Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Article in English | MEDLINE | ID: mdl-35010726

ABSTRACT

The primary role of myostatin is to negatively regulate skeletal muscle growth. The gait speed is a noninvasive, reliable parameter that predicts cardiovascular risk and mortality. This study evaluated the relationship between serum myostatin concentrations and gait speeds in patients who had undergone kidney transplantation (KT). A total of 84 KT recipients were evaluated. A speed of less than 1.0 m/s was categorized into the low gait speed group. We measured serum myostatin concentrations with a commercial enzyme-linked immunosorbent assay. KT recipients in the low gait speed group had significantly older age, as well as higher body weight, body mass index (BMI), skeletal muscle index, serum triglyceride levels, glucose levels, and blood urea nitrogen levels, lower estimated glomerular filtration rates and serum myostatin levels, a higher percentage of steroid use, and a lower proportion of mycophenolate mofetil use. Multivariable logistic regression analysis revealed that lower myostatin levels and lower frequency of mycophenolate mofetil use were independently associated with low gait speed. In multivariable stepwise linear regression analysis, myostatin levels were positively correlated with gait speeds, and age and BMI were negatively correlated with gait speeds. In the study, serum myostatin levels were significantly lower in the low gait speed group. Subjects in the low gait speed group also had greater BMI and older age.


Subject(s)
Kidney Transplantation , Myostatin , Walking Speed , Aged , Body Mass Index , Humans , Muscle, Skeletal , Myostatin/biosynthesis
2.
Endocrinology ; 162(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34265069

ABSTRACT

Cancer-induced skeletal muscle defects show sex-specific differences in severity with men performing poorly compared to women. Hormones and sex chromosomal differences are suggested to mediate these differences, but the functional skeletal muscle markers to document these differences are unknown. We show that the myogenic microRNA miR-486 is a marker of sex-specific differences in cancer-induced skeletal muscle defects. Cancer-induced loss of circulating miR-486 was more severe in men with bladder, lung, and pancreatic cancers compared to women with the same cancer types. In a syngeneic model of pancreatic cancer, circulating and skeletal muscle loss of miR-486 was more severe in male mice compared to female mice. Estradiol (E2) and the clinically used selective estrogen receptor modulator toremifene increased miR-486 in undifferentiated and differentiated myoblast cell line C2C12 and E2-inducible expression correlated with direct binding of estrogen receptor alpha (ERα) to the regulatory region of the miR-486 gene. E2 and toremifene reduced the actions of cytokines such as myostatin, transforming growth factor ß, and tumor necrosis factor α, which mediate cancer-induced skeletal muscle wasting. E2- and toremifene-treated C2C12 myoblast/myotube cells contained elevated levels of active protein kinase B (AKT) with a corresponding decrease in the levels of its negative regulator PTEN, which is a target of miR-486. We propose an ERα:E2-miR-486-AKT signaling axis, which reduces the deleterious effects of cancer-induced cytokines/chemokines on skeletal muscle mass and/or function.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Neoplasms/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Estradiol/pharmacology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscular Diseases/complications , Myostatin/biosynthesis , Neoplasms/complications , Sex Factors , Signal Transduction , Toremifene/pharmacology , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
3.
Front Endocrinol (Lausanne) ; 12: 653179, 2021.
Article in English | MEDLINE | ID: mdl-33897620

ABSTRACT

Obesity is a global health problem and a major risk factor for several metabolic conditions including dyslipidemia, diabetes, insulin resistance and cardiovascular diseases. Obesity develops from chronic imbalance between energy intake and energy expenditure. Stimulation of cellular energy burning process has the potential to dissipate excess calories in the form of heat via the activation of uncoupling protein-1 (UCP1) in white and brown adipose tissues. Recent studies have shown that activation of transforming growth factor-ß (TGF-ß) signaling pathway significantly contributes to the development of obesity, and blockade or inhibition is reported to protect from obesity by promoting white adipose browning and increasing mitochondrial biogenesis. Identification of novel compounds that activate beige/brown adipose characteristics to burn surplus calories and reduce excess storage of fat are actively sought in the fight against obesity. In this review, we present recent developments in our understanding of key modulators of TGF-ß signaling pathways including follistatin (FST) and myostatin (MST) in regulating adipose browning and brown adipose mass and activity. While MST is a key ligand for TGF-ß family, FST can bind and regulate biological activity of several TGF-ß superfamily members including activins, bone morphogenic proteins (BMP) and inhibins. Here, we review the literature supporting the critical roles for FST, MST and other proteins in modulating TGF-ß signaling to influence beige and brown adipose characteristics. We further review the potential therapeutic utility of FST for the treatment of obesity and related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Follistatin/biosynthesis , Metabolic Diseases/metabolism , Myostatin/biosynthesis , Obesity/metabolism , Transforming Growth Factor beta1/metabolism , Adipose Tissue, Beige/metabolism , Animals , Energy Metabolism , Fibronectins/biosynthesis , Follistatin/metabolism , Follistatin-Related Proteins/biosynthesis , Humans , Ligands , Mice , Signal Transduction , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism
4.
Arch Biochem Biophys ; 692: 108511, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32710883

ABSTRACT

(-)-Epigallocatechin-3-gallate (EGCG), the most abundant catechin component in green tea, has been reported to attenuate age-associated insulin resistance, lipogenesis and loss of muscle mass through restoring Akt activity in skeletal muscle in our previous and present studies. Accumulated data has suggested that polyphenols regulate signaling pathways involved in aging process such as inflammation and oxidative stress via modulation of miRNA expression. Here we found that miRNA-486-5p was significantly decreased in both aged senescence accelerated mouse-prone 8 (SAMP8) mice and late passage C2C12 cells. Thus, we further investigated the regulatory effect of EGCG on miRNA-486-5p expression in age-regulated muscle loss. SAMP8 mice were fed with chow diet containing without or with 0.32% EGCG from aged 32 weeks for 8 weeks. Early passage (<12 passages) and late passage (>30 passages) of C2C12 cells were treated without or with EGCG at concentrations of 50 µM for 24h. Our data showed that EGCG supplementation increased miRNA-486-5p expression in both aged SAMP8 mice and late passage C2C12 cells. EGCG stimulated AKT phosphorylation and inhibited FoxO1a-mediated MuRF1 and Atrogin-1 transcription via up-regulating the expression of miR-486 in skeletal muscle of 40-wk-old SAMP8 mice as well as late passage C2C12 cells. In addition, myostatin expression was increased in late passage C2C12 cells and anti-myostatin treatment upregulated the expression of miR-486-5p. Our results identify a unique mechanism of a dietary constituent of green tea and suggest that use of EGCG or compounds derived from it attenuates age-associated muscle loss via myostatin/miRNAs/ubiquitin-proteasome signaling.


Subject(s)
Aging/metabolism , Catechin/analogs & derivatives , Gene Expression Regulation/drug effects , MicroRNAs/metabolism , Muscle Proteins/biosynthesis , Muscular Atrophy/metabolism , Myostatin/biosynthesis , Aging/drug effects , Aging/genetics , Aging/pathology , Animals , Catechin/chemistry , Catechin/pharmacology , Cell Line , Mice , Mice, Transgenic , MicroRNAs/genetics , Muscle Proteins/genetics , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Myostatin/genetics , Tea/chemistry
5.
Article in English | MEDLINE | ID: mdl-31676334

ABSTRACT

The molecular characteristics, expression patterns and functions of the amphibian myostatin (MSTN) gene are unknown. Here, we isolated a full-length Odorrana tormota MSTN cDNA sequence of 1701 bp (Ot-MSTN), containing a putative N-terminal signal peptide, a TGF-ß propeptide domain and an active peptide. Ot-MSTN was expressed in 9 selected tissues examined, and the highest level of expression was in thigh muscle, followed by brain and female gonadal tissue. The expression of Ot-MSTN in multiple O. tormota tissues supported that the activities of MSTN may be not limited to skeletal muscle. Ot-MSTN expression was decreased from stage 31 to stage 40, while the growth rate was increased. The expression of Ot-MSTN in adult male frogs increased with age, indicating that adult male frogs may inhibit the continued hypertrophy of thigh muscle fibers and decrease the growth rate of thigh muscle to ensure muscles do not grow too large. Luciferase reporter assays showed that miR-29b-3p directly targeted the 3'-UTR of Ot-MSTN. miR-29b-3p expression in the thigh muscle of 2 yrs. females who grew faster was significantly lower than that of the slow-growing 2 yrs. male individuals, which showed an opposite trend with Ot-MSTN expression. In addition,miR-29b-3p expression reversed trends of Ot-MSTN expression at different developmental stages in thigh muscle. Therefore, these data indicate that miR-29-3p may negatively regulate the expression of MSTN and regulate thigh muscle growth and development in O. tormota.


Subject(s)
Amphibian Proteins , Gene Expression Regulation/physiology , MicroRNAs , Myostatin , Amphibian Proteins/biosynthesis , Amphibian Proteins/genetics , Animals , Cloning, Molecular , Female , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Myostatin/biosynthesis , Myostatin/genetics , Ranidae
6.
Physiol Behav ; 212: 112697, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31622611

ABSTRACT

Myogenic regulators of muscle development, metabolism and growth differ between fish species in a context-specific manner. Commonly, the analysis of environmental influences on the expression of muscle-related gene regulators in teleosts is based on differences in swimming performance, feeding behaviour and stress-resistance, but the evaluation of behavioural phenotyping of immune and stress-related responsiveness in skeletal muscle is still scarce. Here we challenge proactive and reactive fingerlings of gilthead sea bream (Sparus aurata), one of the most commonly cultured species in the Mediterranean area, with highly pathogenic O1, O2α and O2ß serotypes of Vibrio anguillarum, a widespread opportunistic pathogen of marine animals, to analyse skeletal muscle responses to bath vaccination. Transcripts related to inflammation (interleukin 1ß, il1ß; tumour necrosis factor-α, tnfα; and immunoglobulin M, igm), and muscle metabolism and growth (lipoprotein, lpl; myostatin, mstn-1; myogenin; and growth hormone receptors type I and II, ghr1 and ghr2, respectively) were analysed. Biochemical indicators of muscle metabolism and function (creatine kinase, CK, aspartate aminotransferase, AST; esterase activity, EA; total antioxidant status, TAC and glucose) were also determined. Our results indicate that proactive, but not reactive, fish respond to Vibrio vaccination by increasing the expression levels of mstn-1, myogenin and ghr2 transcripts at short-/medium- term (1 to 3 days' post vaccination). No effect of vaccination was observed in immune indicators or biochemical parameters in either phenotypes, except for elevated levels of EA in reactive fish one-week post vaccination. This suggests that behavioural divergence should be taken into account to evaluate the crosstalk between immune, metabolic and growth processes in muscle of immune-challenged fish.


Subject(s)
Gene Expression Regulation/immunology , Myogenin/biosynthesis , Myostatin/biosynthesis , Receptors, Somatotropin/biosynthesis , Sea Bream/metabolism , Vaccination , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/metabolism , Biomarkers/metabolism , Creatine Kinase/metabolism , Esterases/metabolism , Glucose/metabolism , Muscle, Skeletal/metabolism , Phenotype , Vibrio Infections/prevention & control
7.
Biol Pharm Bull ; 42(9): 1437-1445, 2019.
Article in English | MEDLINE | ID: mdl-31474705

ABSTRACT

Chronic kidney disease (CKD), a chronic catabolic condition, is characterized by muscle wasting and decreased muscle endurance. Many insights into the molecular mechanisms of muscle wasting in CKD have been obtained. A persistent imbalance between protein degradation and synthesis in muscle causes muscle wasting. During muscle wasting, high levels of reactive oxygen species (ROS) and inflammatory cytokines are detected in muscle. These increased ROS and inflammatory cytokine levels induce the expression of myostatin. The myostatin binding to its receptor activin A receptor type IIB stimulates the expression of atrogenes such as atrogin-1 and muscle ring factor 1, members of the muscle-specific ubiquitin ligase family. Impaired mitochondrial function also contributes to reducing muscle endurance. The increased protein-bound uremic toxin, parathyroid hormone, glucocorticoid, and angiotensin II levels that are observed in CKD all have a negative effect on muscle mass and endurance. Among the protein-bound uremic toxins, indoxyl sulfate, an indole-containing compound has the potential to induce muscle atrophy by stimulating ROS-mediated myostatin and atrogenes expression. Indoxyl sulfate also impairs mitochondrial function. Some potential therapeutic approaches based on the muscle wasting mechanisms in CKD are currently in the testing stages.


Subject(s)
Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Renal Insufficiency, Chronic/complications , Sarcopenia/etiology , Cytokines/immunology , Humans , Indican/biosynthesis , Muscle, Skeletal/immunology , Myostatin/biosynthesis , Oxidative Stress/immunology , Proteolysis , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/metabolism , Sarcopenia/immunology , Sarcopenia/metabolism
8.
PLoS One ; 13(10): e0205664, 2018.
Article in English | MEDLINE | ID: mdl-30379863

ABSTRACT

Thoroughbred horses are finely-tuned athletes with a high aerobic capacity relative to skeletal muscle mass, attributable to centuries of genetic selection for speed and stamina. Polymorphisms in the myostatin gene (MSTN), a pronounced inhibitor of skeletal muscle growth, have been shown to almost singularly account for gene-based race distance aptitude in racehorses. In Thoroughbreds, two MSTN polymorphisms, a single nucleotide variation in the first intron (SNP g.66493737C>T) and a non-coding transposable element within the promoter region (a 227 bp SINE insertion) are of particular interest. Until now, it has not been clear which of these variants affect skeletal muscle phenotypes or whether both can impact racing performance. In a large cohort of Thoroughbreds, we observed a complete concordance between the SNP and the SINE insertion. By means of in vitro assays in C2C12 myoblasts, we isolated the SNP variant from the SINE polymorphism and showed the latter is exclusively responsible for adversely affecting transcription initiation and gene expression thereby limiting myostatin protein production. Mapping the MSTN transcription start site in horse skeletal muscle likewise revealed anomalous transcription initiation in the presence of the SINE insertion. Our data provides mechanistic evidence that the SINE insertion uniquely accounts for the MSTN "speed gene" effect on race distance aptitude in the Thoroughbred horse.


Subject(s)
Horses/genetics , Introns , Mutagenesis, Insertional , Myostatin/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Selective Breeding , Animals , Cell Line , Horses/metabolism , Muscle, Skeletal/metabolism , Myostatin/biosynthesis , Phenotype
9.
Biochem Biophys Res Commun ; 503(3): 1409-1414, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30025893

ABSTRACT

Muscle atrophy in metabolic conditions like chronic kidney disease (CKD) and diabetes are associated with glucocorticoid production, dysfunctional insulin/Akt/FoxO3 signaling and increased myostatin expression. We recently found that CREB, a transcription factor proposed to regulate myostatin expression, is highly phosphorylated in some wasting conditions. Based on a novel Akt-PDE3/4 signaling paradigm, we hypothesized that reduced Akt signaling contributes to CREB activation and myostatin expression. C2C12 myotubes were incubated with dexamethasone (Dex), an atrophy-inducing synthetic glucocorticoid. Akt/CREB signaling and myostatin expression were evaluated by immunoblot and qPCR analyses. Inhibitors of Akt, phosphodiesterase (PDE)-3/4, and protein kinase A (PKA) signaling were used to test our hypothesis. Incubating myotubes with Dex for 3-24 h inhibited Akt phosphorylation and enhanced CREB phosphorylation as well as myostatin mRNA and protein. Inhibition of PI3K/Akt signaling with LY294002 similarly increased CREB phosphorylation. Isobutyl-methylxanthine (IBMX, a pan PDE inhibitor), milrinone (PDE3 inhibitor) and rolipram (PDE4 inhibitor) augmented CREB phosphorylation and myostatin expression. Inhibition of protein kinase A by PKI reverted Dex- or IBMX-induced CREB phosphorylation and myostatin expression. Our study provides evidence supporting a newly identified mechanism by which a glucocorticoid-related reduction in Akt signaling contributes to myostatin expression via CREB activation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Glucocorticoids/pharmacology , Muscle Fibers, Skeletal/drug effects , Myostatin/metabolism , Signal Transduction/drug effects , Animals , Cells, Cultured , Mice , Muscle Fibers, Skeletal/metabolism , Myostatin/biosynthesis , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism
10.
Sci Rep ; 7(1): 11877, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28928419

ABSTRACT

Muscle wasting, also known as cachexia, is associated with many chronic diseases, which worsens prognosis of primary illness leading to enhanced mortality. Molecular basis of this metabolic syndrome is not yet completely understood. SIRT6 is a chromatin-bound member of the sirtuin family, implicated in regulating many cellular processes, ranging from metabolism, DNA repair to aging. SIRT6 knockout (SIRT6-KO) mice display loss of muscle, fat and bone density, typical characteristics of cachexia. Here we report that SIRT6 depletion in cardiac as well as skeletal muscle cells promotes myostatin (Mstn) expression. We also observed upregulation of other factors implicated in muscle atrophy, such as angiotensin-II, activin and Acvr2b, in SIRT6 depleted cells. SIRT6-KO mice showed degenerated skeletal muscle phenotype with significant fibrosis, an effect consistent with increased levels of Mstn. Additionally, we observed that in an in vivo model of cancer cachexia, Mstn expression coupled with downregulation of SIRT6. Furthermore, SIRT6 overexpression downregulated the cytokine (TNFα-IFNγ)-induced Mstn expression in C2C12 cells, and promoted myogenesis. From the ChIP assay, we found that SIRT6 controls Mstn expression by attenuating NF-κB binding to the Mstn promoter. Together, these data suggest a novel role for SIRT6 in maintaining muscle mass by controlling expression of atrophic factors like Mstn and activin.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myocardium/metabolism , Myostatin/biosynthesis , Sirtuins/metabolism , Up-Regulation , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Activins/genetics , Activins/metabolism , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Humans , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Myostatin/genetics , NF-kappa B/genetics , Rats , Response Elements , Sirtuins/genetics
11.
Int J Cardiol ; 238: 37-42, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28465115

ABSTRACT

BACKGROUND: Myostatin, a negative regulator of skeletal muscle mass, is up-regulated in the myocardium of heart failure (HF) and increased myostatin is associated with weight loss in animal models with HF. Although there are disparities in pathophysiology and epidemiology between male and female patients with HF, it remains unclear whether there is gender difference in myostatin expression and whether it is associated with weight loss in HF patients. METHODS: Heart tissue samples were collected from patients with advanced heart failure (n=31, female n=5) as well as healthy control donors (n=14, female n=6). Expression levels of myostatin and its related proteins in the heart were evaluated by western blotting analysis. RESULTS: Body mass index was significantly lower in female HF patients than in male counterparts (20.0±4.2 in female vs 25.2±3.8 in male, p=0.04). In female HF patients, both mature myostatin and pSmad2 were significantly up-regulated by 1.9 fold (p=0.05) and 2.5 fold (p<0.01) respectively compared to female donors, while expression of pSmad2 was increased by 2.8 times in male HF patients compared to male healthy subjects, but that of myostatin was not. There was no significant difference in protein expression related to myostatin signaling between male and female patients. CONCLUSION: In this study, myostatin and pSmad2 were significantly up-regulated in the failing heart of female patients, but not male patients, and female patients displayed lower body mass index. Enhanced myostatin signaling in female failing heart may causally contribute to pathogenesis of HF and cardiac cachexia.


Subject(s)
Heart Failure/metabolism , Myostatin/biosynthesis , Signal Transduction/physiology , Smad2 Protein/biosynthesis , Up-Regulation/physiology , Adult , Aged , Female , Heart Failure/pathology , Humans , Male , Middle Aged
12.
Medicine (Baltimore) ; 96(16): e6579, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28422851

ABSTRACT

Patients with coronary heart disease or acute myocardial infarction after cardiac catheterization with stenting referred for phase II cardiac rehabilitation (CR) were grouped according to their preference. Cardio-pulmonary exercise testing (CPET) was used to determine oxygen uptake ((Equation is included in full-text article.)) at peak exercise and anaerobic threshold (AT). The control patients received counseling only while the experiment group received 36 sessions of CR in 3 to 6 months. Exercise physiology parameters and serum myokines (myostatin, insulin-like growth factor-1 (IGF-1), and interleukin-6 (IL-6) were measured pre- and postrehabilitation.There were 29 patients in the experiment group and 10 in the control group, with no significant differences in baseline parameters. The experiment group had prominent progress in aerobic capacity and body composition after CR, but their serum myokine concentrations did not change significantly. Serum myostatin is positively correlated to peak (Equation is included in full-text article.)pre- and post-training, and pretraining AT (Equation is included in full-text article.), after adjusting for age, sex, and body composition. Serum IGF-1 is positively correlated with grip strength before training.Serum myostatin level is positively correlated to aerobic capacity, and IGF-1 level is positively correlated to grip strength in cardiac patients receiving CR.


Subject(s)
Cardiac Rehabilitation/methods , Exercise Test , Insulin-Like Growth Factor I/biosynthesis , Interleukin-6/biosynthesis , Myostatin/biosynthesis , Aged , Cardiac Catheterization , Coronary Disease/rehabilitation , Coronary Disease/surgery , Female , Humans , Male , Middle Aged , Muscle Strength/physiology , Myocardial Infarction/rehabilitation , Myocardial Infarction/surgery , Oxygen Consumption , Prospective Studies , Stents
13.
Int J Med Sci ; 13(9): 680-5, 2016.
Article in English | MEDLINE | ID: mdl-27647997

ABSTRACT

Effects of myostatin (MSTN)-suppression on the regeneration of injured skeletal muscle under unloading condition were investigated by using transgenic mice expressing a dominant-negative form of MSTN (MSTN-DN). Both MSTN-DN and wild-type (WT) mice were subjected to continuous hindlimb suspension (HS) for 6 weeks. Cardiotoxin (CTX) was injected into left soleus muscle under anesthesia 2 weeks after the initiation of HS. Then, the soleus muscles were excised following 6-week HS (4 weeks after CTX-injection). CTX-injection caused to reduce the soleus fiber cross-sectional area (CSA) in WT mice under both unloading and weight-bearing conditions, but not in MSTN-DN mice. Under unloading condition, CTX-injected muscle weight and fiber CSA in MSTN-DN mice were significantly higher than those in WT mice. CTX-injected muscle had many damaged and regenerating fibers having central nuclei in both WT and MSTN-DN mice. Significant increase in the population of Pax7-positive nuclei in CTX-injected muscle was observed in MSTN-DN mice, but not in WT mice. Evidences indicate that the suppression of MSTN cause to increase the regenerative potential of injured soleus muscle via the increase in the population of muscle satellite cells regardless of unloading conditions.


Subject(s)
Hindlimb/growth & development , Muscle, Skeletal/growth & development , Myostatin/biosynthesis , Regeneration , Animals , Cardiotoxins/administration & dosage , Hindlimb/drug effects , Hindlimb/injuries , Hindlimb/physiopathology , Humans , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , Myostatin/antagonists & inhibitors , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Weight-Bearing
14.
Sci Rep ; 6: 32084, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27549031

ABSTRACT

Skeletal muscle atrophy, referred to as sarcopenia, is often observed in chronic kidney disease (CKD) patients, especially in patients who are undergoing hemodialysis. The purpose of this study was to determine whether uremic toxins are involved in CKD-related skeletal muscle atrophy. Among six protein-bound uremic toxins, indole containing compounds, indoxyl sulfate (IS) significantly inhibited proliferation and myotube formation in C2C12 myoblast cells. IS increased the factors related to skeletal muscle breakdown, such as reactive oxygen species (ROS) and inflammatory cytokines (TNF-α, IL-6 and TGF-ß1) in C2C12 cells. IS also enhanced the production of muscle atrophy-related genes, myostatin and atrogin-1. These effects induced by IS were suppressed in the presence of an antioxidant or inhibitors of the organic anion transporter and aryl hydrocarbon receptor. The administered IS was distributed to skeletal muscle and induced superoxide production in half-nephrectomized (1/2 Nx) mice. The chronic administration of IS significantly reduced the body weights accompanied by skeletal muscle weight loss. Similar to the in vitro data, IS induced the expression of myostatin and atrogin-1 in addition to increasing the production of inflammatory cytokines by enhancing oxidative stress in skeletal muscle. These data suggest that IS has the potential to accelerate skeletal muscle atrophy by inducing oxidative stress-mediated myostatin and atrogin-1 expression.


Subject(s)
Gene Expression Regulation/drug effects , Indican/toxicity , Muscle Proteins/biosynthesis , Muscle, Skeletal/drug effects , Myostatin/biosynthesis , Oxidative Stress/drug effects , SKP Cullin F-Box Protein Ligases/biosynthesis , Sarcopenia/chemically induced , Animals , Antioxidants/pharmacology , Cell Division/drug effects , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Myoblasts/drug effects , Myostatin/genetics , Nephrectomy , Organ Size/drug effects , Organic Anion Transporters/antagonists & inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , Sarcopenia/genetics , Sarcopenia/metabolism , Superoxides/metabolism , Uremia/metabolism , Uremia/pathology , Weight Loss/drug effects
15.
Int J Cardiol ; 215: 384-7, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27128567

ABSTRACT

BACKGROUND: Myostatin has been shown to regulate skeletal and cardiac muscle growth. However, its status on long-term hypertrophied myocardium has not been addressed. The purpose of this study was to evaluate the expression of myocardial myostatin and its antagonist follistatin in spontaneously hypertensive rats (SHR) with heart failure. METHODS: Eighteen-month-old SHR were evaluated to identify clinical features of heart failure such as tachypnea/labored respiration and weight loss. After heart failure was detected, rats were subjected to echocardiogram and euthanized. Age-matched normotensive Wistar-Kyoto (WKY) rats were used as controls. Myostatin and follistatin protein expression was assessed by Western blotting. Statistical analysis was performed by Student's t test. RESULTS: All SHR (n=8) presented right ventricular hypertrophy and five had lung congestion. SHR had left chambers hypertrophy and dilation (left atrial diameter: WKY 5.73±0.59; SHR 7.28±1.17mm; p=0.004; left ventricular (LV) diastolic diameter/body weight ratio: WKY 19.6±3.1; SHR 27.7±4.7mm/kg; p=0.001), and LV systolic dysfunction (midwall fractional shortening: WKY 34.9±3.31; SHR 24.8±3.20%; p=0.003). Myocyte diameter (WKY 23.1±1.50, SHR 25.5±1.33µm; p=0.004) and myocardial interstitial collagen fraction (WKY 4.86±0.01; SHR 8.36±0.02%; p<0.001) were increased in the SHR. Myostatin (WKY 1.00±0.16; SHR 0.77±0.23 arbitrary units; p=0.035) and follistatin (WKY 1.00±0.35; SHR 0.49±0.18 arbitrary units; p=0.002) expression was lower in SHR. Myostatin and follistatin expression negatively correlated with LV diastolic diameter-to-body weight ratio and LV systolic diameter, and positively correlated with midwall fractional shortening. CONCLUSION: Myostatin and follistatin protein expression is reduced in the long-term hypertrophied myocardium from spontaneously hypertensive rats with heart failure.


Subject(s)
Heart Failure/metabolism , Hypertension/metabolism , Myocardium/metabolism , Myostatin/biosynthesis , Animals , Body Weight , Echocardiography , Follistatin/biosynthesis , Follistatin/metabolism , Heart Failure/diagnostic imaging , Male , Myocardium/pathology , Myostatin/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/metabolism
16.
Calcif Tissue Int ; 97(6): 602-10, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26340892

ABSTRACT

Vitamin D deficiency is associated with muscle weakness, pain, and atrophy. Serum vitamin D predicts muscle strength and age-related muscle changes. However, precise mechanisms by which vitamin D affects skeletal muscle are unclear. To address this question, this study characterizes the muscle phenotype and gene expression of mice with deletion of vitamin D receptor (VDRKO) or diet-induced vitamin D deficiency. VDRKO and vitamin D-deficient mice had significantly weaker grip strength than their controls. Weakness progressed with age and duration of vitamin D deficiency, respectively. Histological assessment showed that VDRKO mice had muscle fibers that were significantly smaller in size and displayed hyper-nuclearity. Real-time PCR also indicated muscle developmental changes in VDRKO mice with dysregulation of myogenic regulatory factors (MRFs) and increased myostatin in quadriceps muscle (>2-fold). Vitamin D-deficient mice also showed increases in myostatin and the atrophy marker E3-ubiqutin ligase MuRF1. As a potential explanation for grip strength weakness, both groups of mice had down-regulation of genes encoding calcium-handling and sarco-endoplasmic reticulum calcium transport ATPase (Serca) channels. This is the first report of reduced strength, morphological, and gene expression changes in VDRKO and vitamin D-deficient mice where confounding by calcium, magnesium, and phosphate have been excluded by direct testing. Although suggested in earlier in vitro work, this study is the first to report an in vivo association between vitamin D, myostatin, and the regulation of muscle mass. These findings support a direct role for vitamin D in muscle function and corroborate earlier work on the presence of VDR in this tissue.


Subject(s)
Hand Strength , Muscle Fibers, Skeletal/pathology , Myostatin/biosynthesis , Receptors, Calcitriol/deficiency , Vitamin D Deficiency/physiopathology , Animals , Disease Models, Animal , Hand Strength/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Real-Time Polymerase Chain Reaction , Vitamin D Deficiency/metabolism
17.
IUBMB Life ; 67(8): 589-600, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26305594

ABSTRACT

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


Subject(s)
Cell Differentiation/genetics , Muscle Development/genetics , Myostatin/genetics , Transforming Growth Factor beta/genetics , Gene Expression Regulation, Developmental , Humans , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myostatin/biosynthesis , Myostatin/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational
18.
Int J Mol Med ; 36(1): 29-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25955031

ABSTRACT

In the present study, we aimed to determine whether ethanol extracts of Fructus Schisandrae (FS), the dried fruit of Schizandra chinensis Baillon, mitigates the development of dexamethasone-induced muscle atrophy. Adult SPF/VAT outbred CrljOri:CD1 (ICR) mice were either treated with dexamethasone to induce muscle atrophy. Some mice were treated with various concentrations of FS or oxymetholone, a 17α-alkylated anabolic-androgenic steroid. Muscle thickness and weight, calf muscle strength, and serum creatine and creatine kinase (CK) levels were then measured. The administration of FS attenuated the decrease in calf thickness, gastrocnemius muscle thickness, muscle strength and weight, fiber diameter and serum lactate dehydrogenase levels in the gastrocnemius muscle bundles which was induced by dexamethasone in a dose-dependent manner. Treatment with FS also prevented the dexamethasone-induced increase in serum creatine and creatine kinase levels, histopathological muscle fiber microvacuolation and fibrosis, and the immunoreactivity of muscle fibers for nitrotyrosine, 4-hydroxynonenal, inducible nitric oxide synthase and myostatin. In addition, the destruction of the gastrocnemius antioxidant defense system was also inhibited by the administration of FS in a dose-dependent manner. FS downregulated the mRNA expression of atrogin-1 and muscle ring-finger protein-1 (involved in muscle protein degradation), myostatin (a potent negative regulator of muscle growth) and sirtuin 1 (a representative inhibitor of muscle regeneration), but upregulated the mRNA expression of phosphatidylinositol 3-kinase, Akt1, adenosine A1 receptor and transient receptor potential cation channel subfamily V member 4, involved in muscle growth and the activation of protein synthesis. The overall effects of treatment with 500 mg/kg FS were comparable to those observed following treatment with 50 mg/kg oxymetholone. The results from the present study support the hypothesis that FS has a favorable ameliorating effect on muscle atrophy induced by dexamethasone, by exerting anti-inflammatory and antioxidant effects on muscle fibers, which may be due to an increase in protein synthesis and a decrease in protein degradation.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Muscle Strength/drug effects , Muscle, Skeletal/pathology , Muscular Atrophy/drug therapy , Schisandra/metabolism , Aldehydes/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Creatine/blood , Creatine Kinase/blood , Dexamethasone/pharmacology , Fibrosis/drug therapy , Fibrosis/prevention & control , L-Lactate Dehydrogenase/blood , Mice , Mice, Inbred ICR , Muscle Proteins/genetics , Muscle Tonus/drug effects , Muscle, Skeletal/drug effects , Muscular Atrophy/prevention & control , Myostatin/biosynthesis , Myostatin/immunology , Nitric Oxide Synthase Type II/immunology , Oxymetholone/pharmacology , Phosphatidylinositol 3-Kinase/genetics , Protein Biosynthesis/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/biosynthesis , Receptor, Adenosine A1/genetics , SKP Cullin F-Box Protein Ligases/genetics , Sirtuin 1/genetics , TRPV Cation Channels/genetics , Tripartite Motif Proteins , Tyrosine/analogs & derivatives , Tyrosine/immunology , Ubiquitin-Protein Ligases/genetics
19.
Cell Signal ; 27(9): 1895-904, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25958325

ABSTRACT

MicroRNAs (miRNAs or miRs) play a critical role in skeletal muscle development. In a previous study we observed that miR-128 was highly expressed in skeletal muscle. However, its function in regulating skeletal muscle development is not clear. Our hypothesis was that miR-128 is involved in the regulation of the proliferation and differentiation of skeletal myoblasts. In this study, through bioinformatics analyses, we demonstrate that miR-128 specifically targeted mRNA of myostatin (MSTN), a critical inhibitor of skeletal myogenesis, at coding domain sequence (CDS) region, resulting in down-regulating of myostatin post-transcription. Overexpression of miR-128 inhibited proliferation of mouse C2C12 myoblast cells but promoted myotube formation; whereas knockdown of miR-128 had completely opposite effects. In addition, ectopic miR-128 regulated the expression of myogenic factor 5 (Myf5), myogenin (MyoG), paired box (Pax) 3 and 7. Furthermore, an inverse relationship was found between the expression of miR-128 and MSTN protein expression in vivo and in vitro. Taken together, these results reveal that there is a novel pathway in skeletal muscle development in which miR-128 regulates myostatin at CDS region to inhibit proliferation but promote differentiation of myoblast cells.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Down-Regulation/physiology , MicroRNAs/metabolism , Muscle Development/physiology , Myoblasts, Skeletal/metabolism , Myostatin/biosynthesis , Animals , Cell Line , Male , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myoblasts, Skeletal/cytology , Myostatin/genetics
20.
J Appl Physiol (1985) ; 118(12): 1460-6, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25749440

ABSTRACT

We had the unique opportunity to study the skeletal muscle characteristics, at the single fiber level, of a world champion sprint runner who is the current indoor world record holder in the 60-m hurdles (7.30 s) and former world record holder in 110-m hurdles (12.91 s). Muscle biopsies were obtained from the vastus lateralis at rest and 4 h after a high-intensity exercise challenge (4 × 7 repetitions of resistance exercise). Single muscle fiber analyses were conducted for fiber type distribution (myosin heavy chain, MHC), fiber size, contractile function (strength, speed, and power) and mRNA expression (before and after the exercise bout). The world-class sprinter's leg muscle had a high abundance (24%) of the pure MHC IIx muscle fibers with a total fast-twitch fiber population of 71%. Power output of the MHC IIx fibers (35.1 ± 1.4 W/l) was 2-fold higher than MHC IIa fibers (17.1 ± 0.5 W/l) and 14-fold greater than MHC I fibers (2.5 ± 0.1 W/l). Additionally, the MHC IIx fibers were highly responsive to intense exercise at the transcriptional level for genes involved with muscle growth and remodeling (Fn14 and myostatin). To our knowledge, the abundance of pure MHC IIx muscle fibers is the highest observed in an elite sprinter. Further, the power output of the MHC IIa and MHC IIx muscle fibers was greater than any human values reported to date. These data provide a myocellular basis for the high level of sprinting success achieved by this individual.


Subject(s)
Athletes , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/physiology , Running/physiology , Adult , Biopsy , Exercise Test , Gene Expression/genetics , Gene Expression/physiology , Humans , Leg , Male , Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Fast-Twitch/ultrastructure , Muscle Fibers, Skeletal/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myostatin/biosynthesis , Myostatin/genetics , Physical Education and Training , Receptors, Tumor Necrosis Factor/genetics , Resistance Training , Swimming/physiology , TWEAK Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...