Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Sci Rep ; 13(1): 21349, 2023 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-38049482

RESUMO

Although osteoarthritis (OA) is regarded as a disease of the articular cartilage, recent research has demonstrated alterations in periarticular muscles that surround the affected joint. Here, we investigated changes in periarticular muscle during the progression of OA, as well as the cause-and-effect relationship between muscle weakness and OA, in a mouse model of OA by destabilization of the medial meniscus (DMM). Pathological phenotypes in the periarticular muscles were assessed in the early and late stages of OA by DMM. OA pathology and pain behavior in the mice after DMM induction were examined in response to periarticular muscle weakness induced by multiple rounds of barium chloride (BaCl2) injections. The examinations were also performed in myostatin knockout mice with strengthened muscle phenotypes by muscle hypertrophy. Morphological alterations in the tibialis anterior (TA) and quadriceps muscles in DMM mice included variations in muscle-fiber size, aberrant extracellular matrix (ECM) deposition, inflammatory cell infiltration, and decreased muscle mass. Periarticular muscle fibers isolated from DMM mice showed reductions in the number of satellite cells and myogenic capacity of primary myoblast, as well as proliferation. DMM + muscle injury mice also showed exacerbated joint degeneration compared to the DMM vehicles. Myostatin knockout mice were characterized by attenuated OA and the complete abrogation of pain behavior after DMM. Our results suggest an association between muscle weakness and OA progression and pain.


Assuntos
Cartilagem Articular , Osteoartrite , Camundongos , Animais , Miostatina/farmacologia , Osteoartrite/patologia , Cartilagem Articular/patologia , Camundongos Knockout , Debilidade Muscular/patologia , Modelos Animais de Doenças , Músculo Esquelético/patologia , Dor/etiologia , Dor/patologia
2.
Diabetes Metab Syndr ; 17(10): 102856, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37742361

RESUMO

BACKGROUND AND AIM: Sarcopenic Obesity (SO) in the elderly population is a complex and multifactorial condition which refers to the loss of skeletal muscle mass, strength, and function associated with aging, while obesity involves excessive adipose tissue accumulation. The simultaneous occurrence of these two conditions presents a unique set of challenges to public health and clinical management. This narrative review aims to provide an overview of the use of epicatechin (EC) in the treatment of SO and its related complications. METHOD: A survey of studies related to preclinical and clinical evidence of Epicatechin in sarcopenic obesity and its complications was performed in the following database Medline, Scopus, ProQuest, Embase, Web of Science, and Google scholar. Followed by structural activity relationship and pharmacokinetic profile of Epicatechin was discussed in this paper. RESULTS: The main pharmacological effect of Epicatechin is myostatin inhibition activity which has been described by both in vitro and in vivo studies earlier. The SO is directly correlated with the alteration of Myostatin. The pre-clinical and clinical studies suggest that epicatechin can be a potential candidate in the management of SO and its related complication. CONCLUSION: The present review describes the pharmacokinetic profile and structural activity of epicatechin respective to SO and its related complications. The goal of this review is to update the scientific community on the therapeutic potential of epicatechin in SO and age-related factors. Conduction of clinical and pre-clinical trials, also drug dosage optimization may provide with insights on the use of epicatechin in SO.


Assuntos
Catequina , Sarcopenia , Idoso , Humanos , Sarcopenia/tratamento farmacológico , Sarcopenia/etiologia , Catequina/uso terapêutico , Catequina/farmacologia , Miostatina/farmacologia , Músculo Esquelético/fisiologia , Obesidade/complicações , Obesidade/tratamento farmacológico , Obesidade/epidemiologia
3.
Exp Cell Res ; 432(1): 113779, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37709247

RESUMO

AIM: It has long been recognized that resistance exercise can substantially increase skeletal muscle mass and strength, but whether it can protect against glucocorticoid-induced muscle atrophy and its potential mechanism is yet to be determined. This study aimed to investigate the protective effects of resistance exercise in dexamethasone-induced muscle atrophy and elucidate the possible function of exercise-induced protein Sestrin2 in this process. METHODS: Eight-week-old male C57BL/6J mice carried out the incremental mouse ladder exercise for 11 weeks. Two weeks before the end of the intervention, mice were daily intraperitoneally injected with dexamethasone. Body composition, muscle mass, and exercise performance were examined to evaluate muscle atrophy. In vitro, C2C12 cells were used for RT-qPCR, Western Blot, and immunofluorescence experiments to elucidate the potential mechanism. RESULTS: Our results showed that long-term resistance exercise is an effective intervention for dexamethasone-induced muscle atrophy. We also found that Sestrin2 plays a vital role in dexamethasone-induced muscle atrophy. In both animal (P = .0006) and cell models (P = .0266), dexamethasone intervention significantly reduced the protein expression of Sestrin2, which was increased (P = .0112) by resistance exercise. Inversely, overexpression of Sestrin2 improved (P < .0001) dexamethasone-induced myotube cell atrophy by reducing the activation of the ubiquitin-proteasome pathway via inhibiting Forkhead box O3 (FoxO3a) and myostatin (MSTN)/small mother against decapentaplegic (Smad) signaling pathways. CONCLUSION: Taken together, our results indicated that Sestrin2 may serve as an effective molecule that mimics the protective effect of resistance exercise on dexamethasone-induced muscle atrophy.


Assuntos
Músculo Esquelético , Treinamento de Força , Animais , Masculino , Camundongos , Linhagem Celular , Dexametasona/farmacologia , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/prevenção & controle , Atrofia Muscular/metabolismo , Miostatina/metabolismo , Miostatina/farmacologia , Sestrinas/metabolismo
4.
Actual. osteol ; 19(1): 9-17, ago. 2023. tab
Artigo em Espanhol | LILACS, UNISALUD, BINACIS | ID: biblio-1511347

RESUMO

La sarcopenia asociada a la edad es una condición clínica caracterizada por una disminución en la fuerza, calidad y cantidad de masa muscular así como también en la función muscular. Un biomarcador se define como una característica que es medible objetivamente y evaluable como indicador de un proceso biológico normal, patológico o respuesta terapéutica a una intervención farmacológica. Los marcadores bioquímicos propuestos para el estudio de la sarcopenia pueden ser categorizados en dos grupos. El primero de ellos evalúa el estatus musculoesquelético; este panel de marcadores está formado por miostatina/folistatina, procolágeno aminoterminal tipo III e índice de sarcopenia. El segundo grupo de marcadores bioquímicos evalúa factores causales, para lo cual se sugiere medir el factor de crecimiento insulino-símil tipo 1 (IGF-1), dehidroepiandrosterona (DHEAS), cortisol, facto-res inflamatorios [proteína C reactiva (PCR), interleuquina 6 (IL-6) y factor de necrosis tu-moral (TNF-a)]. Las recomendaciones realiza-das están basadas en la evidencia científica disponible en la actualidad y la disponibilidad de la metodología apropiada para cada uno de los biomarcadores. (AU)


Sarcopenia is a progressive and generalized skeletal muscle disorder defined by decrease in the strength, quality and quantity of muscle mass as well as in muscle function. A biomarker is defined as a feature objectively measured and evaluated as an indicator of a normal biologic process, a pathogenic process or a pharmacologic response to therapeutic intervention. The biochemical markers proposed for the study of sarcopenia may be classified in two groups. The first group evaluates the musculoskeletal status, made up by myostatin/follistatin, N-terminal Type III Procollagen and the sarcopenia index. The second evaluates causal factors, where the measurement of the following is suggested: hormones insulin-like growth factor-1 (IGF-I), dehydroepiandrosterone sulphate (DHEAS), cortisol, inflammatory factors [C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-a (TNF-a)]. The recommendations made are based on scientific evidence currently available and the appropriate methodology availability for each biomarker. (AU)


Assuntos
Humanos , Biomarcadores/metabolismo , Sarcopenia/tratamento farmacológico , Músculos/efeitos dos fármacos , Hormônios Esteroides Gonadais/análise , Pró-Colágeno , Creatinina , Hormônios Peptídicos/análise , Folistatina/farmacologia , Adipocinas/farmacologia , Miostatina/farmacologia , Sarcopenia/diagnóstico , Músculos/metabolismo
5.
Cell Commun Signal ; 21(1): 93, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37143106

RESUMO

BACKGROUND: Extravillous trophoblast (EVT) cell invasion is a tightly regulated process that requires for a normal pregnancy. The epithelial-mesenchymal transition (EMT) has been implicated in EVT cell invasion. Growth differentiation factor-8 (GDF-8), a member of the transforming growth factor-beta (TGF-ß) superfamily, is expressed in the human placenta and promotes EVT cell invasion by upregulating the expression of matrix metalloproteinase 2 (MMP2). However, the underlying molecular mechanism of GDF-8-induced MMP2 expression remains undetermined. Therefore, the present study aims to examine the role of Snail and Slug, the EMT-related transcriptional regulators, in GDF-8-stimulated MMP2 expression and cell invasion in HTR-8/SVneo human EVT cell line and primary cultures of human EVT cells. METHODS: HTR-8/SVneo and primary cultures of human EVT cells were used to examine the effect of GDF-8 on MMP2 expression and explore the underlying mechanism. For gene silencing and overexpression, the HTR-8/SVneo cell line was used to make the experiments more technically feasible. The cell invasiveness was measured by Matrigel-coated transwell invasion assay. RESULTS: GDF-8 stimulated MMP2 expression in both HTR-8/SVneo and primary EVT cells. The stimulatory effect of GDF-8 on MMP2 expression was blocked by the inhibitor of TGF-ß type-I receptors, SB431542. Treatment with GDF-8 upregulated Snail and Slug expression in both HTR-8/SVneo and primary EVT cells. The stimulatory effects of GDF-8 on Snail and Slug expression were blocked by pretreatment of SB431542 and siRNA-mediated knockdown of SMAD4. Interestingly, using the siRNA knockdown approach, our results showed that Snail but not Slug was required for the GDF-8-induced MMP2 expression and cell invasion in HTR-8/SVneo cells. The reduction of MMP2 expression in the placentas with preeclampsia (PE) was also observed. CONCLUSIONS: These findings discover the physiological function of GDF-8 in the human placenta and provide important insights into the regulation of MMP2 expression in human EVT cells. Video Abstract.


Assuntos
Metaloproteinase 2 da Matriz , Trofoblastos , Feminino , Humanos , Gravidez , Movimento Celular , Metaloproteinase 2 da Matriz/metabolismo , Miostatina/metabolismo , Miostatina/farmacologia , RNA Interferente Pequeno/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Trofoblastos/metabolismo
6.
Endocrinol Metab (Seoul) ; 37(4): 684-697, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-36065648

RESUMO

BACKGRUOUND: Muscle atrophy is caused by an imbalance between muscle growth and wasting. Delta-like 1 homolog (DLK1), a protein that modulates adipogenesis and muscle development, is a crucial regulator of myogenic programming. Thus, we investigated the effect of exogenous DLK1 on muscular atrophy. METHODS: We used muscular atrophy mouse model induced by dexamethasone (Dex). The mice were randomly divided into three groups: (1) control group, (2) Dex-induced muscle atrophy group, and (3) Dex-induced muscle atrophy group treated with DLK1. The effects of DLK1 were also investigated in an in vitro model using C2C12 myotubes. RESULTS: Dex-induced muscular atrophy in mice was associated with increased expression of muscle atrophy markers and decreased expression of muscle differentiation markers, while DLK1 treatment attenuated these degenerative changes together with reduced expression of the muscle growth inhibitor, myostatin. In addition, electron microscopy revealed that DLK1 treatment improved mitochondrial dynamics in the Dex-induced atrophy model. In the in vitro model of muscle atrophy, normalized expression of muscle differentiation markers by DLK1 treatment was mitigated by myostatin knockdown, implying that DLK1 attenuates muscle atrophy through the myostatin pathway. CONCLUSION: DLK1 treatment inhibited muscular atrophy by suppressing myostatin-driven signaling and improving mitochondrial biogenesis. Thus, DLK1 might be a promising candidate to treat sarcopenia, characterized by muscle atrophy and degeneration.


Assuntos
Miostatina , Sarcopenia , Animais , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/metabolismo , Atrofia Muscular/prevenção & controle , Miostatina/metabolismo , Miostatina/farmacologia , Sarcopenia/metabolismo , Transdução de Sinais
7.
Ann N Y Acad Sci ; 1517(1): 203-212, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36072988

RESUMO

Citrate is an indispensable component of bone. Reduced levels of citrate in bone and serum are reported in the elderly and in osteoporosis patients. Myostatin (Mstn) is implicated in skeletal homeostasis, but its effects on osteogenesis remain incompletely understood. Nox4 has critical roles in bone homeostasis. TGF-ß/Mstn-associated Smad2/3 signaling has been linked to Nox4 expression. Insulin-like growth factor (IGF-1) has been shown to counteract many regulatory effects of Mstn. However, the crosstalk among Mstn, IGF-1, and Nox4 is not well understood; the interactive effects of those factors on citrate secretion, osteogenic differentiation, and bone remodeling remain unclear. In this study, we demonstrated that osteogenic differentiation induced an IGF-1-dependent upregulation of citrate secretion that was suppressed by Mstn. Inhibition of Nox4 prevented Mstn-induced reduction of citrate secretion. In addition, Mstn reduced bone nodule formation; these changes were prevented by Nox4 inhibition. Moreover, Mstn increased the ratio of RANKL to OPG mRNAs to favor osteoclast activation. These results indicate that Mstn negatively regulates osteogenesis by increasing levels of Nox4, which reduced IGF-1 expression, citrate secretion, and bone mineralization while also altering the RANKL to OPG ratio. These findings provide new and highly relevant insights into the osseous effects of myostatin.


Assuntos
Células-Tronco Mesenquimais , Miostatina , Camundongos , Animais , Miostatina/metabolismo , Miostatina/farmacologia , Fator de Crescimento Insulin-Like I/metabolismo , Osteogênese , NADP/metabolismo , Células-Tronco Mesenquimais/metabolismo , Citratos/metabolismo , Oxirredutases/metabolismo , Músculo Esquelético/metabolismo
8.
Oxid Med Cell Longev ; 2022: 3497644, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35663205

RESUMO

During exercise, the body's organs and skeletal muscles produce reactive oxygen species (ROS). Excessive ROS can destroy cellular lipids, sugars, proteins, and nucleotides and lead to cancer. The production of nicotinamide adenine dinucleotide phosphate (NADPH) by the pentose phosphate pathway (PPP) is an auxiliary process of the cellular antioxidant system that supplements the reducing power of glutathione (GSH) to eliminate ROS in the cell. Myostatin (MSTN) is mainly expressed in skeletal muscle and participates in the regulation of skeletal muscle growth and development. Loss of MSTN leads to muscular hypertrophy, and MSTN deficiency upregulates glycolysis. However, the effect of MSTN on the PPP has not been reported. This study investigated the effect of MSTN on muscle antioxidant capacity from a metabolic perspective. We found that reducing MSTN modulates AMP-activated protein kinase (AMPK), a key molecule in cellular energy metabolism that directly regulates glucose metabolism through phosphorylation. Downregulation of MSTN promotes tyrosine modification of glucose-6-phosphate-dehydrogenase (G6PD) by AMPK and is regulated by the Smad signaling pathway. The Smad2/3 complex acts as a transcription factor to inhibit the AMPK expression. These results suggest that reduced MSTN expression inhibits the Smad signaling pathway, promotes AMPK expression, enhances the activity of G6PD enzyme, and enhances the antioxidant capacity of nonenzymatic GSH.


Assuntos
Proteínas Quinases Ativadas por AMP , Miostatina , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Antioxidantes/metabolismo , Bovinos , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Miostatina/farmacologia , Espécies Reativas de Oxigênio/metabolismo
9.
Biol Res Nurs ; 24(3): 294-307, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35332795

RESUMO

Purpose: We aimed to determine the effects of 12 weeks of soy milk consumption combined with resistance training (RT) on body composition, physical performance, and skeletal muscle regulatory markers in older men. Methods: In this randomized clinical trial study, 60 healthy elderly men (age = 65.63 ± 3.16 years) were randomly assigned to four groups: resistance training (RT; n = 15), soy milk consumption (SMC; n = 15), resistance training + soy milk (RSM; n = 15), and control (CON; n = 15) groups. The study was double-blind for the soy milk/placebo. Participants in RT and RSM groups performed resistance training (3 times/week) for 12 weeks. Participants in the SMC and RSM groups consumed 240 mL of soy milk daily. Body composition [body mass (BM), body fat percent (BFP), waist-hip ratio (WHR), and fat mass (FM)], physical performance [upper body strength (UBS), lower body strength (LBS), VO2max, upper anaerobic power, lower anaerobic power, and handgrip strength], and serum markers [follistatin, myostatin, myostatin-follistatin ratio (MFR), and growth and differentiation factor 11 (GDF11)] were evaluated before and after interventions. Results: All 3 interventions significantly (p < 0.05) increased serum follistatin concentrations (RT = 1.7%, SMC = 2.9%, RSM = 7.8%) and decreased serum myostatin (RT = -1.3% SMC = -5.4%, RSM = -0.5%) and GDF11 concentrations (RT = -1.4%, SMC = -1.4%, RSM = -9.0%), and MFR (RT = -2.6%, SMC = -3.2%, RSM = -12%). In addition, we observed significant reduction in all 3 intervention groups in BFP (RT = -3.6%, SMC = -1.4%, RSM = -6.0%), WHR (RT = -2.2%, SMC = -2.1%, RSM = -4.3%), and FM (RT = -9.6%, SMC = -3.8%, RSM = -11.0%). Moreover, results found significant increase only in RT and RSM groups for muscle mass (RT = 3.8% and RSM = 11.8%), UBS (RT = 10.9% and RSM = 21.8%), LBS (RT = 4.3% and RSM = 7.8%), upper anaerobic power (RT = 7.8% and RSM = 10.3%), and lower anaerobic power (RT = 4.6% and RSM = 8.9%). Handgrip strength were significantly increased in all 3 intervention groups (RT = 7.0%, SMC = 6.9%, RSM = 43.0%). VO2max significantly increased only in RSM (1.7%) after 12 weeks of intervention. Additionally, significant differences were observed between the changes for all variables in the RSM group compared to RT, SMC, and CON groups (p < 0.05). Conclusions: There were synergistic effects of soy milk and RT for skeletal muscle regulatory markers, body composition, and physical performance. Results of the present study support the importance of soy milk in conjunction with RT for older men.


Assuntos
Treinamento de Força , Leite de Soja , Idoso , Biomarcadores , Composição Corporal/fisiologia , Proteínas Morfogenéticas Ósseas/farmacologia , Folistatina/farmacologia , Fatores de Diferenciação de Crescimento/farmacologia , Força da Mão , Humanos , Masculino , Pessoa de Meia-Idade , Força Muscular/fisiologia , Músculo Esquelético/fisiologia , Miostatina/farmacologia , Desempenho Físico Funcional , Treinamento de Força/métodos
10.
Mol Nutr Food Res ; 66(1): e2100549, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34710274

RESUMO

SCOPE: Allulose is shown to increase the muscle weight in diet-induced obese mice. However, there are no studies on the effects of allulose in age-associated sarcopenia. This study aims to elucidate the mechanisms of action for allulose in age associated by analyzing the transcriptional patterns in aged mice. METHODS AND RESULTS: The 48-week-old mice are fed with AIN-93diet containing allulose for 12 weeks. Allulose supplementation increases the muscle mass and grip strength in aged mice. Allulose increases the insulin-like growth factor 1 (IGF-1) and its downstream factor expressions which 40 are related protein synthesis, while inhibits the myostatin expression related protein degradation. In mRNA-seq analysis, allulose supplementation significantly decreases in Adiponectin, Adipsin, cell death inducing DFFA like effector (CIDEC), Haptoglobin, Neuroglobin, and stearoyl-CoA desaturase-1 (SCD1) and increases in cytokine-inducible SH2-containing protein (CISH) and ceramide synthase 1 (CerS1) that are regulate protein turn over in gastrocnemius. Also, allulose alleviates autophagy in muscle with regulated mammalian target of rapamycin (mTOR) signaling pathway and increases the anti-oxidant enzyme activity. CONCLUSION: These findings suggest that allulose improves the age-associated sarcopenia with enhancing antioxidant properties by altering mRNA and protein expression.


Assuntos
Sarcopenia , Animais , Frutose , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Mamíferos , Camundongos , Camundongos Obesos , Músculo Esquelético/metabolismo , Miostatina/genética , Miostatina/metabolismo , Miostatina/farmacologia , Sarcopenia/tratamento farmacológico , Sarcopenia/metabolismo , Sarcopenia/prevenção & controle
11.
Br J Pharmacol ; 179(6): 1237-1250, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34553378

RESUMO

BACKGROUND AND PURPOSE: Duchenne muscular dystrophy (DMD) is a degenerative muscle disease with no effective drug treatment. This study investigated the positive effects of fenofibrate on dystrophic muscles. EXPERIMENTAL APPROACH: Myostatin expression in serum and muscle tissue from patients with Duchenne muscular dystrophy and mdx mice were tested. Primary myoblasts isolated from mdx mice were challenged with an inflammatory stimulus and treated with fenofibrate. In animal experiments, 6-week-old male mdx mice were treated with fenofibrate (100 mg kg-1 ) administered orally once per day for 6 weeks. Effects of fenofibrate were evaluated by tests of muscle function plus histology and biochemical analyses of serum. Expression of myostatin, MuRF1, and atrogin-1 in skeletal muscle was evaluated by western blotting and real-time PCR. Total and oxidative myosin heavy chain (MHC) were assessed via immunofluorescence. KEY RESULTS: Expression of myostatin protein was increased in dystrophic muscle of patients with Duchenne muscular dystrophy and mdx mice. Fenofibrate enhanced myofibre differentiation by down-regulating the expression of myostatin protein but not mRNA in primary myoblasts of mdx mice. Fenofibrate significantly improved muscle function while ameliorating muscle damage in mdx mice. These benefits were accompanied by an anti-inflammatory effect. Fenofibrate treatment returned myofibre function by inhibiting the expressions of myostatin, MuRF1, and atrogin-1 protein in the gastrocnemius muscle and diaphragm, while leaving the mRNA level of myostatin unaffected. CONCLUSIONS AND IMPLICATIONS: Fenofibrate substantially slows muscle dystrophy by promoting the degradation of myostatin protein, which may indicate a new therapeutic focus for patients with Duchenne muscular dystrophy.


Assuntos
Fenofibrato , Distrofia Muscular de Duchenne , Animais , Fenofibrato/farmacologia , Fenofibrato/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Miostatina/metabolismo , Miostatina/farmacologia , Miostatina/uso terapêutico
12.
Pflugers Arch ; 473(6): 969-976, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33895875

RESUMO

Myostatin is a signaling molecule produced by skeletal muscle cells (myokine) that inhibits muscle hypertrophy and has further paracrine and endocrine effects in other organs including bone. Myostatin binds to activin receptor type 2B which forms a complex with transforming growth factor-ß type I receptor (TGF-ßRI) and induces intracellular p38MAPK and NFκB signaling. Fibroblast growth factor 23 (FGF23) is a paracrine and endocrine mediator produced by bone cells and regulates phosphate and vitamin D metabolism in the kidney. P38MAPK and NFκB-dependent store-operated Ca2+ entry (SOCE) are positive regulators of FGF23 production. Here, we explored whether myostatin influences the synthesis of FGF23. Fgf23 gene expression was determined by qRT-PCR and FGF23 protein by ELISA in UMR106 osteoblast-like cells. UMR106 cells expressed activin receptor type 2A and B. Myostatin upregulated Fgf23 gene expression and protein production. The myostatin effect on Fgf23 was significantly attenuated by TGF-ßRI inhibitor SB431542, p38MAPK inhibitor SB202190, and NFκB inhibitor withaferin A. Moreover, SOCE inhibitor 2-APB blunted the myostatin effect on Fgf23. Taken together, myostatin is a stimulator of Fgf23 expression in UMR106 cells, an effect at least partially mediated by downstream TGF-ßRI/p38MAPK signaling as well as NFκB-dependent SOCE.


Assuntos
Fator de Crescimento de Fibroblastos 23/metabolismo , Miostatina/farmacologia , Osteoblastos/metabolismo , Receptores de Ativinas/metabolismo , Animais , Benzamidas/farmacologia , Cálcio/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Dioxóis/farmacologia , Fator de Crescimento de Fibroblastos 23/genética , Imidazóis/farmacologia , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Osteoblastos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Ratos , Vitanolídeos/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Mol Metab ; 43: 101120, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33220490

RESUMO

OBJECTIVE: Increasing muscle mass and activating beige fat both have great potential for ameliorating obesity and its comorbidities. Myostatin null mice have increased skeletal muscle mass and are protected from obesity and its sequelae. Deletion of myostatin has also been suggested to result in the activation of beige adipocytes, thermogenic fat cells with anti-obesity and anti-diabetes properties. It is not known whether beige fat activation contributes to the protection from obesity in myostatin null mice. METHODS: To investigate the role of beige fat activation in the metabolic benefits associated with myostatin deletion, we crossed myostatin null mice to adipocyte-specific PRDM16 knockout mice. We analyzed this new mouse model using molecular profiling, whole mount three-dimensional tissue imaging, tissue respiration, and glucose and insulin tolerance tests in models of diet-induced obesity. RESULTS: Here, we report that PRDM16 is required for the activation of beige fat in the absence of myostatin. However, we show in both male and female mice that beige fat activation is dispensable for the protection from obesity, glucose intolerance, insulin resistance, and hepatic steatosis mediated by myostatin deletion. CONCLUSION: These findings demonstrate that increasing muscle mass can compensate for the inactivation of beige fat and raise the possibility of targeting muscle mass as a therapeutic approach to offset the deleterious effects of adipose tissue dysfunction in obesity and metabolic syndrome.


Assuntos
Tecido Adiposo Bege/metabolismo , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Adipócitos Bege/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Bege/fisiologia , Animais , Regulação da Temperatura Corporal/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fígado Gorduroso/metabolismo , Feminino , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miostatina/genética , Miostatina/farmacologia , Obesidade/metabolismo , Termogênese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Biomolecules ; 10(5)2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32365803

RESUMO

Alongside in vivo models, a simpler and more mechanistic approach is required to study the effects of myostatin on skeletal muscle because myostatin is an important negative regulator of muscle size. In this study, myostatin was administered to murine (C2C12) and human (CHQ) myoblasts and myotubes. Canonical and noncanonical signaling downstream to myostatin, related ligands, and their receptor were analyzed. The effects of tumorkines were analyzed after coculture of C2C12 and colon cancer-C26 cells. The effects of myostatin on canonical and noncanonical signaling were strongly reduced in C2C12 cells after differentiation. This may be explained by increased follistatin, an endogenous blocker of myostatin and altered expression of activin receptor ligands. In contrast, CHQ cells were equally responsive to myostatin, and follistatin remained unaltered. Both myostatin administration and the coculture stimulated pathways associated with inflammation, especially in C2C12 cells. In conclusion, the effects of myostatin on intracellular signaling may be cell line- or organism-specific, and C2C12 myotubes seem to be a nonoptimal in vitro model for investigating the effects of myostatin on canonical and noncanonical signaling in skeletal muscle. This may be due to altered expression of activin receptor ligands and their regulators during muscle cell differentiation.


Assuntos
Diferenciação Celular , Mioblastos/metabolismo , Miostatina/farmacologia , Transdução de Sinais , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Folistatina/metabolismo , Folistatina/farmacologia , Humanos , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citologia , Mioblastos/efeitos dos fármacos
15.
Int J Mol Sci ; 21(10)2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32429150

RESUMO

Myostatin is a negative regulator of muscle cell growth and proliferation. Furthermore, myostatin directly affects the expression of 14q32 microRNAs by binding the 14q32 locus. Direct inhibition of 14q32 microRNA miR-495-3p decreased postinterventional restenosis via inhibition of both vascular smooth muscle cell (VSMC) proliferation and local inflammation. Here, we aimed to investigate the effects of myostatin in a mouse model for postinterventional restenosis. In VSMCs in vitro, myostatin led to the dose-specific downregulation of 14q32 microRNAs miR-433-3p, miR-494-3p, and miR-495-3p. VSMC proliferation was inhibited, where cell migration and viability remained unaffected. In a murine postinterventional restenosis model, myostatin infusion did not decrease restenosis, neointimal area, or lumen stenosis. Myostatin inhibited expression of both proliferation marker PCNA and of 14q32 microRNAs miR-433-3p, miR-494-3p, and miR-495-3p dose-specifically in cuffed femoral arteries. However, 14q32 microRNA expression remained unaffected in macrophages and macrophage activation as well as macrophage influx into lesions were not decreased. In conclusion, myostatin did not affect postinterventional restenosis. Although myostatin inhibits 14q32 microRNA expression and proliferation in VSMCs, myostatin had no effect on macrophage activation and infiltration. Our findings underline that restenosis is driven by both VSMC proliferation and local inflammation. Targeting only one of these components is insufficient to prevent restenosis.


Assuntos
Reestenose Coronária/genética , Regulação da Expressão Gênica , Inflamação/genética , MicroRNAs/genética , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Miostatina/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cromossomos de Mamíferos/genética , Artéria Femoral/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Sci Rep ; 10(1): 4561, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32165710

RESUMO

Insulin resistance is associated with aging in mice and humans. We have previously shown that administration of recombinant GDF11 (rGDF11) to aged mice alters aging phenotypes in the brain, skeletal muscle, and heart. While the closely related protein GDF8 has a role in metabolism, limited data are available on the potential metabolic effects of GDF11 or GDF8 in aging. To determine the metabolic effects of these two ligands, we administered rGDF11 or rGDF8 protein to young or aged mice fed a standard chow diet, short-term high-fat diet (HFD), or long-term HFD. Under nearly all of these diet conditions, administration of exogenous rGDF11 reduced body weight by 3-17% and significantly improved glucose tolerance in aged mice fed a chow (~30% vs. saline) or HF (~50% vs. saline) diet and young mice fed a HFD (~30%). On the other hand, exogenous rGDF8 showed signifcantly lesser effect or no effect at all on glucose tolerance compared to rGDF11, consistent with data demonstrating that GFD11 is a more potent signaling ligand than GDF8. Collectively, our results show that administration of exogenous rGDF11, but not rGDF8, can reduce diet-induced weight gain and improve metabolic homeostasis.


Assuntos
Envelhecimento/metabolismo , Peso Corporal/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/administração & dosagem , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina , Miostatina/administração & dosagem , Envelhecimento/sangue , Envelhecimento/efeitos dos fármacos , Animais , Proteínas Morfogenéticas Ósseas/farmacologia , Metabolismo Energético/efeitos dos fármacos , Fatores de Diferenciação de Crescimento/administração & dosagem , Fatores de Diferenciação de Crescimento/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miostatina/farmacologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
17.
Int J Mol Sci ; 21(3)2020 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-32050585

RESUMO

Myostatin is a myokine that regulates muscle function and mass, producing muscle atrophy. Myostatin induces the degradation of myofibrillar proteins, such as myosin heavy chain or troponin. The main pathway that mediates protein degradation during muscle atrophy is the ubiquitin proteasome system, by increasing the expression of atrogin-1 and MuRF-1. In addition, myostatin activates the NF-κB signaling pathway. Renin-angiotensin system (RAS) also regulates muscle mass. Angiotensin (1-7) (Ang-(1-7)) has anti-atrophic properties in skeletal muscle. In this paper, we evaluated the effect of Ang-(1-7) on muscle atrophy and signaling induced by myostatin. The results show that Ang-(1-7) prevented the decrease of the myotube diameter and myofibrillar protein levels induced by myostatin. Ang-(1-7) also abolished the increase of myostatin-induced reactive oxygen species production, atrogin-1, MuRF-1, and TNF-α gene expressions and NF-κB signaling activation. Ang-(1-7) inhibited the activity mediated by myostatin through Mas receptor, as is demonstrated by the loss of all Ang-(1-7)-induced effects when the Mas receptor antagonist A779 was used. Our results show that the effects of Ang-(1-7) on the myostatin-dependent muscle atrophy and signaling are blocked by MK-2206, an inhibitor of Akt/PKB. Together, these data indicate that Ang-(1-7) inhibited muscle atrophy and signaling induced by myostatin through a mechanism dependent on Mas receptor and Akt/PKB.


Assuntos
Angiotensina I/farmacologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Miostatina/farmacologia , NF-kappa B/metabolismo , Fragmentos de Peptídeos/farmacologia , Transdução de Sinais , Animais , Linhagem Celular , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
18.
Cells ; 9(2)2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32041253

RESUMO

Transforming Growth Factor ß (TGF-ß) is involved in fibrosis as well as the regulation of muscle mass, and contributes to the progressive pathology of muscle wasting disorders. However, little is known regarding the time-dependent signalling of TGF-ß in myoblasts and myotubes, as well as how TGF-ß affects collagen type I expression and the phenotypes of these cells. Here, we assessed effects of TGF-ß on gene expression in C2C12 myoblasts and myotubes after 1, 3, 9, 24 and 48 h treatment. In myoblasts, various myogenic genes were repressed after 9, 24 and 48 h, while in myotubes only a reduction in Myh3 expression was observed. In both myoblasts and myotubes, TGF-ß acutely induced the expression of a subset of genes involved in fibrosis, such as Ctgf and Fgf-2, which was subsequently followed by increased expression of Col1a1. Knockdown of Ctgf and Fgf-2 resulted in a lower Col1a1 expression level. Furthermore, the effects of TGF-ß on myogenic and fibrotic gene expression were more pronounced than those of myostatin, and knockdown of TGF-ß type I receptor Tgfbr1, but not receptor Acvr1b, resulted in a reduction in Ctgf and Col1a1 expression. These results indicate that, during muscle regeneration, TGF-ß induces fibrosis via Tgfbr1 by stimulating the autocrine signalling of Ctgf and Fgf-2.


Assuntos
Colágeno Tipo I/metabolismo , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/efeitos dos fármacos , Mioblastos/efeitos dos fármacos , Miostatina/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fatores de Tempo
19.
Am J Physiol Cell Physiol ; 317(4): C674-C686, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31268780

RESUMO

G protein-coupled receptor kinase 2 (GRK2) is an important protein involved in ß-adrenergic receptor desensitization. In addition, studies have shown GRK2 can modulate different metabolic processes in the cell. For instance, GRK2 has been recently shown to promote mitochondrial biogenesis and increase ATP production. However, the role of GRK2 in skeletal muscle and the signaling mechanisms that regulate GRK2 remain poorly understood. Myostatin is a well-known myokine that has been shown to impair mitochondria function. Here, we have assessed the role of myostatin in regulating GRK2 and the subsequent downstream effect of myostatin regulation of GRK2 on mitochondrial respiration in skeletal muscle. Myostatin treatment promoted the loss of GRK2 protein in myoblasts and myotubes in a time- and dose-dependent manner, which we suggest was through enhanced ubiquitin-mediated protein loss, as treatment with proteasome inhibitors partially rescued myostatin-mediated loss of GRK2 protein. To evaluate the effects of GRK2 on mitochondrial respiration, we generated stable myoblast lines that overexpress GRK2. Stable overexpression of GRK2 resulted in increased mitochondrial content and enhanced mitochondrial/oxidative respiration. Interestingly, although overexpression of GRK2 was unable to prevent myostatin-mediated impairment of mitochondrial respiratory function, elevated levels of GRK2 blocked the increased autophagic flux observed following treatment with myostatin. Overall, our data suggest a novel role for GRK2 in regulating mitochondria mass and mitochondrial respiration in skeletal muscle.


Assuntos
Autofagia/efeitos dos fármacos , Quinase 2 de Receptor Acoplado a Proteína G/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mioblastos/efeitos dos fármacos , Miostatina/farmacologia , Animais , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Camundongos , Mitocôndrias/metabolismo , Células Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Miostatina/metabolismo , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
20.
Physiol Rep ; 7(13): e14152, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31250564

RESUMO

Liver diseases such as non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are characterized by excess hepatic accumulation of lipid droplets and triglycerides which are associated with defective insulin action. Myostatin (Mstn) and adiponectin, secreted by muscle cells and adipocytes, respectively, play important roles in regulating insulin signaling and energy metabolism. The mechanisms underlying the actions of Mstn and adiponectin remain largely unknown. Moreover, the interactions between Mstn and adiponectin in regulating gene expression critical for fatty acid metabolism and insulin action in hepatocytes have not been investigated. The effects of Mstn and AdipoRon, a synthetic adiponectin receptor agonist that is orally active, alone or in combination, on hepatic gene expression and function was investigated. While Mstn increased fatty acid (FA) accumulation and desensitized cellular responses to insulin, AdipoRon protected against Mstn-induced defects in hepatic gene expression and function. In addition, these effects of Mstn were associated with reduced AMPK and PPARα activities which were reversed by AdipoRon. Finally, AdipoRon was able to prevent Mstn-induced activation of the Smad2/3 pathway. These data suggest crosstalk between Mstn-induced Smad2/3 and adiponectin-induced AMPK/PPARα pathways, which may play important roles in the regulation of hepatic gene expression critical for FA metabolism and insulin signaling. In addition, the data suggest that AdipoRon, as an adiponectin receptor agonist, may serve a therapeutic role to reduce the hepatic contribution to the disorders of fat metabolism and insulin action.


Assuntos
Ácidos Graxos/biossíntese , Hepatócitos/efeitos dos fármacos , Insulina/metabolismo , Piperidinas/farmacologia , Quinases Proteína-Quinases Ativadas por AMP , Animais , Linhagem Celular , Ácidos Graxos/genética , Hepatócitos/metabolismo , Camundongos , Miostatina/farmacologia , PPAR alfa/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...