Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 360
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 61(1): 84-97, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26687681

RESUMO

Autophagy, a cellular self-eating mechanism, is important for maintaining cell survival and tissue homeostasis in various stressed conditions. Although the molecular mechanism of autophagy induction has been well studied, how cells terminate autophagy process remains elusive. Here, we show that ULK1, a serine/threonine kinase critical for autophagy initiation, is a substrate of the Cul3-KLHL20 ubiquitin ligase. Upon autophagy induction, ULK1 autophosphorylation facilitates its recruitment to KLHL20 for ubiquitination and proteolysis. This autophagy-stimulated, KLHL20-dependent ULK1 degradation restrains the amplitude and duration of autophagy. Additionally, KLHL20 governs the degradation of ATG13, VPS34, Beclin-1, and ATG14 in prolonged starvation through a direct or indirect mechanism. Impairment of KLHL20-mediated regulation of autophagy dynamics potentiates starvation-induced cell death and aggravates diabetes-associated muscle atrophy. Our study identifies a key role of KLHL20 in autophagy termination by controlling autophagy-dependent turnover of ULK1 and VPS34 complex subunits and reveals the pathophysiological functions of this autophagy termination mechanism.


Assuntos
Autofagia , Proteínas de Transporte/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Culina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Proteínas de Transporte/genética , Classe III de Fosfatidilinositol 3-Quinases/genética , Proteínas Culina/genética , Complicações do Diabetes/enzimologia , Complicações do Diabetes/genética , Complicações do Diabetes/patologia , Retroalimentação Fisiológica , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Atrofia Muscular/enzimologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Proteólise , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Proteínas de Transporte Vesicular/metabolismo
2.
Am J Physiol Cell Physiol ; 322(3): C382-C394, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35044855

RESUMO

Sarcolipin (SLN) is a small regulatory protein that inhibits the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, in that large increases in SLN content are found in response to development, atrophy, overload, and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious as it may reduce muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism that protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fiber-type shifts, and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.


Assuntos
Desenvolvimento Muscular , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimologia , Atrofia Muscular/enzimologia , Distrofias Musculares/enzimologia , Proteolipídeos/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Animais , Sinalização do Cálcio , Humanos , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Atrofia Muscular/patologia , Atrofia Muscular/fisiopatologia , Distrofias Musculares/patologia , Distrofias Musculares/fisiopatologia
3.
Am J Physiol Cell Physiol ; 320(1): C45-C56, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33052072

RESUMO

UBR5 is an E3 ubiquitin ligase positively associated with anabolism, hypertrophy, and recovery from atrophy in skeletal muscle. The precise mechanisms underpinning UBR5's role in the regulation of skeletal muscle mass remain unknown. The present study aimed to elucidate these mechanisms by silencing the UBR5 gene in vivo. To achieve this aim, we electroporated a UBR5-RNAi plasmid into mouse tibialis anterior muscle to investigate the impact of reduced UBR5 on anabolic signaling MEK/ERK/p90RSK and Akt/GSK3ß/p70S6K/4E-BP1/rpS6 pathways. Seven days after UBR5 RNAi electroporation, although reductions in overall muscle mass were not detected, the mean cross-sectional area (CSA) of green fluorescent protein (GFP)-positive fibers were reduced (-9.5%) and the number of large fibers were lower versus the control. Importantly, UBR5-RNAi significantly reduced total RNA, muscle protein synthesis, ERK1/2, Akt, and GSK3ß activity. Although p90RSK phosphorylation significantly increased, total p90RSK protein levels demonstrated a 45% reduction with UBR5-RNAi. Finally, these early events after 7 days of UBR5 knockdown culminated in significant reductions in muscle mass (-4.6%) and larger reductions in fiber CSA (-18.5%) after 30 days. This was associated with increased levels of phosphatase PP2Ac and inappropriate chronic elevation of p70S6K and rpS6 between 7 and 30 days, as well as corresponding reductions in eIF4e. This study demonstrates that UBR5 plays an important role in anabolism/hypertrophy, whereby knockdown of UBR5 culminates in skeletal muscle atrophy.


Assuntos
Metabolismo Energético , Músculo Esquelético/enzimologia , Atrofia Muscular/enzimologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Glicogênio Sintase Quinase 3 beta/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais , Fatores de Tempo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
4.
J Cell Sci ; 132(17)2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31391242

RESUMO

The muscle-specific RING-finger protein MuRF1 (also known as TRIM63) constitutes a bona fide ubiquitin ligase that routes proteins like several different myosin heavy chain proteins (MyHC) to proteasomal degradation during muscle atrophy. In two unbiased screens, we identified DCAF8 as a new MuRF1-binding partner. MuRF1 physically interacts with DCAF8 and both proteins localize to overlapping structures in muscle cells. Importantly, similar to what is seen for MuRF1, DCAF8 levels increase during atrophy, and the downregulation of either protein substantially impedes muscle wasting and MyHC degradation in C2C12 myotubes, a model system for muscle differentiation and atrophy. DCAF proteins typically serve as substrate receptors for cullin 4-type (Cul4) ubiquitin ligases (CRL), and we demonstrate that DCAF8 and MuRF1 associate with the subunits of such a protein complex. Because genetic downregulation of DCAF8 and inhibition of cullin activity also impair myotube atrophy in C2C12 cells, our data imply that the DCAF8 promotes muscle wasting by targeting proteins like MyHC as an integral substrate receptor of a Cul4A-containing ring ubiquitin ligase complex (CRL4A).This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células COS , Proteínas de Transporte , Chlorocebus aethiops , Humanos , Camundongos , Atrofia Muscular/enzimologia , Ratos , Transfecção
5.
Cell Physiol Biochem ; 55(4): 489-504, 2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34416105

RESUMO

BACKGROUND/AIMS: Diaphragm dysfunction with increased reactive oxygen species (ROS) occurs within 72 hrs post-myocardial infarction (MI) in mice and may contribute to loss of inspiratory maximal pressure and endurance in patients. METHODS: We used wild-type (WT) and whole-body Nox4 knockout (Nox4KO) mice to measure diaphragm bundle force in vitro with a force transducer, mitochondrial respiration in isolated fiber bundles with an O2 sensor, mitochondrial ROS by fluorescence, mRNA (RT-PCR) and protein (immunoblot), and fiber size by histology 72 hrs post-MI. RESULTS: MI decreased diaphragm fiber cross-sectional area (CSA) (~15%, p = 0.015) and maximal specific force (10%, p = 0.005), and increased actin carbonylation (5-10%, p = 0.007) in both WT and Nox4KO. Interestingly, MI did not affect diaphragm mRNA abundance of MAFbx/atrogin-1 and MuRF-1 but Nox4KO decreased it by 20-50% (p < 0.01). Regarding the mitochondria, MI and Nox4KO decreased the protein abundance of citrate synthase and subunits of electron transport system (ETS) complexes and increased mitochondrial O2 flux (JO2) and H2O2 emission (JH2O2) normalized to citrate synthase. Mitochondrial electron leak (JH2O2/JO2) in the presence of ADP was lower in Nox4KO and not changed by MI. CONCLUSION: Our study shows that the early phase post-MI causes diaphragm atrophy, contractile dysfunction, sarcomeric actin oxidation, and decreases citrate synthase and subunits of mitochondrial ETS complexes. These factors are potential causes of loss of inspiratory muscle strength and endurance in patients, which likely contribute to the pathophysiology in the early phase post-MI. Whole-body Nox4KO did not prevent the diaphragm abnormalities induced 72 hrs post-MI, suggesting that systemic pharmacological inhibition of Nox4 will not benefit patients in the early phase post-MI.


Assuntos
Diafragma/enzimologia , Mitocôndrias Musculares/enzimologia , Contração Muscular , Atrofia Muscular/enzimologia , Infarto do Miocárdio/enzimologia , NADPH Oxidase 4/deficiência , Animais , Diafragma/patologia , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias Musculares/genética , Mitocôndrias Musculares/patologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , NADPH Oxidase 4/metabolismo
6.
Biochem Biophys Res Commun ; 570: 184-190, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34293592

RESUMO

OBJECTIVE: Inflammation contributes to skeletal muscle atrophy via protein degradation induced by p38 mitogen-activated protein kinase (MAPK) phosphorylation. Meanwhile, pulsed ultrasound irradiation provides the mechanical stimulation to the target tissue, and has been reported to show anti-inflammatory effects. This study investigated the preventive effects of pulsed ultrasound irradiation on muscle atrophy induced by lipopolysaccharide (LPS) in C2C12 myotubes. METHODS: C2C12 myotubes were used in this research. The pulsed ultrasound (a frequency of 3 MHz, duty cycle of 20%, intensity of 0.5 W/cm2) was irradiated to myotube before LPS administration. RESULTS: The LPS increased phosphorylation of p38 MAPK and decreased the myofibril and myosin heavy chain protein (P < 0.05), followed by atrophy in C2C12 myotubes. The pulsed ultrasound irradiation attenuated p38 MAPK phosphorylation and myotube atrophy induced by LPS (P < 0.05). CONCLUSIONS: Pulsed ultrasound irradiation has the preventive effects on inflammation-induced muscle atrophy through inhibiting phosphorylation of p38 MAPK.


Assuntos
Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Atrofia Muscular/enzimologia , Atrofia Muscular/patologia , Ondas Ultrassônicas , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Lipopolissacarídeos , Camundongos , Proteínas Musculares/metabolismo , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/prevenção & controle , Fosforilação , Proteínas Ligases SKP Culina F-Box/metabolismo
7.
Molecules ; 26(2)2021 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-33466753

RESUMO

Skeletal muscle loss is a detrimental side-effect of numerous chronic diseases that dramatically increases mortality and morbidity. The alteration of protein homeostasis is generally due to increased protein breakdown while, protein synthesis may also be down-regulated. The ubiquitin proteasome system (UPS) is a master regulator of skeletal muscle that impacts muscle contractile properties and metabolism through multiple levers like signaling pathways, contractile apparatus degradation, etc. Among the different actors of the UPS, the E3 ubiquitin ligases specifically target key proteins for either degradation or activity modulation, thus controlling both pro-anabolic or pro-catabolic factors. The atrogenes MuRF1/TRIM63 and MAFbx/Atrogin-1 encode for key E3 ligases that target contractile proteins and key actors of protein synthesis respectively. However, several other E3 ligases are involved upstream in the atrophy program, from signal transduction control to modulation of energy balance. Controlling E3 ligases activity is thus a tempting approach for preserving muscle mass. While indirect modulation of E3 ligases may prove beneficial in some situations of muscle atrophy, some drugs directly inhibiting their activity have started to appear. This review summarizes the main signaling pathways involved in muscle atrophy and the E3 ligases implicated, but also the molecules potentially usable for future therapies.


Assuntos
Atrofia Muscular/enzimologia , Atrofia Muscular/fisiopatologia , Biossíntese de Proteínas , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Transdução de Sinais
8.
FASEB J ; 33(6): 7778-7790, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30894018

RESUMO

Cachexia is frequently accompanied by severe metabolic derangements, although the mechanisms responsible for this debilitating condition remain unclear. Pyruvate dehydrogenase kinase (PDK)4, a critical regulator of cellular energetic metabolism, was found elevated in experimental models of cancer, starvation, diabetes, and sepsis. Here we aimed to investigate the link between PDK4 and the changes in muscle size in cancer cachexia. High PDK4 and abnormal energetic metabolism were found in the skeletal muscle of colon-26 tumor hosts, as well as in mice fed a diet enriched in Pirinixic acid, previously shown to increase PDK4 levels. Viral-mediated PDK4 overexpression in myotube cultures was sufficient to promote myofiber shrinkage, consistent with enhanced protein catabolism and mitochondrial abnormalities. On the contrary, blockade of PDK4 was sufficient to restore myotube size in C2C12 cultures exposed to tumor media. Our data support, for the first time, a direct role for PDK4 in promoting cancer-associated muscle metabolic alterations and skeletal muscle atrophy.-Pin, F., Novinger, L. J., Huot, J. R., Harris, R. A., Couch, M. E., O'Connell, T. M., Bonetto, A. PDK4 drives metabolic alterations and muscle atrophy in cancer cachexia.


Assuntos
Caquexia/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Neoplasias/complicações , Piruvato Desidrogenase Quinase de Transferência de Acetil/fisiologia , Animais , Caquexia/etiologia , Linhagem Celular , Masculino , Camundongos , Mitocôndrias Musculares/enzimologia , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/enzimologia , Atrofia Muscular/enzimologia , Oxirredução
9.
J Pathol ; 246(4): 433-446, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30066461

RESUMO

Skeletal muscle atrophy following unloading or immobilization represents a major invalidating event in bedridden patients. Among mechanisms involved in atrophy development, a controversial role is played by neuronal NOS (nNOS; NOS1), whose dysregulation at the protein level and/or subcellular distribution also characterizes other neuromuscular disorders. This study aimed to investigate unloading-induced changes in nNOS before any evidence of myofiber atrophy, using vastus lateralis biopsies obtained from young healthy subjects after a short bed-rest and rat soleus muscles after exposure to short unloading periods. Our results showed that (1) changes in nNOS subcellular distribution using NADPH-diaphorase histochemistry to detect enzyme activity were observed earlier than using immunofluorescence to visualize the protein; (2) loss of active nNOS from the physiological subsarcolemmal localization occurred before myofiber atrophy, i.e. in 8-day bed-rest biopsies and in 6 h-unloaded rat soleus, and was accompanied by increased nNOS activity in the sarcoplasm; (3) nNOS (Nos1) transcript and protein levels decreased significantly in the rat soleus after 6 h and 1 day unloading, respectively, to return to ambulatory levels after 4 and 7 days of unloading, respectively; (4) unloading-induced nNOS redistribution appeared dependent on mitochondrial-derived oxidant species, indirectly measured by tropomyosin disulfide bonds which had increased significantly in the rat soleus already after a 6 h-unloading bout; (5) activity of displaced nNOS molecules is required for translocation of the FoxO3 transcription factor to myofiber nuclei. FoxO3 nuclear localization in rat soleus increased after 6 h unloading (about four-fold the ambulatory level), whereas it did not when nNOS expression and activity were inhibited in vivo before and during 6 h unloading. In conclusion, this study demonstrates that the redistribution of active nNOS molecules from sarcolemma to sarcoplasm not only is ahead of the atrophy of unloaded myofibers, and is induced by increased production of mitochondrial superoxide anion, but also drives FoxO3 activation to initiate muscle atrophy. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Atrofia Muscular/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Estresse Oxidativo , Músculo Quadríceps/enzimologia , Sarcolema/enzimologia , Animais , Repouso em Cama , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Proteína Forkhead Box O3/metabolismo , Voluntários Saudáveis , Elevação dos Membros Posteriores , Humanos , Masculino , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/fisiopatologia , NADP/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Transporte Proteico , Músculo Quadríceps/patologia , Músculo Quadríceps/fisiopatologia , Ratos Wistar , Sarcolema/patologia , Superóxidos/metabolismo , Fatores de Tempo
10.
Biochem J ; 475(14): 2257-2269, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29959184

RESUMO

Signaling molecule phosphatidylinositol 4,5-bisphosphate is produced primarily by phosphatidylinositol 4-phosphate 5-kinase (PIP5K). PIP5K is essential for the development of the human neuronal system, which has been exemplified by a recessive genetic disorder, lethal congenital contractural syndrome type 3, caused by a single aspartate-to-asparagine mutation in the kinase domain of PIP5Kγ. So far, the exact role of this aspartate residue has yet to be elucidated. In this work, we conducted structural, functional and computational studies on a zebrafish PIP5Kα variant with a mutation at the same site. Compared with the structure of the wild-type (WT) protein in the ATP-bound state, the ATP-associating glycine-rich loop of the mutant protein was severely disordered and the temperature factor of ATP was significantly higher. Both observations suggest a greater degree of disorder of the bound ATP, whereas neither the structure of the catalytic site nor the Km toward ATP was substantially affected by the mutation. Microsecond molecular dynamics simulation revealed that negative charge elimination caused by the mutation destabilized the involved hydrogen bonds and affected key electrostatic interactions in the close proximity of ATP. Taken together, our data indicated that the disease-related aspartate residue is a key node in the interaction network crucial for effective ATP binding. This work provides a paradigm of how a subtle but critical structural perturbation caused by a single mutation at the ATP-binding site abolishes the kinase activity, emphasizing that stabilizing substrate in a productive conformational state is crucial for catalysis.


Assuntos
Contratura/enzimologia , Simulação de Dinâmica Molecular , Atrofia Muscular/enzimologia , Mutação , Fosfotransferases (Aceptor do Grupo Álcool)/química , Proteínas de Peixe-Zebra/química , Peixe-Zebra , Trifosfato de Adenosina/química , Trifosfato de Adenosina/genética , Animais , Contratura/genética , Humanos , Atrofia Muscular/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Domínios Proteicos , Proteínas de Peixe-Zebra/genética
11.
Am J Physiol Cell Physiol ; 314(2): C177-C190, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29092819

RESUMO

Protein arginine methyltransferase 1 (PRMT1), PRMT4, and PRMT5 catalyze the methylation of arginine residues on target proteins. Previous work suggests that these enzymes regulate skeletal muscle plasticity. However, the function of PRMTs during disuse-induced muscle remodeling is unknown. The purpose of our study was to determine whether denervation-induced muscle disuse alters PRMT expression and activity in skeletal muscle, as well as to contextualize PRMT biology within the early disuse-evoked events that precede atrophy, which remain largely undefined. Mice were subjected to 6, 12, 24, 72, or 168 h of unilateral hindlimb denervation. Muscle mass decreased by ~30% after 72 or 168 h of neurogenic disuse, depending on muscle fiber type composition. The expression, localization, and activities of PRMT1, PRMT4, and PRMT5 were modified, exhibiting changes in gene expression and activity that were PRMT-specific. Rapid alterations in canonical muscle atrophy signaling such as forkhead box protein O1, muscle RING-finger protein-1, as well as peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) content, AMP-activated protein kinase (AMPK) and p38 mitogen-activated protein kinase, were observed before measurable decrements in muscle mass. Denervation-induced modifications in AMPK-PRMT1 and PGC-1α-PRMT1 binding revealed a novel, putative PRMT1-AMPK-PGC-1α signaling axis in skeletal muscle. Here, PGC-1α-PRMT1 binding was elevated after 6 h of disuse, whereas AMPK-PRMT1 interactions were reduced following 168 h of denervation. Our data suggest that PRMT biology is integral to the mechanisms that precede and initiate skeletal muscle atrophy during conditions of neurogenic disuse. This study furthers our understanding of the role of PRMTs in governing skeletal muscle plasticity.


Assuntos
Plasticidade Celular , Músculo Esquelético/enzimologia , Músculo Esquelético/inervação , Atrofia Muscular/enzimologia , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Denervação Muscular , Músculo Esquelético/patologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fenótipo , Ligação Proteica , Proteína-Arginina N-Metiltransferases/genética , Transdução de Sinais , Fatores de Tempo
12.
Am J Physiol Cell Physiol ; 314(6): C721-C731, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29513566

RESUMO

Unloading-mediated muscle atrophy is associated with increased reactive oxygen species (ROS) production. We previously demonstrated that elevated ubiquitin ligase casitas B-lineage lymphoma-b (Cbl-b) resulted in the loss of muscle volume (Nakao R, Hirasaka K, Goto J, Ishidoh K, Yamada C, Ohno A, Okumura Y, Nonaka I, Yasutomo K, Baldwin KM, Kominami E, Higashibata A, Nagano K, Tanaka K, Yasui N, Mills EM, Takeda S, Nikawa T. Mol Cell Biol 29: 4798-4811, 2009). However, the pathological role of ROS production associated with unloading-mediated muscle atrophy still remains unknown. Here, we showed that the ROS-mediated signal transduction caused by microgravity or its simulation contributes to Cbl-b expression. In L6 myotubes, the assessment of redox status revealed that oxidized glutathione was increased under microgravity conditions, and simulated microgravity caused a burst of ROS, implicating ROS as a critical upstream mediator linking to downstream atrophic signaling. ROS generation activated the ERK1/2 early-growth response protein (Egr)1/2-Cbl-b signaling pathway, an established contributing pathway to muscle volume loss. Interestingly, antioxidant treatments such as N-acetylcysteine and TEMPOL, but not catalase, blocked the clinorotation-mediated activation of ERK1/2. The increased ROS induced transcriptional activity of Egr1 and/or Egr2 to stimulate Cbl-b expression through the ERK1/2 pathway in L6 myoblasts, since treatment with Egr1/2 siRNA and an ERK1/2 inhibitor significantly suppressed clinorotation-induced Cbl-b and Egr expression, respectively. Promoter and gel mobility shift assays revealed that Cbl-b was upregulated via an Egr consensus oxidative responsive element at -110 to -60 bp of the Cbl-b promoter. Together, this indicates that under microgravity conditions, elevated ROS may be a crucial mechanotransducer in skeletal muscle cells, regulating muscle mass through Cbl-b expression activated by the ERK-Egr signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Atrofia Muscular/enzimologia , Mioblastos Esqueléticos/enzimologia , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ausência de Peso , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antioxidantes/farmacologia , Células COS , Chlorocebus aethiops , Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glutationa/metabolismo , Mecanotransdução Celular , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/prevenção & controle , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/patologia , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-cbl/genética , Ratos , Voo Espacial , Fatores de Tempo , Regulação para Cima , Simulação de Ausência de Peso
13.
Am J Physiol Regul Integr Comp Physiol ; 315(2): R429-R433, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29693432

RESUMO

The phosphodiesterase 4 (PDE4)-cAMP pathway plays a predominant role in mediating skeletal muscle proteolysis in burn injury. The present investigations to determine the PDE4 isoform(s) involved in this action revealed that burn injury increased the expression of rat skeletal muscle PDE4B mRNA by sixfold but had little or no effect on expression of other PDE4 isoforms. These observations led us to study the effects of burn in PDE4B knockout (KO) rats. As reported by us previously, burn injury significantly increased extensor digitorum longus (EDL) muscle total and myofibrillar proteolysis in wild-type (WT) rats, but there were no significant effects on either total or myofibrillar protein breakdown in EDL muscle of PDE4B KO rats with burn injury. Moreover, burn injury increased PDE4 activity in the skeletal muscle of WT rats, but this was reduced by >80% in PDE4B KO rats. Also, burn injury decreased skeletal muscle cAMP concentration in WT rats but had no significant effects in the muscles of PDE4B KO rats. Incubation of the EDL muscle of burn-PDE4B KO rats with an inhibitor of the exchange factor directly activated by cAMP, but not with a protein kinase A inhibitor, eliminated the protective effects of PDE4B KO on EDL muscle proteolysis and increased muscle proteolysis to the same extent as in the EDL of burn-WT rats. These novel findings confirm a major role for PDE4B in skeletal muscle proteolysis in burn injury and suggest that an innovative therapy based on PDE4B-selective inhibitors could be developed to treat skeletal muscle cachexia in burn injury without the fear of causing emesis, which is associated with PDE4D inhibition.


Assuntos
Queimaduras/complicações , Caquexia/prevenção & controle , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/deficiência , Músculo Esquelético/enzimologia , Atrofia Muscular/prevenção & controle , Animais , Queimaduras/enzimologia , Queimaduras/genética , Caquexia/enzimologia , Caquexia/etiologia , Caquexia/genética , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Atrofia Muscular/enzimologia , Atrofia Muscular/genética , Proteólise , Ratos Sprague-Dawley , Ratos Transgênicos , Sistemas do Segundo Mensageiro
14.
Int J Mol Sci ; 19(11)2018 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-30424476

RESUMO

Molecular mechanisms that trigger disuse-induced postural muscle atrophy as well as myosin phenotype transformations are poorly studied. This review will summarize the impact of 5' adenosine monophosphate -activated protein kinase (AMPK) activity on mammalian target of rapamycin complex 1 (mTORC1)-signaling, nuclear-cytoplasmic traffic of class IIa histone deacetylases (HDAC), and myosin heavy chain gene expression in mammalian postural muscles (mainly, soleus muscle) under disuse conditions, i.e., withdrawal of weight-bearing from ankle extensors. Based on the current literature and the authors' own experimental data, the present review points out that AMPK plays a key role in the regulation of signaling pathways that determine metabolic, structural, and functional alternations in skeletal muscle fibers under disuse.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Atrofia Muscular/enzimologia , Transtornos Musculares Atróficos/enzimologia , Animais , Metabolismo Energético , Humanos , Atrofia Muscular/patologia , Transtornos Musculares Atróficos/patologia , Transdução de Sinais
15.
J Am Soc Nephrol ; 27(9): 2797-808, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26880799

RESUMO

Muscle proteolysis in CKD is stimulated when the ubiquitin-proteasome system is activated. Serum glucocorticoid-regulated kinase 1 (SGK-1) is involved in skeletal muscle homeostasis, but the role of this protein in CKD-induced muscle wasting is unknown. We found that, compared with muscles from healthy controls, muscles from patients and mice with CKD express low levels of SGK-1. In mice, SGK-1-knockout (SGK-1-KO) induced muscle loss that correlated with increased expression of ubiquitin E3 ligases known to facilitate protein degradation by the ubiquitin-proteasome, and CKD substantially aggravated this response. SGK-1-KO also altered the phosphorylation levels of transcription factors FoxO3a and Smad2/3. In C2C12 muscle cells, expression of dominant negative FoxO3a or knockdown of Smad2/3 suppressed the upregulation of E3 ligases induced by loss of SGK-1. Additionally, SGK-1 overexpression increased the level of phosphorylated N-myc downstream-regulated gene 1 protein, which directly interacted with and suppressed the phosphorylation of Smad2/3. Overexpression of SGK-1 in wild-type mice with CKD had similar effects on the phosphorylation of FoxO3a and Smad2/3 and prevented CKD-induced muscle atrophy. Finally, mechanical stretch of C2C12 muscle cells or treadmill running of wild-type mice with CKD stimulated SGK-1 production, and treadmill running inhibited proteolysis in muscle. These protective responses were absent in SGK-1-KO mice. Thus, SGK-1 could be a mechanical sensor that mediates exercise-induced improvement in muscle wasting stimulated by CKD.


Assuntos
Proteína Forkhead Box O3/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Atrofia Muscular/enzimologia , Atrofia Muscular/etiologia , Proteínas Serina-Treonina Quinases/fisiologia , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/enzimologia , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia , Animais , Camundongos
16.
J Biol Chem ; 290(26): 16202-14, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25944903

RESUMO

FBXO32 (MAFbx/Atrogin-1) is an E3 ubiquitin ligase that is markedly up-regulated in muscle atrophy. Although some data indicate that FBXO32 may play an important role in tumorigenesis, the molecular mechanism of FBXO32 in tumorigenesis has been poorly understood. Here, we present evidence that FBXO32 targets the oncogenic protein c-Myc for ubiquitination and degradation through the proteasome pathway. Phosphorylation of c-Myc at Thr-58 and Ser-62 is dispensable for FBXO32 to induce c-Myc degradation. Mutation of the lysine 326 in c-Myc reduces c-Myc ubiquitination and prevents the c-Myc degradation induced by FBXO32. Furthermore, overexpression of FBXO32 suppresses c-Myc activity and inhibits cell growth, but knockdown of FBXO32 enhances c-Myc activity and promotes cell growth. Finally, we show that FBXO32 is a direct downstream target of c-Myc, highlighting a negative feedback regulation loop between c-Myc and FBXO32. Thus, FBXO32 may function by targeting c-Myc. This work explains the function of FBXO32 and highlights its mechanisms in tumorigenesis.


Assuntos
Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Motivos de Aminoácidos , Animais , Proliferação de Células , Humanos , Camundongos , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/enzimologia , Atrofia Muscular/genética , Fosforilação , Ligação Proteica , Proteólise , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Ligases SKP Culina F-Box/genética , Ubiquitinação
17.
Biochim Biophys Acta ; 1852(3): 490-506, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25496993

RESUMO

Muscle wasting impairs physical performance, increases mortality and reduces medical intervention efficacy in chronic diseases and cancer. Developing proficient intervention strategies requires improved understanding of the molecular mechanisms governing muscle mass wasting and recovery. Involvement of muscle protein- and myonuclear turnover during recovery from muscle atrophy has received limited attention. The insulin-like growth factor (IGF)-I signaling pathway has been implicated in muscle mass regulation. As glycogen synthase kinase 3 (GSK-3) is inhibited by IGF-I signaling, we hypothesized that muscle-specific GSK-3ß deletion facilitates the recovery of disuse-atrophied skeletal muscle. Wild-type mice and mice lacking muscle GSK-3ß (MGSK-3ß KO) were subjected to a hindlimb suspension model of reversible disuse-induced muscle atrophy and followed during recovery. Indices of muscle mass, protein synthesis and proteolysis, and post-natal myogenesis which contribute to myonuclear accretion, were monitored during the reloading of atrophied muscle. Early muscle mass recovery occurred more rapidly in MGSK-3ß KO muscle. Reloading-associated changes in muscle protein turnover were not affected by GSK-3ß ablation. However, coherent effects were observed in the extent and kinetics of satellite cell activation, proliferation and myogenic differentiation observed during reloading, suggestive of increased myonuclear accretion in regenerating skeletal muscle lacking GSK-3ß. This study demonstrates that muscle mass recovery and post-natal myogenesis from disuse-atrophy are accelerated in the absence of GSK-3ß.


Assuntos
Diferenciação Celular , Quinase 3 da Glicogênio Sintase/metabolismo , Desenvolvimento Muscular , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiologia , Atrofia Muscular/enzimologia , Regeneração , Animais , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/fisiopatologia
18.
Arch Biochem Biophys ; 600: 56-60, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27136709

RESUMO

AMP-activated protein kinase (AMPK) is a master regulator of skeletal muscle metabolic pathways. Recently, AMPK activation by AICAR has been shown to increase myofibrillar protein degradation in C2C12 myotubes via stimulating autophagy and ubiquitin proteasome system. However, the impact of AMPKα on denervation induced muscle atrophy has not been tested. In this study, we performed sciatic denervation on hind limb muscles in both wild type (WT) and AMPKα2(-/-) mice. We found that AMPKα was phosphorylated in atrophic muscles following denervation. In addition, deletion of AMPKα2 significantly attenuated denervation induced skeletal muscle wasting and protein degradation, as evidenced by preserved muscle mass and myofiber area, as well as lower levels of ubiquitinated protein, Atrogin-1 and MuRF-1 expression, and LC3-II/I ratio in tibial anterior (TA) muscles. Interestingly, the phosphorylated FoxO3a at Ser253 was significantly decreased in atrophic TA muscles, which was preserved in AMPKα2(-/-) mice. Collectively, our data support the notion that the activation of AMPKα2 contributes to the atrophic effects of denervation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Denervação Muscular , Atrofia Muscular/enzimologia , Atrofia Muscular/prevenção & controle , Proteínas Quinases Ativadas por AMP/genética , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Atrofia Muscular/patologia
19.
Am J Physiol Endocrinol Metab ; 308(2): E144-58, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25406264

RESUMO

Skeletal muscle atrophy is a common and debilitating condition that remains poorly understood at the molecular level. To better understand the mechanisms of muscle atrophy, we used mouse models to search for a skeletal muscle protein that helps to maintain muscle mass and is specifically lost during muscle atrophy. We discovered that diverse causes of muscle atrophy (limb immobilization, fasting, muscle denervation, and aging) strongly reduced expression of the enzyme spermine oxidase. Importantly, a reduction in spermine oxidase was sufficient to induce muscle fiber atrophy. Conversely, forced expression of spermine oxidase increased muscle fiber size in multiple models of muscle atrophy (immobilization, fasting, and denervation). Interestingly, the reduction of spermine oxidase during muscle atrophy was mediated by p21, a protein that is highly induced during muscle atrophy and actively promotes muscle atrophy. In addition, we found that spermine oxidase decreased skeletal muscle mRNAs that promote muscle atrophy (e.g., myogenin) and increased mRNAs that help to maintain muscle mass (e.g., mitofusin-2). Thus, in healthy skeletal muscle, a relatively low level of p21 permits expression of spermine oxidase, which helps to maintain basal muscle gene expression and fiber size; conversely, during conditions that cause muscle atrophy, p21 expression rises, leading to reduced spermine oxidase expression, disruption of basal muscle gene expression, and muscle fiber atrophy. Collectively, these results identify spermine oxidase as an important positive regulator of muscle gene expression and fiber size, and elucidate p21-mediated repression of spermine oxidase as a key step in the pathogenesis of skeletal muscle atrophy.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/metabolismo , Miogenina/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Animais , Jejum/fisiologia , GTP Fosfo-Hidrolases/genética , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Denervação Muscular , Fibras Musculares Esqueléticas/enzimologia , Atrofia Muscular/enzimologia , Atrofia Muscular/genética , Miogenina/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Restrição Física/fisiologia , Poliamina Oxidase
20.
Am J Physiol Heart Circ Physiol ; 308(9): H1039-50, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25724497

RESUMO

Microcirculatory dysfunction may cause tissue malperfusion and progression to organ failure in the later stages of sepsis, but the role of smooth muscle contractile dysfunction is uncertain. Mice were given intraperitoneal LPS, and mesenteric arteries were harvested at 6-h intervals for analyses of gene expression and contractile function by wire myography. Contractile (myosin and actin) and regulatory [myosin light chain kinase and phosphatase subunits (Mypt1, CPI-17)] mRNAs and proteins were decreased in mesenteric arteries at 24 h concordant with reduced force generation to depolarization, Ca(2+), and phenylephrine. Vasodilator sensitivity to DEA/nitric oxide (NO) and cGMP under Ca(2+) clamp were increased at 24 h after LPS concordant with a switch to Mypt1 exon 24- splice variant coding for a leucine zipper (LZ) motif required for PKG-1α activation of myosin phosphatase. This was reproduced by smooth muscle-specific deletion of Mypt1 exon 24, causing a shift to the Mypt1 LZ+ isoform. These mice had significantly lower resting blood pressure than control mice but similar hypotensive responses to LPS. The vasodilator sensitivity of wild-type mice to DEA/NO, but not cGMP, was increased at 6 h after LPS. This was abrogated in mice with a redox dead version of PKG-1α (Cys42Ser). Enhanced vasorelaxation in early endotoxemia is mediated by redox signaling through PKG-1α but in later endotoxemia by myosin phosphatase isoform shifts enhancing sensitivity to NO/cGMP as well as smooth muscle atrophy. Muscle atrophy and modulation may be a novel target to suppress microcirculatory dysfunction; however, inactivation of inducible NO synthase, treatment with the IL-1 antagonist IL-1ra, or early activation of α-adrenergic signaling did not suppressed this response.


Assuntos
Lipopolissacarídeos , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimologia , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Óxido Nítrico/metabolismo , Fosfoproteínas/metabolismo , Sepse/enzimologia , Transdução de Sinais , Vasodilatação , Animais , GMP Cíclico/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I/deficiência , Proteína Quinase Dependente de GMP Cíclico Tipo I/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Genótipo , Peptídeos e Proteínas de Sinalização Intracelular , Isoenzimas , Masculino , Artérias Mesentéricas/enzimologia , Artérias Mesentéricas/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação , Proteínas Musculares/genética , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiopatologia , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/enzimologia , Atrofia Muscular/fisiopatologia , Quinase de Cadeia Leve de Miosina/deficiência , Quinase de Cadeia Leve de Miosina/genética , Fosfatase de Miosina-de-Cadeia-Leve/genética , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Oxirredução , Fenótipo , Fosfoproteínas/genética , RNA Mensageiro/metabolismo , Sepse/induzido quimicamente , Sepse/genética , Sepse/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA