Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Pharm Biol ; 60(1): 1106-1115, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35639355

ABSTRACT

CONTEXT: Obesity, one of the major public health problems worldwide, has attracted increasing attention. Ginsenoside Rb1 is the most abundant active component of Panax ginseng C.A.Mey (Araliaceae) and is reported to have beneficial effects on obesity and diabetes. However, the mechanisms by which Rb1 regulates obesity remain to be explored. OBJECTIVE: This paper intends to further explore the mechanism of Rb1 in regulating obesity. MATERIALS AND METHODS: The C57BL/6 obese mice were divided into two groups: the control (CTR) and Rb1. The CTR group [intraperitoneally (ip) administered with saline] and the Rb1 group (ip administered with Rb1, 40 mg/kg/d) were treated daily for four weeks. In vitro, Rb1 (0, 10, 20, 40 µM) was added to differentiated C2C12 cells and Rb1 (0, 20, 40 µM) was added to 3T3-L1 cells. After 24 h, total RNA and protein from C2C12 cells and 3T3-L1 cells were used to detect myostatin (MSTN) and fibronectin type III domain-containing 5 (FNDC5) expression. RESULTS: Rb1 reduced the body weight and adipocyte size. Improved glucose tolerance and increased basic metabolic activity were also found in Rb1 treated mice. MSTN was downregulated in differentiated C2C12 cells, 3T3-L1 cells and adipose tissues upon Rb1 treatment. FNDC5 was increased after Rb1 treatment. However, MSTN overexpression attenuated Rb1-mediated decrease accumulation of lipid droplets in differentiated 3T3-L1 adipocytes. DISCUSSION & CONCLUSIONS: Rb1 may ameliorate obesity in part through the MSTN/FNDC5 signalling pathway. Our results showed that Rb1 can be used as an effective drug in the treatment of human obesity.


Subject(s)
Ginsenosides , Myostatin , Obesity , Panax , Animals , Fibronectins , Ginsenosides/pharmacology , Mice , Mice, Inbred C57BL , Myostatin/genetics , Obesity/drug therapy , Obesity/metabolism
2.
Trop Anim Health Prod ; 54(2): 98, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35141787

ABSTRACT

The purpose of this study was to examine the effect of dietary supplementation with methyl methionine sulfonium chloride (MMSC), and L-carnitine (L-CAR) alone or in combination on the growth performance of broilers through their impact on the expression of IGF-1 and MSTN genes associated with growth in broilers. One-day-old female Ross 308 broiler chicks were allocated into four groups, each of which received a broiler starter diet and water daily ad libitum. The control group (group 1) was given drinking water without any additives. Group 2 received 0.25 g L-carnitine per liter of drinking water, group 3 received 0.25 g MMSC per liter of drinking water, and group 4 received 0.25 g of both L-carnitine and MMSC per liter of drinking water. Birds were given a starter diet to 21 days after which they received a broiler grower diet to 35 days when the experiment ended. There were five replicate groups of 12 birds per treatment. Body weights and feed intake were recorded weekly. Compared to the control group of birds, supplementation with MMSC either alone or in combination with L-carnitine resulted in an increase in growth rate or feed utilization efficiency; L-carnitine by itself had no effect. MMSC supplementation, again either alone or in combination with L-carnitine, increased jejunal and ileal villi height, increased serum total proteins and globulins, downregulated myostatin (MSTN) mRNA, and upregulated insulin growth factor-1 (IGF-1) mRNA expression. Supplementation with L-carnitine alone showed none of these effects. We conclude that MMSC supplementation improved growth performance through the upregulation of IGF-1 mRNA expression and downregulation of MSTN mRNA expression.


Subject(s)
Animal Nutritional Physiological Phenomena , Chickens , Insulin-Like Growth Factor I , Myostatin/genetics , Vitamin U , Animal Feed/analysis , Animals , Carnitine , Chickens/genetics , Chickens/growth & development , Chlorides , Diet/veterinary , Dietary Supplements , Female , Insulin , Insulin-Like Growth Factor I/genetics , Methionine/analogs & derivatives
3.
Int J Mol Sci ; 22(2)2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33467209

ABSTRACT

Skeletal muscle is the most abundant tissue and constitutes about 40% of total body mass. Herein, we report that crude water extract (CWE) of G. uralensis enhanced myoblast proliferation and differentiation. Pretreatment of mice with the CWE of G. uralensis prior to cardiotoxin-induced muscle injury was found to enhance muscle regeneration by inducing myogenic gene expression and downregulating myostatin expression. Furthermore, this extract reduced nitrotyrosine protein levels and atrophy-related gene expression. Of the five different fractions of the CWE of G. uralensis obtained, the ethyl acetate (EtOAc) fraction more significantly enhanced myoblast proliferation and differentiation than the other fractions. Ten bioactive compounds were isolated from the EtOAc fraction and characterized by GC-MS and NMR. Of these compounds (4-hydroxybenzoic acid, liquiritigenin, (R)-(-)-vestitol, isoliquiritigenin, medicarpin, tetrahydroxymethoxychalcone, licochalcone B, liquiritin, liquiritinapioside, and ononin), liquiritigenin, tetrahydroxymethoxychalcone, and licochalcone B were found to enhance myoblast proliferation and differentiation, and myofiber diameters in injured muscles were wider with the liquiritigenin than the non-treated one. Computational analysis showed these compounds are non-toxic and possess good drug-likeness properties. These findings suggest that G. uralensis-extracted components might be useful therapeutic agents for the management of muscle-associated diseases.


Subject(s)
Glycyrrhiza uralensis/chemistry , Muscular Atrophy/drug therapy , Plant Extracts/chemistry , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Chalcones/chemistry , Chalcones/pharmacology , Chalcones/therapeutic use , Flavanones/chemistry , Flavanones/pharmacology , Flavanones/therapeutic use , Male , Mice , Mice, Inbred C57BL , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Myostatin/genetics , Myostatin/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Tyrosine/analogs & derivatives , Tyrosine/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.
Nutrients ; 12(5)2020 Apr 28.
Article in English | MEDLINE | ID: mdl-32354126

ABSTRACT

Muscle wasting is caused by various factors, such as aging, cancer, diabetes, and chronic kidney disease, and significantly decreases the quality of life. However, therapeutic interventions for muscle atrophy have not yet been well-developed. In this study, we investigated the effects of schisandrin A (SNA), a component extracted from the fruits of Schisandra chinensis, on dexamethasone (DEX)-induced muscle atrophy in mice and studied the underlying mechanisms. DEX+SNA-treated mice had significantly increased grip strength, muscle weight, and muscle fiber size compared with DEX+vehicle-treated mice. In addition, SNA treatment significantly reduced the expression of muscle degradation factors such as myostatin, MAFbx (atrogin1), and muscle RING-finger protein-1 (MuRF1) and enhanced the expression of myosin heavy chain (MyHC) compared to the vehicle. In vitro studies using differentiated C2C12 myotubes also showed that SNA treatment decreased the expression of muscle degradation factors induced by dexamethasone and increased protein synthesis and expression of MyHCs by regulation of Akt/FoxO and Akt/70S6K pathways, respectively. These results suggest that SNA reduces protein degradation and increases protein synthesis in the muscle, contributing to the amelioration of dexamethasone-induced muscle atrophy and may be a potential candidate for the prevention and treatment of muscle atrophy.


Subject(s)
Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Dexamethasone/adverse effects , Gene Expression/drug effects , Lignans/pharmacology , Lignans/therapeutic use , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/prevention & control , Phytotherapy , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Schisandra/chemistry , Animals , Cells, Cultured , Cyclooctanes/administration & dosage , Cyclooctanes/isolation & purification , Lignans/administration & dosage , Lignans/isolation & purification , Male , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscular Atrophy/chemically induced , Muscular Atrophy/physiopathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myostatin/genetics , Myostatin/metabolism , Organ Size/drug effects , Polycyclic Compounds/administration & dosage , Polycyclic Compounds/isolation & purification , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
Fish Shellfish Immunol ; 98: 710-719, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31707005

ABSTRACT

Myostatin, through type I receptor (kinase 4, 5, ALK4/5), functions to participate in the immune system and negatively regulate muscle growth in mammals. However, the role of myostatin (mstn) in the immune system of teleosts is largely unknown. In a previous study, we cloned the mstn1 cDNA encoding myostatin in Qi river crucian carp (Carassius auratus). In the present study, we have cloned mstn2 cDNA, which was characterized and analyzed together with mstn1. Tissue distribution analysis showed that both mstn genes are expressed in numerous tissues, with mstn1 dominantly expressed in the muscle and brain, whereas mstn2 is mainly expressed in the brain. During embryogenesis, mstn1 and mstn2 exhibit different expression patterns. Both mstn1 and mstn2 expression increased stepwise in the brain at different developmental stages. Furthermore, both genes are differentially regulated during different periods of fasting/re-feeding. Following the exposure of C. auratus to polyI:C, lipopolysaccharide (LPS), and Aeromonas hydrophila, both genes were upregulated in different tissues, which indicated that they might be involved in the immune response against pathogenic invasion. Blocking the Mstn signal pathway with SB-431542 (a chemical inhibitor of ALK4/5) resulted in significantly increased body length and weight. However, the mortality of SB-431542-treated fish was higher after A. hydrophila challenge. Moreover, decreased expression of lysozymes (lyz), complement component 3 (c3), ß-defensin 3 (defb3), and interferon γ (ifnγ) were exhibited in treated fish, compared with the controls. Furthermore, the expression of nf-κb1, three pro-inflammatory cytokines (il1ß, il6, and tnfα), and inflammatory cytokines (il8 and il10) were significantly increased in both the SB-431542-treated group and the control after A. hydrophila infection, suggesting that the NF-κB pathway was not suppressed in the SB-431542-treated fish. Taken together, our data suggest that both mstn1 and mstn2 play important roles in early body development, muscle growth, and the immune system by acting downstream of the NF-κB signal pathway.


Subject(s)
Carps/genetics , Carps/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Myostatin/genetics , Myostatin/immunology , Aeromonas hydrophila/physiology , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Goldfish/genetics , Goldfish/immunology , Gram-Negative Bacterial Infections/immunology , Lipopolysaccharides/pharmacology , Poly I-C/pharmacology
7.
Gene ; 682: 1-12, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30267811

ABSTRACT

The myostatin (mstn) and myostatinb (mstnb) gene of Cranoglanis bouderius were cloned and sequenced and their expressions under nutritional restriction were characterized. The full cDNA sequences of mstn and mstnb were 1878 bp and 1928 bp, containing an open reading frame of 1170 bp and 1119 bp, which encoded 390 and 373 amino acids, respectively. The deduced mstn and mstnb sequence structures were similar to other members of TGF-ß superfamily, including the TGF beta pro-peptide, TGF beta domain, proteolytic processing site and nine conserved cysteines in the C-terminal. In addition, four mstn gene duplications were found in Cranoglanis bouderius. Sequence alignment and phylogenetic tree analyses indicated that the mstn gene and mstnb gene had a close relationship with Siluriformes fish, and the mstn and mstnb genes were roughly classified into two groups. RT-PCR analysis revealed that the mstn and mstnb were expressed in a variety of tissues in Cranoglanis bouderius although the mstn was highly expressed in skeletal muscle and the mstnb was mainly expressed in brain. We speculate that the mstn gene but not mstnb is likely to play a key role in managing muscle growth. A fasting-re-feeding experiment was used to evaluate the effects of starvation on mstn and mstnb expressions in juvenile Cranoglanis bouderius for 5 weeks. The result showed that the mstn and mstnb transcript levels varied among tissues. The mRNA expression levels of mstn in muscle, brain and liver gradually decreased during starvation and returned to the normal level after re-feeding. The mstnb mRNA levels in muscle, brain, liver, spleen, intestine and kidney increased during an early fast time but ultimately decreased with prolonged fasting time. The mstnb transcript levels in muscle, brain and liver increased significantly after re-feeding. In summary, the results supported that the mstn and mstnb may not be limited to control of muscle growth in fish but could also be involved in other biological functions.


Subject(s)
Catfishes/genetics , Eating , Fasting , Myostatin/genetics , Animals , Brain/metabolism , Cloning, Molecular , DNA, Complementary/metabolism , Fish Proteins/genetics , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Open Reading Frames , Phylogeny , RNA, Messenger/metabolism , Species Specificity
8.
Fish Shellfish Immunol ; 80: 416-425, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29920384

ABSTRACT

The present study was conducted to evaluate the supplementation of three autochthonous Bacillus strains (B. subtilis, B. amyloliquefaciens and B. cereus) and a commercial B. amyloliquefaciensin doses of 1 × 1010 CFU/kg on the growth performance, hematology, antioxidant activities, digestive enzyme levels, immune status and disease resistance of Clarias gariepinus. A total of 300 fish (75.23 ±â€¯1.6 g) were randomly divided into 5 groups (each group was subdivided into 2 subgroups, 30 fish/each). The control group was fed basal diet (D0). Diets D1, D2, D3 and D4were supplemented with B. subtilis, B. amyloliquefaciens, B. cereus and a commercial B. amyloliquefaciens, respectively. During the course of the experiment, D3 showed the best body weight, weight gain, specific growth rate and food conversion ratio. The measured hemogram blood parameters had the highest significant increase in D3. WBCs and monocyte counts had no significant differences among the experimental groups. The serum antioxidant and digestive enzymes were the highest in D3 and were the lowest in D0. After 15 d, the non-specific immune parameters were markedly increased in fish fed probiotic-containing diet compared with the control. After 30 d, the highest significant immune parameters were observed in D3; D1 and D2 had no significant differences in serum lysozyme activity, nitric oxide and IgM compared with D0. Myostatin cDNA levels were adversely affected by probiotic supplements compare with the control. The PACAP expression showed the highest significant value in D3 followed by D1and D4then D2. The relative survival percentages of the Aeromonas sobria challenged C. gariepinus were the highest in D3, D2, D4 and then D1. Among the three isolated Bacillus species, dietary supplementation with the B. cereus had the highest performance in C. gariepinus compared with the commercial B. amyloliquefaciens and the control group.


Subject(s)
Bacillus , Catfishes , Probiotics , Aeromonas , Amylases/blood , Animal Feed , Animals , Body Weight , Catfishes/genetics , Catfishes/growth & development , Catfishes/immunology , Catfishes/microbiology , Disease Resistance , Erythrocyte Count , Fish Diseases , Gastrointestinal Microbiome , Gram-Negative Bacterial Infections/veterinary , Lipase/blood , Myostatin/genetics , Peptide Hydrolases/blood , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , RNA, Messenger/metabolism , Superoxide Dismutase/metabolism
9.
Curr Protein Pept Sci ; 18(12): 1273-1287, 2017.
Article in English | MEDLINE | ID: mdl-28595527

ABSTRACT

Creatine, a very popular supplement among athletic populations, is of growing interest for clinical applications. Since over 90% of creatine is stored in skeletal muscle, the effect of creatine supplementation on muscle metabolism is a widely studied area. While numerous studies over the past few decades have shown that creatine supplementation has many favorable effects on skeletal muscle physiology and metabolism, including enhancing muscle mass (growth/hypertrophy); the underlying mechanisms are poorly understood. This report reviews studies addressing the mechanisms of action of creatine supplementation on skeletal muscle growth/hypertrophy. Early research proposed that the osmotic effect of creatine supplementation serves as a cellular stressor (osmosensing) that acts as an anabolic stimulus for protein synthesis signal pathways. Other reports indicated that creatine directly affects muscle protein synthesis via modulations of components in the mammalian target of rapamycin (mTOR) pathway. Creatine may also directly affect the myogenic process (formation of muscle tissue), by altering secretions of myokines, such as myostatin and insulin-like growth factor-1, and expressions of myogenic regulatory factors, resulting in enhanced satellite cells mitotic activities and differentiation into myofiber. Overall, there is still no clear understanding of the mechanisms of action regarding how creatine affects muscle mass/growth, but current evidence suggests it may exert its effects through multiple approaches, with converging impacts on protein synthesis and myogenesis.


Subject(s)
Creatine/administration & dosage , Dietary Supplements , Muscle Development/drug effects , Myogenic Regulatory Factors/genetics , Protein Biosynthesis/drug effects , Animals , Cell Differentiation , Gene Expression Regulation , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/agonists , Myogenic Regulatory Factors/metabolism , Myostatin/genetics , Myostatin/metabolism , Protein Biosynthesis/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
10.
Genet Mol Res ; 16(1)2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28362981

ABSTRACT

Propolis can be used as growth enhancer due to its antimicrobial, antioxidant, and immune-stimulant properties, but its effects on morphometry and muscle gene expression are largely unknown. The present study evaluates the influence of propolis on muscle morphometry and myostatin gene expression in Nile tilapia (Oreochromis niloticus) bred in net cages. Reversed males (GIFT strain) with an initial weight of 170 ± 25 g were distributed in a (2 x 4) factorial scheme, with two diets (DPRO, commercial diet with 4% propolis ethanol extract and DCON, commercial diet without propolis, control) and four assessment periods (0, 35, 70, and 105 experimental days). Muscles were evaluated at each assessment period. Histomorphometric analysis classified the fiber diameters into four groups: <20 µm; 20-30 µm; 30-50 µm; and > 50 µm. RT-qPCR was performed to assess myostatin gene expression. Fibers < 20 µm diameter were more frequent in DPRO than in DCON at all times. Fiber percentages >30 µm (30-50 and > 50 µm) at 70 days were 25.39% and 40.07% for DPRO and DCON, respectively. There was greater myostatin gene expression at 105 days, averaging 1.93 and 1.89 for DCON and DPRO, respectively, with no significant difference in any of the analyzed periods. Propolis ethanol extract did not affect the diameter of muscle fibers or the gene expression of myostatin. Future studies should describe the mechanisms of natural products' effects on muscle growth and development since these factors are highly relevant for fish production performance.


Subject(s)
Cichlids/anatomy & histology , Ethanol/administration & dosage , Muscle, Skeletal/drug effects , Myostatin/genetics , Propolis/chemistry , Animals , Aquaculture/methods , Cichlids/genetics , Ethanol/chemistry , Ethanol/pharmacology , Fish Proteins/genetics , Gene Expression Regulation/drug effects , Male , Muscle, Skeletal/ultrastructure , Organ Size/drug effects , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/pharmacology
11.
Lasers Med Sci ; 32(9): 2167-2171, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27942989

ABSTRACT

Acupuncture treatment utilizes the stimulation of metal acupuncture needles that are manually inserted into a living body. In the last decades, laser light has been used as an alternative to needles to stimulate acupuncture points. We previously reported suppression of myostatin (Mstn) gene expression in skeletal muscle by means of femtosecond laser (FL) irradiation, after electroacupuncture, in which acupuncture needles are stimulated with a low-frequency microcurrent. The purpose of the study here was to investigate the efficacy of FL irradiation in mouse skeletal muscle with regard to protein synthesis. After irradiation of the hindlimbs, we first analyzed Mstn gene expression and Mstn protein level in the skeletal muscle. We then evaluated phosphorylation of the mammalian target of rapamycin (mTOR) and its downstream target 70-kDa ribosomal protein S6 kinase (p70S6K). The results showed that FL irradiation significantly reduced the amount of Mstn protein and enhanced the phosphorylation of p70S6K in of the mTOR/S6K signaling pathway. We suggest that FL irradiation activated the protein synthetic pathway in the skeletal muscle. In conclusion, we determined that FL irradiation can serve as an alternative for acupuncture needles and has the potential of being a new non-invasive acupuncture treatment of skeletal muscle.


Subject(s)
Acupuncture Therapy/methods , Lasers , Animals , Creatine Kinase/blood , Gene Expression Regulation , Male , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myostatin/genetics , Myostatin/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
12.
PLoS One ; 11(11): e0165843, 2016.
Article in English | MEDLINE | ID: mdl-27812153

ABSTRACT

Our laboratory recently produced genetically engineered (GE) Meishan pigs containing a ZFN-edited myostatin loss-of-function mutant. These GE pigs develop and grow as normal as wild type pigs but produce pork with greater lean yield and lower fat mass. To assess any potential subchronic toxicity risks of this GE pork, a 90-day feeding study was conducted in Sprague-Dawley rats. Rats were randomly divided into five groups, and fed for 90 days with basic diet and basic diets formulated with low dose and high dose pork prepared from wild type pigs and GE pigs, respectively. Animal behaviors and clinical signs were monitored twice daily, and body weight and food consumption were measured and recorded weekly. At days 45 and 90, blood tests (lipid panel, electrolytes, parameters related to liver and kidney functions, and complete blood counts) were performed. Additionally, gross pathology and histopathological analyses were performed for major organs in each group. Data analysis shows that there were no significant differences in growth rate, food consumption, and blood test parameters between rat groups fed with GE pork and wild type pork. Although differences in some liver function parameters (such as aspartate aminotransferase, total proteins, albumin, and alkaline phosphatase) and white blood cell counts (such as lymphocyte percentage and monocyte percentage) were observed between rats fed with high dose GE pork and basic diet, all test results in rats fed with GE pork are in the normal range. Additionally, there are no apparent lesions noted in all organs isolated from rats in all five feeding groups on days 45 and 90. Overall, our results clearly indicate that food consumption of GE pork produced by ZFN-edited myostatin loss-of-function mutant pigs did not have any long-term adverse effects on the health status in rats.


Subject(s)
Animal Feed , Food Safety , Food, Genetically Modified/adverse effects , Red Meat/adverse effects , Swine , Animal Feed/adverse effects , Animals , Body Weight/drug effects , Eating/drug effects , Female , Hematology , Lipids/blood , Liver Function Tests , Male , Mutation , Myostatin/genetics , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Time Factors
13.
J Nutr ; 144(8): 1196-203, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24919692

ABSTRACT

Short successive periods of muscle disuse, due to injury or illness, can contribute significantly to the loss of muscle mass with aging (sarcopenia). It has been suggested that increasing the protein content of the diet may be an effective dietary strategy to attenuate muscle disuse atrophy. We hypothesized that protein supplementation twice daily would preserve muscle mass during a short period of limb immobilization. Twenty-three healthy older (69 ± 1 y) men were subjected to 5 d of one-legged knee immobilization by means of a full-leg cast with (PRO group; n = 11) or without (CON group; n = 12) administration of a dietary protein supplement (20.7 g of protein, 9.3 g of carbohydrate, and 3.0 g of fat) twice daily. Two d prior to and immediately after the immobilization period, single-slice computed tomography scans of the quadriceps and single-leg 1 repetition maximum strength tests were performed to assess muscle cross-sectional area (CSA) and leg muscle strength, respectively. Additionally, muscle biopsies were collected to assess muscle fiber characteristics as well as mRNA and protein expression of selected genes. Immobilization decreased quadriceps' CSAs by 1.5 ± 0.7% (P < 0.05) and 2.0 ± 0.6% (P < 0.05), and muscle strength by 8.3 ± 3.3% (P < 0.05) and 9.3 ± 1.6% (P < 0.05) in the CON and PRO groups, respectively, without differences between groups. Skeletal muscle myostatin, myogenin, and muscle RING-finger protein-1 (MuRF1) mRNA expression increased following immobilization in both groups (P < 0.05), whereas muscle atrophy F-box/atrogen-1 (MAFBx) mRNA expression increased in the PRO group only (P < 0.05). In conclusion, dietary protein supplementation (∼20 g twice daily) does not attenuate muscle loss during short-term muscle disuse in healthy older men. This trial was registered at clinicaltrials.gov as NCT01588808.


Subject(s)
Dietary Proteins/administration & dosage , Dietary Supplements , Muscular Disorders, Atrophic/drug therapy , Quadriceps Muscle/drug effects , Sarcopenia/prevention & control , Aged , Diet , Energy Intake , Humans , Immobilization , Male , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Strength/drug effects , Myogenin/genetics , Myogenin/metabolism , Myostatin/genetics , Myostatin/metabolism , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Surveys and Questionnaires , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Zebrafish ; 11(3): 265-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24813227

ABSTRACT

Myostatin (Mstn), a member of the transforming growth factor ß superfamily, plays an inhibiting role in mammalian muscle growth. Mammals like human, cattle, mouse, sheep, and dog carrying null alleles of Mstn display a double-muscle phenotype. Mstn is conserved in fish; however, little is known whether the fish with mutated mstn display a similar phenotype to mammals because of the lack of mutant fish with mstn null alleles. Previously, we knocked out one of the duplicated copies of myostatin gene (mstna) in yellow catfish using zinc-finger nucleases. In this study, we report the identification of the second myostatin gene (mstnb) and knockout of mstnb in yellow catfish. The gene comprises three exons. It is predicted to encode 373 amino acid residues. The predicted protein exhibits 59.3% identity with yellow catfish Mstna and 57.3% identity with human MSTN. Employing TALEN (transcription activator-like effector nucleases) technology, we obtained two founders (from four randomly selected founders) of yellow catfish carrying the mutated mstnb gene in their germ cells. Totally, six mutated alleles of mstnb were obtained from the founders. Among the six alleles, four are nonframeshift and two are frameshift mutation. The frameshift mutated alleles include mstnb(nju22), an 8 bp deletion, and mstnb(nju24), a complex type of mutation comprising a 7 bp deletion and a 12 bp insertion. They are predicted to encode function null Mstnb. Our results will help to understand the roles of mstn genes in fish growth.


Subject(s)
Catfishes/genetics , Endonucleases/metabolism , Fish Proteins/genetics , Myostatin/genetics , Amino Acid Sequence , Animals , Base Sequence , Catfishes/embryology , Catfishes/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Embryo, Nonmammalian/metabolism , Female , Fish Proteins/chemistry , Fish Proteins/metabolism , Gene Knockout Techniques , Gene Targeting , Inheritance Patterns , Male , Molecular Sequence Data , Myostatin/chemistry , Myostatin/metabolism , Phylogeny
15.
Am J Physiol Endocrinol Metab ; 305(1): E33-40, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23632633

ABSTRACT

Although it is well established that the lack of myostatin (Mstn) promotes skeletal muscle hypertrophy, the corresponding changes regarding force generation have been studied mainly in vitro and remain conflicting. Furthermore, the metabolic underpinnings of these changes are very poorly documented. To clarify this issue, we have investigated strictly noninvasively in vivo the impact of the lack of Mstn on gastrocnemius muscle function and energetics in Mstn-targeted knockout (Mstn-/-) mice using ¹H-magnetic resonance (MR) imaging and ³¹P-MR spectroscopy during maximal repeated isometric contractions induced by transcutaneous electrostimulation. In Mstn-/- animals, although body weight, gastrocnemius muscle volume, and absolute force were larger (+38, +118, and +34%, respectively) compared with wild-type (Mstn+/+) mice, specific force (calculated from MR imaging measurements) was significantly lower (-36%), and resistance to fatigue was decreased. Besides, Mstn deficiency did not affect phosphorylated compound concentrations and intracellular pH at rest but caused a large increase in ATP cost of contraction (up to +206% compared with Mstn+/+) throughout the stimulation period. Further, Mstn deficiency limits the shift toward oxidative metabolism during muscle activity despite the fact that oxidative ATP synthesis capacity was not altered. Our data demonstrate in vivo that the absence of Mstn impairs both mechanical performance and energy cost of contraction in hypertrophic muscle. These findings must be kept in mind when considering Mstn as a potential therapeutic target for increasing muscle mass in patients suffering from muscle-wasting disorders.


Subject(s)
Energy Metabolism/physiology , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Myostatin/genetics , Myostatin/metabolism , Physical Conditioning, Animal/physiology , Adenosine Triphosphate/metabolism , Animals , Biomechanical Phenomena/genetics , Electric Stimulation , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology
16.
Eur J Nutr ; 52(2): 477-87, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22476926

ABSTRACT

PURPOSE: The aim of the study was to examine the acute effects of pre-exercise ingestion of protein, carbohydrate, and a non-caloric placebo on serum concentrations of insulin and cortisol, and the intramuscular gene expression of myostatin- and ubiquitin proteasome pathway (UPP)-related genes following a bout of resistance exercise. METHODS: Ten untrained college-aged men participated in three resistance exercise sessions (3 × 10 at 80 % 1RM for bilateral hack squat, leg press, and leg extension) in a cross-over fashion, which were randomly preceded by protein, carbohydrate, or placebo ingestion 30 min prior to training. Pre-supplement/pre-exercise, 2 h and 6 h post-exercise muscle biopsies were obtained during each session and analyzed for mRNA fold changes in myostatin (MSTN), activin IIB, follistatin-like 3 (FSTL3), SMAD specific E3 ubiquitin protein ligase 1 (SMURF1), forkhead box O3, F-box protein 32 (FBXO32), and Muscle RING-finger protein-1, with beta-actin serving as the housekeeping gene. Gene expression of all genes was analyzed using real-time PCR. RESULTS: Acute feeding appeared to have no significant effect on myostatin or UPP biomarkers. However, resistance exercise resulted in a significant downregulation of MSTN and FBXO32 mRNA expression and a significant upregulation in FSTL3 and SMURF1 mRNA expression (p < 0.05). CONCLUSIONS: An acute bout of resistance exercise results in acute post-exercise alterations in intramuscular mRNA expression of myostatin and UPP markers suggestive of skeletal muscle growth. However, carbohydrate and protein feeding surrounding resistance exercise appear to have little influence on the acute expression of these markers.


Subject(s)
Biomarkers/blood , Eating , Hydrocortisone/blood , Insulin/blood , Myostatin/genetics , Resistance Training , Ubiquitin-Protein Ligases/metabolism , Actins/genetics , Actins/metabolism , Cross-Over Studies , Dietary Carbohydrates/administration & dosage , Dietary Proteins/administration & dosage , Down-Regulation , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Male , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Myostatin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Single-Blind Method , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Up-Regulation , Young Adult
17.
Oxid Med Cell Longev ; 2013: 782497, 2013.
Article in English | MEDLINE | ID: mdl-24454989

ABSTRACT

OBJECTIVE: The antidiabetes drug astragalus polysaccharide (APS) is capable of increasing insulin sensitivity in skeletal muscle and improving whole-body glucose homeostasis. Recent studies suggest that skeletal muscle secreted growth factor myostatin plays an important role in regulating insulin signaling and insulin resistance. We hypothesized that regulation of skeletal muscle myostatin expression may be involved in the improvement of insulin sensitivity by APS. METHODS: APS was administered to 13-week-old diabetic KKAy and nondiabetic C57BL/6J mice for 8 weeks. Complementary studies examined APS effects on the saturated acid palmitate-induced insulin resistance and myostatin expression in C2C12 cells. RESULTS: APS treatment ameliorated hyperglycemia, hyperlipidemia, and insulin resistance and decreased the elevation of myostatin expression and malondialdehyde production in skeletal muscle of noninsulin-dependent diabetic KKAy mice. In C2C12 cells in vitro, saturated acid palmitate-induced impaired glucose uptake, overproduction of ROS, activation of extracellular regulated protein kinases (ERK), and NF-κB were partially restored by APS treatment. The protective effects of APS were mimicked by ERK and NF-κB inhibitors, respectively. CONCLUSION: Our study demonstrates elevated myostatin expression in skeletal muscle of type 2 diabetic KKAy mice and in cultured C2C12 cells exposed to palmitate. APS is capable of improving insulin sensitivity and decreasing myostatin expression in skeletal muscle through downregulating ROS-ERK-NF-κB pathway.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Muscle, Skeletal/metabolism , Myostatin/metabolism , NF-kappa B/metabolism , Polysaccharides/pharmacology , Reactive Oxygen Species/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cell Line , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Fatty Acids/blood , Gene Expression Regulation/drug effects , Insulin Resistance , MAP Kinase Signaling System/drug effects , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Myostatin/genetics , Palmitic Acid/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Am J Physiol Endocrinol Metab ; 303(5): E614-23, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22739107

ABSTRACT

Physical activity is required to attenuate the loss of skeletal muscle mass with aging. Short periods of muscle disuse, due to sickness or hospitalization, reduce muscle protein synthesis rates, resulting in rapid muscle loss. The present study investigates the capacity of neuromuscular electrical stimulation (NMES) to increase in vivo skeletal muscle protein synthesis rates in older type 2 diabetes patients. Six elderly type 2 diabetic men (70 ± 2 yr) were subjected to 60 min of one-legged NMES. Continuous infusions with L-[ring-¹³C6]phenylalanine were applied, with blood and muscle samples being collected regularly to assess muscle protein synthesis rates in both the stimulated (STIM) and nonstimulated control (CON) leg during 4 h of recovery after NMES. Furthermore, mRNA expression of key genes implicated in the regulation of muscle mass were measured over time in the STIM and CON leg. Muscle protein synthesis rates were greater in the STIM compared with the CON leg during recovery from NMES (0.057 ± 0.008 vs. 0.045 ± 0.008%/h, respectively, P < 0.01). Skeletal muscle myostatin mRNA expression in the STIM leg tended to increase immediately following NMES compared with the CON leg (1.63- vs. 1.00-fold, respectively, P = 0.07) but strongly declined after 2 and 4 h of recovery in the STIM leg only. In conclusion, this is the first study to show that NMES directly stimulates skeletal muscle protein synthesis rates in vivo in humans. NMES likely represents an effective interventional strategy to attenuate muscle loss in elderly individuals during bed rest and/or in other disuse states.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Electric Stimulation Therapy , Muscle Proteins/biosynthesis , Neuromuscular Junction/physiopathology , Quadriceps Muscle/metabolism , Sarcopenia/prevention & control , Aged , Atrophy/etiology , Atrophy/metabolism , Atrophy/pathology , Atrophy/prevention & control , Bed Rest/adverse effects , Biopsy, Needle , Carbon Isotopes , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Electric Stimulation Therapy/methods , Gene Expression Regulation , Humans , Immobilization/adverse effects , Kinetics , Male , Muscle Proteins/genetics , Myostatin/biosynthesis , Myostatin/genetics , Phenylalanine/blood , Phenylalanine/metabolism , Quadriceps Muscle/pathology , Quadriceps Muscle/physiopathology , RNA, Messenger/metabolism , Sarcopenia/complications , Sarcopenia/etiology
19.
Am J Physiol Endocrinol Metab ; 300(5): E790-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21304067

ABSTRACT

Arthritis is a chronic inflammatory illness that induces cachexia, which has a direct impact on morbidity and mortality. Fenofibrate, a selective PPARα activator prescribed to treat human dyslipidemia, has been reported to decrease inflammation in rheumatoid arthritis patients. The aim of this study was to elucidate whether fenofibrate is able to ameliorate skeletal muscle wasting in adjuvant-induced arthritis, an experimental model of rheumatoid arthritis. On day 4 after adjuvant injection, control and arthritic rats were treated with 300 mg/kg fenofibrate until day 15, when all rats were euthanized. Fenofibrate decreased external signs of arthritis and liver TNFα and blocked arthritis-induced decreased in PPARα expression in the gastrocnemius muscle. Arthritis decreased gastrocnemius weight, which results from a decrease in cross-section area and myofiber size, whereas fenofibrate administration to arthritic rats attenuated the decrease in both gastrocnemius weight and fast myofiber size. Fenofibrate treatment prevented arthritis-induced increase in atrogin-1 and MuRF1 expression in the gastrocnemius. Neither arthritis nor fenofibrate administration modify Akt-FoxO3 signaling. Myostatin expression was not modified by arthritis, but fenofibrate decreased myostatin expression in the gastrocnemius of arthritic rats. Arthritis increased muscle expression of MyoD, PCNA, and myogenin in the rats treated with vehicle but not in those treated with fenofibrate. The results indicate that, in experimental arthritis, fenofibrate decreases skeletal muscle atrophy through inhibition of the ubiquitin-proteasome system and myostatin.


Subject(s)
Arthritis, Experimental/pathology , Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , Muscle Proteins/biosynthesis , Muscle, Skeletal/pathology , Myostatin/biosynthesis , Myostatin/genetics , PPAR gamma/agonists , SKP Cullin F-Box Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Animals , Arthritis, Experimental/drug therapy , Atrophy , Body Weight/drug effects , Eating/drug effects , Gene Expression/drug effects , Lipids/blood , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/genetics , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Organ Size/drug effects , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
20.
Amino Acids ; 40(2): 585-94, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20623149

ABSTRACT

Depletion of skeletal muscle protein mainly results from enhanced protein breakdown, caused by activation of proteolytic systems such as the Ca2+-dependent and the ATP-ubiquitin-dependent ones. In the last few years, enhanced expression and bioactivity of myostatin have been reported in several pathologies characterized by marked skeletal muscle depletion. More recently, high myostatin levels have been associated with glucocorticoid-induced hypercatabolism. The search for therapeutical strategies aimed at preventing/correcting protein hypercatabolism has been directed to inhibit humoral mediators known for their pro-catabolic action, such as TNFα. The present study has been aimed to investigate the involvement of TNFα in the regulation of both myostatin expression and intracellular protein catabolism, and the possibility to interfere with such modulations by means of amino acid supplementation. For this purpose, C2C12 myotubes exposed to TNFα in the presence or in the absence of amino acid (glutamine or leucine) supplementation have been used. Myotube treatment with TNFα leads to both hyperexpression of the muscle-specific ubiquitin ligase atrogin-1, and enhanced activity of the Ca(2+)-dependent proteolytic system. These changes are associated with increased myostatin expression. Glutamine supplementation effectively prevents TNFα-induced muscle protein loss and restores normal myostatin levels. The results shown in the present study indicate a direct involvement of TNFα in the onset of myotube protein loss and in the perturbation of myostatin-dependent signaling. In addition, the protective effect exerted by glutamine suggests that amino acid supplementation could represent a possible strategy to improve muscle mass.


Subject(s)
Down-Regulation , Glutamine/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Myostatin/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Mice , Muscle Proteins/genetics , Myostatin/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL