopatias Alcoólicas/tratamento farmacológico , Hepatopatias Alcoólicas/metabolismo , Hepatopatias Alcoólicas/patologia , Camundongos , Deficiências na Proteostase/dietoterapia , Deficiências na Proteostase/etiologia , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia , Sarcopenia/tratamento farmacológico , Sarcopenia/etiologia , Sarcopenia/metabolismo , Sarcopenia/patologia
5.
FASEB J ; 33(3): 4021-4034, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30509128

RESUMO

It is well known that an increase in mechanical loading can induce skeletal muscle hypertrophy, and a long standing model in the field indicates that mechanical loads induce hypertrophy via a mechanism that requires signaling through the mechanistic target of rapamycin complex 1 (mTORC1). Specifically, it has been widely proposed that mechanical loads activate signaling through mTORC1 and that this, in turn, promotes an increase in the rate of protein synthesis and the subsequent hypertrophic response. However, this model is based on a number of important assumptions that have not been rigorously tested. In this study, we created skeletal muscle specific and inducible raptor knockout mice to eliminate signaling by mTORC1, and with these mice we were able to directly demonstrate that mechanical stimuli can activate signaling by mTORC1, and that mTORC1 is necessary for mechanical load-induced hypertrophy. Surprisingly, however, we also obtained multiple lines of evidence that indicate that mTORC1 is not required for a mechanical load-induced increase in the rate of protein synthesis. This observation highlights an important shortcoming in our understanding of how mechanical loads induce hypertrophy and illustrates that additional mTORC1-independent mechanisms play a critical role in this process.-You, J.-S., McNally, R. M., Jacobs, B. L., Privett, R. E., Gundermann, D. M., Lin, K.-H., Steinert, N. D., Goodman, C. A., Hornberger, T. A. The role of raptor in the mechanical load-induced regulation of mTOR signaling, protein synthesis, and skeletal muscle hypertrophy.


Assuntos
Músculo Esquelético/metabolismo , Esforço Físico , Proteína Regulatória Associada a mTOR/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Hipertrofia/etiologia , Hipertrofia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Biossíntese de Proteínas , Proteína Regulatória Associada a mTOR/genética , Transdução de Sinais
6.
Exerc Sport Sci Rev ; 47(3): 188-194, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30870215

RESUMO

The mechanistic target of rapamycin (mTOR) exerts both rapamycin-sensitive and rapamycin-insensitive signaling events, and the rapamycin-sensitive components of mTOR signaling have been widely implicated in the pathway through which resistance exercise induces skeletal muscle hypertrophy. This review explores the hypothesis that rapamycin-insensitive components of mTOR signaling also contribute to this highly important process.


Assuntos
Músculo Esquelético/anatomia & histologia , Músculo Esquelético/metabolismo , Treinamento Resistido , Serina-Treonina Quinases TOR/fisiologia , Humanos , Proteínas Musculares/biossíntese , Proteólise , Transdução de Sinais
7.
J Biol Chem ; 292(17): 6987-6997, 2017 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-28289099

RESUMO

Mechanistic target of rapamycin (mTOR) signaling is necessary to generate a mechanically induced increase in skeletal muscle mass, but the mechanism(s) through which mechanical stimuli regulate mTOR signaling remain poorly defined. Recent studies have suggested that Ras homologue enriched in brain (Rheb), a direct activator of mTOR, and its inhibitor, the GTPase-activating protein tuberin (TSC2), may play a role in this pathway. To address this possibility, we generated inducible and skeletal muscle-specific knock-out mice for Rheb (iRhebKO) and TSC2 (iTSC2KO) and mechanically stimulated muscles from these mice with eccentric contractions (EC). As expected, the knock-out of TSC2 led to an elevation in the basal level of mTOR signaling. Moreover, we found that the magnitude of the EC-induced activation of mTOR signaling was significantly blunted in muscles from both inducible and skeletal muscle-specific knock-out mice for Rheb and iTSC2KO mice. Using mass spectrometry, we identified six sites on TSC2 whose phosphorylation was significantly altered by the EC treatment. Employing a transient transfection-based approach to rescue TSC2 function in muscles of the iTSC2KO mice, we demonstrated that these phosphorylation sites are required for the role that TSC2 plays in the EC-induced activation of mTOR signaling. Importantly, however, these phosphorylation sites were not required for an insulin-induced activation of mTOR signaling. As such, our results not only establish a critical role for Rheb and TSC2 in the mechanical activation of mTOR signaling, but they also expose the existence of a previously unknown branch of signaling events that can regulate the TSC2/mTOR pathway.


Assuntos
Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Encéfalo/metabolismo , Feminino , Homozigoto , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Monoméricas de Ligação ao GTP/genética , Contração Muscular , Músculo Esquelético/metabolismo , Neuropeptídeos/genética , Fosforilação , Plasmídeos/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Sirolimo/química , Tamoxifeno/química , Proteína 2 do Complexo Esclerose Tuberosa
8.
Exerc Sport Sci Rev ; 46(2): 76-85, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29346157

RESUMO

We propose that phosphoproteomic-based studies will radically advance our knowledge about exercise-regulated signaling events. However, these studies use cutting-edge technologies that can be difficult for nonspecialists to understand. Hence, this review is intended to help nonspecialists 1) understand the fundamental technologies behind phosphoproteomic analysis and 2) use various bioinformatic tools that can be used to interrogate phosphoproteomic datasets.


Assuntos
Exercício Físico/fisiologia , Proteômica , Transdução de Sinais , Biologia Computacional , Conjuntos de Dados como Assunto , Humanos , Espectrometria de Massas , Fosforilação
10.
J Physiol ; 595(15): 5209-5226, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28542873

RESUMO

KEY POINTS: Mechanical signals play a critical role in the regulation of muscle mass, but the molecules that sense mechanical signals and convert this stimulus into the biochemical events that regulate muscle mass remain ill-defined. Here we report a mass spectrometry-based workflow to study the changes in protein phosphorylation that occur in mouse skeletal muscle 1 h after a bout of electrically evoked maximal-intensity contractions (MICs). Our dataset provides the first comprehensive map of the MIC-regulated phosphoproteome. Using unbiased bioinformatics approaches, we demonstrate that our dataset leads to the identification of many well-known MIC-regulated signalling pathways, as well as to a plethora of novel MIC-regulated events. We expect that our dataset will serve as a fundamentally important resource for muscle biologists, and help to lay the foundation for entirely new hypotheses in the field. ABSTRACT: The maintenance of skeletal muscle mass is essential for health and quality of life. It is well recognized that maximal-intensity contractions, such as those which occur during resistance exercise, promote an increase in muscle mass. Yet, the molecules that sense the mechanical information and convert it into the signalling events (e.g. phosphorylation) that drive the increase in muscle mass remain undefined. Here we describe a phosphoproteomics workflow to examine the effects of electrically evoked maximal-intensity contractions (MICs) on protein phosphorylation in mouse skeletal muscle. While a preliminary phosphoproteomics experiment successfully identified a number of MIC-regulated phosphorylation events, a large proportion of these identifications were present on highly abundant myofibrillar proteins. We subsequently incorporated a centrifugation-based fractionation step to deplete the highly abundant myofibrillar proteins and performed a second phosphoproteomics experiment. In total, we identified 5983 unique phosphorylation sites of which 663 were found to be regulated by MIC. GO term enrichment, phosphorylation motif analyses, and kinase-substrate predictions indicated that the MIC-regulated phosphorylation sites were chiefly modified by mTOR, as well as multiple isoforms of the MAPKs and CAMKs. Moreover, a high proportion of the regulated phosphorylation sites were found on proteins that are associated with the Z-disc, with over 74% of the Z-disc proteins experiencing robust changes in phosphorylation. Finally, our analyses revealed that the phosphorylation state of two Z-disc kinases (striated muscle-specific serine/threonine protein kinase and obscurin) was dramatically altered by MIC, and we propose ways these kinases could play a fundamental role in skeletal muscle mechanotransduction.


Assuntos
Contração Muscular/fisiologia , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Animais , Estimulação Elétrica , Masculino , Espectrometria de Massas , Camundongos Endogâmicos C57BL , Fosforilação , Proteômica
11.
J Cell Sci ; 128(2): 219-24, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25413344

RESUMO

A fundamental requirement of cells is their ability to transduce and interpret their mechanical environment. This ability contributes to regulation of growth, differentiation and adaptation in many cell types. The intermediate filament (IF) system not only provides passive structural support to the cell, but recent evidence points to IF involvement in active biological processes such as signaling, mechanotransduction and gene regulation. However, the mechanisms that underlie these processes are not well known. Skeletal muscle cells provide a convenient system to understand IF function because the major muscle-specific IF, desmin, is expressed in high abundance and is highly organized. Here, we show that desmin plays both structural and regulatory roles in muscle cells by demonstrating that desmin is required for the maintenance of myofibrillar alignment, nuclear deformation, stress production and JNK-mediated stress sensing. Finite element modeling of the muscle IF system suggests that desmin immediately below the sarcolemma is the most functionally significant. This demonstration of biomechanical integration by the desmin IF system suggests that it plays an active biological role in muscle in addition to its accepted structural role.


Assuntos
Desmina/metabolismo , Filamentos Intermediários/metabolismo , Músculo Esquelético/metabolismo , Miofibrilas/metabolismo , Animais , Desmina/genética , Humanos , Filamentos Intermediários/ultraestrutura , Mecanotransdução Celular/genética , Camundongos Knockout , Músculo Esquelético/ultraestrutura , Miofibrilas/ultraestrutura , Sarcolema/genética , Sarcolema/metabolismo , Estresse Mecânico
12.
Proc Natl Acad Sci U S A ; 111(44): 15756-61, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25336758

RESUMO

Peroxisome proliferator-activated receptor gamma coactivator 1-alpha 4 (PGC-1α4) is a protein isoform derived by alternative splicing of the PGC1α mRNA and has been shown to promote muscle hypertrophy. We show here that G protein-coupled receptor 56 (GPR56) is a transcriptional target of PGC-1α4 and is induced in humans by resistance exercise. Furthermore, the anabolic effects of PGC-1α4 in cultured murine muscle cells are dependent on GPR56 signaling, because knockdown of GPR56 attenuates PGC-1α4-induced muscle hypertrophy in vitro. Forced expression of GPR56 results in myotube hypertrophy through the expression of insulin-like growth factor 1, which is dependent on Gα12/13 signaling. A murine model of overload-induced muscle hypertrophy is associated with increased expression of both GPR56 and its ligand collagen type III, whereas genetic ablation of GPR56 expression attenuates overload-induced muscle hypertrophy and associated anabolic signaling. These data illustrate a signaling pathway through GPR56 which regulates muscle hypertrophy associated with resistance/loading-type exercise.


Assuntos
Regulação da Expressão Gênica/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Condicionamento Físico Animal , Receptores Acoplados a Proteínas G/biossíntese , Transdução de Sinais/fisiologia , Animais , Colágeno Tipo III/biossíntese , Hipertrofia/metabolismo , Fator de Crescimento Insulin-Like I/biossíntese , Masculino , Camundongos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/metabolismo
13.
FASEB J ; 29(10): 4092-106, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26178167

RESUMO

Prolonged immobilization (IM) causes skeletal muscle atrophy characterized by mitochondrial deterioration and proteolysis. Muscle remobilization (RM) increases reactive oxygen species generation, proinflammatory cytokine expression, and oxidative stress, preventing muscle from quick recovery. Thus, we hypothesized that overexpression of peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) via in vivo transfection would promote mitochondrial biogenesis and antioxidant defense, thus ameliorating the aforementioned deteriorations in a mouse model with 14-d IM followed by 5-d RM. PGC-1α transfection in tibialis anterior muscle resulted in a 7.2- and 4-fold increase in PGC-1α content in cytosol and nucleus, respectively. Mitochondrial biogenic (cytochrome c, mitochondrial transcription factor A), morphologic (mitochondrial density, mDNA/nDNA ratio), and functional (cytochrome c oxidase activity, ATP synthesis rate) markers, as well as fiber cross-sectional area, significantly increased in IM-RM muscle by PGC-1α overexpression. These effects were accompanied by an 18% decrease in H2O2, 30% decrease in nuclear factor-κB-DNA binding, and 25% reduction of IL-1ß and-6 production in IM-RM muscle. There was a 34% increase in superoxide dismutase-2 activity, along with a 3.5-fold increase in NAD-dependent deacetylase sirtuin-3 expression caused by enhanced PGC-1α-estrogen-related receptor α binding. Our findings highlighted the importance of PGC-1α in protecting muscle from metabolic and redox disturbances caused by IM.


Assuntos
Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Fatores de Transcrição/metabolismo , Animais , Western Blotting , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Feminino , Expressão Gênica , Mediadores da Inflamação/metabolismo , Camundongos , Microscopia Eletrônica , Mitocôndrias/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/fisiopatologia , Atrofia Muscular/etiologia , Atrofia Muscular/genética , Oxirredução , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Espécies Reativas de Oxigênio/metabolismo , Restrição Física/efeitos adversos , Restrição Física/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxido Dismutase/metabolismo , Fatores de Tempo , Fatores de Transcrição/genética , Transfecção
14.
J Biol Chem ; 289(3): 1551-63, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24302719

RESUMO

The activation of mTOR signaling is essential for mechanically induced changes in skeletal muscle mass, and previous studies have suggested that mechanical stimuli activate mTOR (mammalian target of rapamycin) signaling through a phospholipase D (PLD)-dependent increase in the concentration of phosphatidic acid (PA). Consistent with this conclusion, we obtained evidence which further suggests that mechanical stimuli utilize PA as a direct upstream activator of mTOR signaling. Unexpectedly though, we found that the activation of PLD is not necessary for the mechanically induced increases in PA or mTOR signaling. Motivated by this observation, we performed experiments that were aimed at identifying the enzyme(s) that promotes the increase in PA. These experiments revealed that mechanical stimulation increases the concentration of diacylglycerol (DAG) and the activity of DAG kinases (DGKs) in membranous structures. Furthermore, using knock-out mice, we determined that the ζ isoform of DGK (DGKζ) is necessary for the mechanically induced increase in PA. We also determined that DGKζ significantly contributes to the mechanical activation of mTOR signaling, and this is likely driven by an enhanced binding of PA to mTOR. Last, we found that the overexpression of DGKζ is sufficient to induce muscle fiber hypertrophy through an mTOR-dependent mechanism, and this event requires DGKζ kinase activity (i.e. the synthesis of PA). Combined, these results indicate that DGKζ, but not PLD, plays an important role in mechanically induced increases in PA and mTOR signaling. Furthermore, this study suggests that DGKζ could be a fundamental component of the mechanism(s) through which mechanical stimuli regulate skeletal muscle mass.


Assuntos
Diacilglicerol Quinase/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Ácidos Fosfatídicos/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Diacilglicerol Quinase/genética , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/patologia , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Músculo Esquelético/patologia , Tamanho do Órgão/genética , Ácidos Fosfatídicos/genética , Serina-Treonina Quinases TOR/genética
15.
J Exp Biol ; 218(Pt 2): 276-84, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25452506

RESUMO

Mammalian hibernators provide an extreme example of naturally occurring challenges to muscle homeostasis. The annual hibernation cycle is characterized by shifts between summer euthermy with tissue anabolism and accumulation of body fat reserves, and winter heterothermy with fasting and tissue catabolism. The circannual patterns of skeletal muscle remodelling must accommodate extended inactivity during winter torpor, the motor requirements of transient winter active periods, and sustained activity following spring emergence. Muscle volume in thirteen-lined ground squirrels (Ictidomys tridecemlineatus) calculated from MRI upper hindlimb images (n=6 squirrels, n=10 serial scans) declined from hibernation onset, reaching a nadir in early February. Paradoxically, mean muscle volume rose sharply after February despite ongoing hibernation, and continued total body mass decline until April. Correspondingly, the ratio of muscle volume to body mass was steady during winter atrophy (October-February) but increased (+70%) from February to May, which significantly outpaced changes in liver or kidney examined by the same method. Generally stable myocyte cross-sectional area and density indicated that muscle remodelling is well regulated in this hibernator, despite vastly altered seasonal fuel and activity levels. Body composition analysis by echo MRI showed lean tissue preservation throughout hibernation amid declining fat mass by the end of winter. Muscle protein synthesis was 66% depressed in early but not late winter compared with a summer fasted baseline, while no significant changes were observed in the heart, liver or intestine, providing evidence that could support a transition in skeletal muscle regulation between early and late winter, prior to spring emergence and re-feeding.


Assuntos
Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Sciuridae/fisiologia , Animais , Peso Corporal , Feminino , Hibernação/fisiologia , Membro Posterior , Masculino , Proteínas Musculares/análise , Músculo Esquelético/crescimento & desenvolvimento , Atrofia Muscular , Biossíntese de Proteínas , Sciuridae/crescimento & desenvolvimento , Estações do Ano
16.
J Muscle Res Cell Motil ; 35(1): 11-21, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24162376

RESUMO

It is well recognized that mechanical signals play a critical role in the regulation of skeletal muscle mass, and the maintenance of muscle mass is essential for mobility, disease prevention and quality of life. Furthermore, over the last 15 years it has become established that signaling through a protein kinase called the mammalian (or mechanistic) target of rapamycin (mTOR) is essential for mechanically-induced changes in protein synthesis and muscle mass, however, the mechanism(s) via which mechanical stimuli regulate mTOR signaling have not been defined. Nonetheless, advancements are being made, and an emerging body of evidence suggests that the late endosome/lysosomal (LEL) system might play a key role in this process. Therefore, the purpose of this review is to summarize this body of evidence. Specifically, we will first explain why the Ras homologue enriched in brain (Rheb) and phosphatidic acid (PA) are considered to be direct activators of mTOR signaling. We will then describe the process of endocytosis and its involvement in the formation of LEL structures, as well as the evidence which indicates that mTOR and its direct activators (Rheb and PA) are all enriched at the LEL. Finally, we will summarize the evidence that has implicated the LEL in the regulation of mTOR by various growth regulatory inputs such as amino acids, growth factors and mechanical stimuli.


Assuntos
Endossomos/metabolismo , Músculo Esquelético/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Humanos , Biossíntese de Proteínas , Transdução de Sinais
18.
Elife ; 122024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38466320

RESUMO

An increase in mechanical loading, such as that which occurs during resistance exercise, induces radial growth of muscle fibers (i.e. an increase in cross-sectional area). Muscle fibers are largely composed of myofibrils, but whether radial growth is mediated by an increase in the size of the myofibrils (i.e. myofibril hypertrophy) and/or the number of myofibrils (i.e. myofibrillogenesis) is not known. Electron microscopy (EM) can provide images with the level of resolution that is needed to address this question, but the acquisition and subsequent analysis of EM images is a time- and cost-intensive process. To overcome this, we developed a novel method for visualizing myofibrils with a standard fluorescence microscope (fluorescence imaging of myofibrils with image deconvolution [FIM-ID]). Images from FIM-ID have a high degree of resolution and contrast, and these properties enabled us to develop pipelines for automated measurements of myofibril size and number. After extensively validating the automated measurements, we used both mouse and human models of increased mechanical loading to discover that the radial growth of muscle fibers is largely mediated by myofibrillogenesis. Collectively, the outcomes of this study offer insight into a fundamentally important topic in the field of muscle growth and provide future investigators with a time- and cost-effective means to study it.


Approximately 45% of human body mass is made of skeletal muscle. These muscles contract and relax to provide the mechanical forces needed for breathing, moving, keeping warm and performing many other essential processes. Both sedentary and active adults lose approximately 30-40% of this muscle mass by the age of 80, increasing their risk of disease, disability and death. As a result, there is much interest in developing therapies that can restore, maintain and increase muscle mass in older individuals. Muscles are made of multiple fibers that are in turn largely composed of smaller units known as myofibrils. Previous studies have shown that performing resistance training or other exercise that increases the mechanical loads placed on muscles stimulates muscle growth. This growth is largely due to increased girth of the existing muscle fibers. However, it remained unclear whether this was due to myofibrils growing in size, increasing in number, or a combination of both. To address this question, Jorgenson et al. developed a fluorescence imaging method called FIM-ID to count the number and measure the size of myofibrils within cross-sections of skeletal muscle. Using FIM-ID to study samples of mouse and human muscle fibers then revealed that increasing mechanical loads on muscles increased the number of myofibrils and this was largely responsible for muscle fiber growth. FIM-ID mostly relies on common laboratory instruments and free open-source software is used to count and measure the myofibrils. Jorgenson et al. hope that this will allow as many other researchers as possible to use FIM-ID to study myofibrils in the future. A better understanding of how the body controls the number of myofibrils may lead to the development of therapies that can mimic the effects of exercise on muscles to maintain or even increase muscle mass in human patients.


Assuntos
Músculo Esquelético , Miofibrilas , Humanos , Animais , Camundongos , Fibras Musculares Esqueléticas , Hipertrofia , Imagem Óptica
19.
J Physiol ; 591(18): 4611-20, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23732640

RESUMO

The goal of this study was to determine whether the mechanical activation of mechanistic target of rapamycin (mTOR) signalling is associated with changes in phosphorylation of tuberous sclerosis complex-2 (TSC2) and targeting of mTOR and TSC2 to the lysosome. As a source of mechanical stimulation, mouse skeletal muscles were subjected to eccentric contractions (ECs). The results demonstrated that ECs induced hyper-phosphorylation of TSC2 and at least part of this increase occurred on residue(s) that fall within RxRxxS/T consensus motif(s). Furthermore, in control muscles, we found that both mTOR and TSC2 are highly enriched at the lysosome. Intriguingly, ECs enhanced the lysosomal association of mTOR and almost completely abolished the lysosomal association of TSC2. Based on these results, we developed a new model that could potentially explain how mechanical stimuli activate mTOR signalling. Furthermore, this is the first study to reveal that the activation of mTOR is associated with the translocation of TSC2 away from the lysosome. Since a large number of signalling pathways rely on TSC2 to control mTOR signalling, our results have potentially revealed a fundamental mechanism via which not only mechanical, but also various other types of stimuli, control mTOR signalling.


Assuntos
Lisossomos/metabolismo , Contração Muscular , Músculo Esquelético/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/fisiologia , Fosforilação , Transporte Proteico , Transdução de Sinais , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/química
20.
Exerc Sport Sci Rev ; 41(2): 107-15, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23089927

RESUMO

Protein synthesis rates commonly are measured using isotopic tracers to quantify the incorporation of a labeled amino acid into muscle proteins. Here we provide evidence supporting our hypothesis that the nonisotopic SUnSET technique is a valid and accurate method for the measurement of in vivo changes in protein synthesis at the whole-muscle and single-muscle fiber levels.


Assuntos
Proteínas Musculares/biossíntese , Músculo Esquelético/metabolismo , Inibidores da Síntese de Proteínas , Puromicina , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA