Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
NPJ Parkinsons Dis ; 8(1): 159, 2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36396647

ABSTRACT

Parkinson's Disease (PD) is a chronic and progressive neurodegenerative disease manifesting itself with tremors, muscle stiffness, bradykinesia, dementia, and depression. Mutations of mitochondrial E3 ligase, PARKIN, have been associated with juvenile PD. Previous studies have characterized muscle atrophy and motor deficits upon loss of functional Parkin in fly and rodent models. However, the mechanisms behind pathophysiology of Parkin deficient muscle remains to be elusive. Here, results suggested that knock down of Parkin significantly increases proteolytic activities in skeletal muscle cell line, the C2C12 myotubes. However, the atrogene levels increase moderately in Parkin deficient cell line. To further investigate the role of Parkin in skeletal muscle atrophy, Parkin knock out (KO) and wild type mice were subjected to 48 h starvation. After 48 h fasting, a greater reduction in skeletal muscle weights was observed in Parkin KO mice as compared to age matched wild type control, suggesting elevated proteolytic activity in the absence of Parkin. Subsequent microarray analyses revealed further enhanced expression of FOXO and ubiquitin pathway in fasted Parkin KO mice. Furthermore, a greater reduction in the expression of cytoskeleton genes was observed in Parkin KO mice following 48 h fasting. Collectively, these results suggest that Parkin deficiency exacerbates fasting-induced skeletal muscle wasting, through upregulating genes involved in catabolic activities in skeletal muscle.

2.
Am J Physiol Cell Physiol ; 315(2): C164-C185, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29561660

ABSTRACT

Parkinson's disease is a neurodegenerative disease characterized by tremors, muscle stiffness, and muscle weakness. Molecular genetic analysis has confirmed that mutations in PARKIN and PINK1 genes, which play major roles in mitochondrial quality control and mitophagy, are frequently associated with Parkinson's disease. PARKIN is an E3 ubiquitin ligase that translocates to mitochondria during loss of mitochondrial membrane potential to increase mitophagy. Although muscle dysfunction is noted in Parkinson's disease, little is known about the involvement of PARKIN in the muscle phenotype of Parkinson's disease. In this study, we report that the mitochondrial uncoupler CCCP promotes PINK1/PARKIN-mediated mitophagy in myogenic C2C12 cells. As a result of this excess mitophagy, we show that CCCP treatment of myotubes leads to the development of myotube atrophy in vitro. Surprisingly, we also found that siRNA-mediated knockdown of Parkin results in impaired mitochondrial turnover. In addition, knockdown of Parkin led to myotubular atrophy in vitro. Consistent with these in vitro results, Parkin knockout muscles showed impaired mitochondrial function and smaller myofiber area, suggesting that Parkin function is required for post-natal skeletal muscle growth and development.


Subject(s)
Mitochondria/metabolism , Muscular Atrophy/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mitophagy/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Kinases/metabolism
3.
Oncotarget ; 8(58): 98553-98566, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228710

ABSTRACT

BACKGROUND: Irisin is an exercise induced myokine that is shown to promote browning of adipose tissue and hence, increase energy expenditure. Furthermore, our unpublished results indicate that Irisin improves myogenic differentiation and induces skeletal muscle hypertrophy. Since exercise induced skeletal muscle hypertrophy improves muscle strength, we wanted to investigate if ectopic injection of Irisin peptide improves skeletal muscle function in a mouse model of muscular dystrophy. This utility of Irisin peptide is yet to be studied in animal models. METHODS: In order to test this hypothesis, we expressed and purified recombinant murine Irisin peptide from E. coli. Three- to six-week-old male mdx mice were injected IP with either vehicle (dialysis buffer) or Irisin recombinant peptide for two or four weeks, three times-a-week. RESULTS: Irisin injection increased muscle weights and enhanced grip strength in mdx mice. Improved muscle strength can be attributed to the significant hypertrophy observed in the Irisin injected mdx mice. Moreover, Irisin treatment resulted in reduced accumulation of fibrotic tissue and myofiber necrosis in mdx mice. In addition, Irisin improved sarcolemmal stability, which is severely compromised in mdx mice. CONCLUSION: Irisin injection induced skeletal muscle hypertrophy, improved muscle strength and reduced necrosis and fibrotic tissue in a murine dystrophy model. These results demonstrate the potential therapeutic value of Irisin in muscular dystrophy.

4.
Nat Commun ; 8(1): 1104, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29062100

ABSTRACT

Exercise induces expression of the myokine irisin, which is known to promote browning of white adipose tissue and has been shown to mediate beneficial effects following exercise. Here we show that irisin induces expression of a number of pro-myogenic and exercise response genes in myotubes. Irisin increases myogenic differentiation and myoblast fusion via activation of IL6 signaling. Injection of irisin in mice induces significant hypertrophy and enhances grip strength of uninjured muscle. Following skeletal muscle injury, irisin injection improves regeneration and induces hypertrophy. The effects of irisin on hypertrophy are due to activation of satellite cells and enhanced protein synthesis. In addition, irisin injection rescues loss of skeletal muscle mass following denervation by enhancing satellite cell activation and reducing protein degradation. These data suggest that irisin functions as a pro-myogenic factor in mice.


Subject(s)
Atrophy/prevention & control , Fibronectins/metabolism , Hypertrophy/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Adipose Tissue, White/metabolism , Animals , Atrophy/etiology , Atrophy/genetics , Atrophy/metabolism , Denervation/adverse effects , Fibronectins/administration & dosage , Fibronectins/genetics , Humans , Hypertrophy/genetics , Hypertrophy/physiopathology , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Stem Cells/cytology , Stem Cells/metabolism
7.
J Am Med Dir Assoc ; 17(1): 65-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26410107

ABSTRACT

BACKGROUND: Many recent guidelines and consensus on sarcopenia have incorporated gait speed and grip strength as diagnostic criteria without addressing early posture changes adopted to maintain gait speed before weakness is clinically evident. OBJECTIVES: Older adults are known to compensate well for declining physiological reserve through environmental modification and posture adaptation. This study aimed to analyze and identify significant posture adaptation in older adults that is required to maintain gait speed in the face of increasing vulnerability. This would be a useful guide for early posture correction exercise interventions to prevent further decline, in addition to traditional gait, balance, and strength training. DESIGN: A community-based cross-sectional study. SETTING AND PARTICIPANTS: The participants comprised 90 healthy community-dwelling Chinese men between the ages of 60 and 80 years and 20 Chinese adults between the ages of 21 and 50 years within the normal BMI range as a comparison group. MEASUREMENTS: All the participants underwent handgrip strength testing, 6-minute walk, timed up-and-go (TUG), and motion analysis for gait characteristics. Low function was characterized by slow walking speed (<1.0 m/s) and/or slow TUG (>10 seconds), whereas low strength was determined by hand grip dynamometer testing (<26 kg). The degree of frailty was classified using the Canadian Study for Health and Ageing Clinical Frailty Scale (CSHA-CFS) to differentiate between healthy and vulnerable older adults. RESULTS: As expected, the vulnerable older adults had lower functional performance and strength compared with the healthy older adults group. However, a significant number demonstrated posture adaptations in walking in all 3 groups, including those who maintained a good walking speed (>1.0 m/s). The extent of such adaptation was larger in the vulnerable group as compared with the healthy group. CONCLUSION: Although gait speed is a robust parameter for screening older adults for sarcopenia and frailty, our data suggest that identifying trunk posture adaptation before the onset of decline in gait speed will help in planning interventions in the at-risk community-dwelling older adults even before gait speed declines.


Subject(s)
Adaptation, Physiological , Frail Elderly , Gait/physiology , Posture/physiology , Torso/physiology , Walking/physiology , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Exercise Test , Hand Strength/physiology , Humans , Male , Middle Aged , Sarcopenia/physiopathology , Young Adult
9.
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
10.
Am J Physiol Endocrinol Metab ; 309(2): E122-31, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25921579

ABSTRACT

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a ubiquitously expressed gene with higher levels observed in skeletal muscle. Recently, our laboratory showed (Bonala S, Lokireddy S, Arigela H, Teng S, Wahli W, Sharma M, McFarlane C, Kambadur R. J Biol Chem 287: 12935-12951, 2012) that PPARß/δ modulates myostatin activity to induce myogenesis in skeletal muscle. In the present study, we show that PPARß/δ-null mice display reduced body weight, skeletal muscle weight, and myofiber atrophy during postnatal development. In addition, a significant reduction in satellite cell number was observed in PPARß/δ-null mice, suggesting a role for PPARß/δ in muscle regeneration. To investigate this, tibialis anterior muscles were injured with notexin, and muscle regeneration was monitored on days 3, 5, 7, and 28 postinjury. Immunohistochemical analysis revealed an increased inflammatory response and reduced myoblast proliferation in regenerating muscle from PPARß/δ-null mice. Histological analysis confirmed that the regenerated muscle fibers of PPARß/δ-null mice maintained an atrophy phenotype with reduced numbers of centrally placed nuclei. Even though satellite cell numbers were reduced before injury, satellite cell self-renewal was found to be unaffected in PPARß/δ-null mice after regeneration. Previously, our laboratory had showed (Bonala S, Lokireddy S, Arigela H, Teng S, Wahli W, Sharma M, McFarlane C, Kambadur R. J Biol Chem 287: 12935-12951, 2012) that inactivation of PPARß/δ increases myostatin signaling and inhibits myogenesis. Our results here indeed confirm that inactivation of myostatin signaling rescues the atrophy phenotype and improves muscle fiber cross-sectional area in both uninjured and regenerated tibialis anterior muscle from PPARß/δ-null mice. Taken together, these data suggest that absence of PPARß/δ leads to loss of satellite cells, impaired skeletal muscle regeneration, and postnatal myogenesis. Furthermore, our results also demonstrate that functional antagonism of myostatin has utility in rescuing these effects.


Subject(s)
Muscle Development/genetics , PPAR delta/genetics , PPAR-beta/genetics , Satellite Cells, Skeletal Muscle/metabolism , Animals , Down-Regulation/genetics , Gene Silencing , Growth and Development/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Muscular Atrophy/genetics
12.
J Cell Biochem ; 115(11): 1908-17, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24909401

ABSTRACT

Conversion of skin fibroblasts into myoblasts by transducing the cells with myogenic master regulator MyoD has been in practice for more than two decades. The purpose of such conversion is due to scarcity of muscle biopsies during muscle wasting, hence conversion of fibroblasts to myogenic lineage from various genetic backgrounds offers a great alternative for cell therapies. Here, we have investigated if eliminating Myostatin, a potent negative regulator of myogenesis, could improve the myogenic conversion of fibroblasts. In the present study, we have isolated primary muscle fibroblasts from the skeletal muscles of wild-type (WT) and myostatin null (Mstn(-/-)) mice and transduced the muscle fibroblasts with MyoD using adenoviral, lentiviral transduction, and electroporation methods. In contrast to what we predicted, it is only in WT muscle fibroblasts we detected significant ectopic expression of MyoD, and myogenic conversion. Muscle fibroblasts from Mstn(-/-) genotype failed to take up as much MyoD using the three methods and, therefore, failed to form myotubes. The aforesaid condition of greater MyoD uptake by WT muscle fibroblasts was attributed to the presence of adenoviral receptors, which facilitated adenoviral transduction. However, in Mstn(-/-) fibroblasts we detected negligible levels of adenovirus receptors. Moreover, we also detected significantly higher levels of MyoD antagonists, c-Fos, c-Jun, and cyclin D1 in Mstn(-/-) muscle fibroblasts. Taken together, our results demonstrate that lack of myostatin reduces myogenic potential of muscle fibroblasts by inhibiting MyoD function.


Subject(s)
Fibroblasts/cytology , Muscle Development , Muscle, Skeletal/cytology , MyoD Protein/genetics , Myostatin/deficiency , Animals , Cell Differentiation , Cells, Cultured , Cyclin D1/metabolism , Genetic Therapy , Mice , Mice, Transgenic , MyoD Protein/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transduction, Genetic
13.
PLoS One ; 9(1): e87687, 2014.
Article in English | MEDLINE | ID: mdl-24498167

ABSTRACT

Growth factors, such as myostatin (Mstn), play an important role in regulating post-natal myogenesis. In fact, loss of Mstn has been shown to result in increased post-natal muscle growth through enhanced satellite cell functionality; while elevated levels of Mstn result in dramatic skeletal muscle wasting through a mechanism involving reduced protein synthesis and increased ubiquitin-mediated protein degradation. Here we show that miR-27a/b plays an important role in feed back auto-regulation of Mstn and thus regulation of post-natal myogenesis. Sequence analysis of Mstn 3' UTR showed a single highly conserved miR-27a/b binding site and increased expression of miR-27a/b was correlated with decreased expression of Mstn and vice versa both in vitro and in mice in vivo. Moreover, we also show that Mstn gene expression was regulated by miR-27a/b. Treatment with miR-27a/b-specific AntagomiRs resulted in increased Mstn expression, reduced myoblast proliferation, impaired satellite cell activation and induction of skeletal muscle atrophy that was rescued upon either blockade of, or complete absence of, Mstn. Consistent with this, miR-27a over expression resulted in reduced Mstn expression, skeletal muscle hypertrophy and an increase in the number of activated satellite cells, all features consistent with impaired Mstn function. Loss of Smad3 was associated with increased levels of Mstn, concomitant with decreased miR-27a/b expression, which is consistent with impaired satellite cell function and muscular atrophy previously reported in Smad3-null mice. Interestingly, treatment with Mstn resulted in increased miR-27a/b expression, which was shown to be dependent on the activity of Smad3. These data highlight a novel auto-regulatory mechanism in which Mstn, via Smad3 signaling, regulates miR-27a/b and in turn its own expression. In support, Mstn-mediated inhibition of Mstn 3' UTR reporter activity was reversed upon miR-27a/b-specific AntagomiR transfection. Therefore, miR-27a/b, through negatively regulating Mstn, plays a role in promoting satellite cell activation, myoblast proliferation and preventing muscle wasting.


Subject(s)
Gene Expression Regulation/physiology , MicroRNAs/metabolism , Muscle Development/physiology , Myostatin/biosynthesis , Satellite Cells, Skeletal Muscle/metabolism , Smad3 Protein/metabolism , 3' Untranslated Regions/physiology , Animals , Cell Line , Male , Mice , Mice, Mutant Strains , MicroRNAs/genetics , Myostatin/genetics , Satellite Cells, Skeletal Muscle/cytology , Smad3 Protein/genetics
14.
J Biol Chem ; 289(9): 5784-98, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24425880

ABSTRACT

One of the features of uncontrolled type 1 diabetes is oxidative stress that induces DNA damage and cell death. Skeletal muscle atrophy is also considerable in type 1 diabetes, however, the signaling mechanisms that induce oxidative stress culminating in muscle atrophy are not fully known. Here, we show that in Streptozotocin-induced diabetic wild type mice, hypo-phosphorylation of Akt, resulted in activation of Foxa2 transcription factor in the muscle. Foxa2 transcriptionally up-regulated Myostatin, contributing to exaggerated oxidative stress leading to DNA damage via p63/REDD1 pathway in skeletal muscle of Streptozotocin-treated wild type mice. In Myostatin(-/-) mice however, Streptozotocin treatment did not reduce Akt phosphorylation despite reduced IRS-1 signaling. Moreover, Foxa2 levels remained unaltered in Myostatin(-/-) mice, while levels of p63/REDD1 were higher compared with wild type mice. Consistent with these results, relatively less DNA damage and muscle atrophy was observed in Myostatin(-/-) muscle in response to Streptozotocin treatment. Taken together, our results for the first time show the role of Foxa2 in Myostatin regulation in skeletal muscle in diabetic mice. Altogether, these results demonstrate the mechanism by which Myostatin contributes to DNA damage in skeletal muscle of the diabetic mice that would lead to myofiber degeneration.


Subject(s)
DNA Damage , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Muscle Fibers, Skeletal/metabolism , Myostatin/metabolism , Oxidative Stress , Animals , Cell Line , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Myostatin/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Biol Chem ; 289(11): 7654-70, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24451368

ABSTRACT

To date a plethora of evidence has clearly demonstrated that continued high calorie intake leads to insulin resistance and type-2 diabetes with or without obesity. However, the necessary signals that initiate insulin resistance during high calorie intake remain largely unknown. Our results here show that in response to a regimen of high fat or high glucose diets, Mstn levels were induced in muscle and liver of mice. High glucose- or fat-mediated induction of Mstn was controlled at the level of transcription, as highly conserved carbohydrate response and sterol-responsive (E-box) elements were present in the Mstn promoter and were revealed to be critical for ChREBP (carbohydrate-responsive element-binding protein) or SREBP1c (sterol regulatory element-binding protein 1c) regulation of Mstn expression. Further molecular analysis suggested that the increased Mstn levels (due to high glucose or fatty acid loading) resulted in increased expression of Cblb in a Smad3-dependent manner. Casitas B-lineage lymphoma b (Cblb) is an ubiquitin E3 ligase that has been shown to specifically degrade insulin receptor substrate 1 (IRS1) protein. Consistent with this, our results revealed that elevated Mstn levels specifically up-regulated Cblb, resulting in enhanced ubiquitin proteasome-mediated degradation of IRS1. In addition, over expression or knock down of Cblb had a major impact on IRS1 and pAkt levels in the presence or absence of insulin. Collectively, these observations strongly suggest that increased glucose levels and high fat diet, both, result in increased circulatory Mstn levels. The increased Mstn in turn is a potent inducer of insulin resistance by degrading IRS1 protein via the E3 ligase, Cblb, in a Smad3-dependent manner.


Subject(s)
Diet/adverse effects , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Myostatin/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Smad3 Protein/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Dietary Fats/administration & dosage , Gene Expression Regulation , Glucose/administration & dosage , Glucose/metabolism , Hep G2 Cells , Humans , Lentivirus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Muscle, Skeletal/metabolism , Nuclear Proteins/metabolism , Palmitates/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription Factors/metabolism
16.
Mol Endocrinol ; 28(3): 317-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24438338

ABSTRACT

Smad (Sma and Mad-related protein) 2/3 are downstream signaling molecules for TGF-ß and myostatin (Mstn). Recently, Mstn was shown to induce reactive oxygen species (ROS) in skeletal muscle via canonical Smad3, nuclear factor-κB, and TNF-α pathway. However, mice lacking Smad3 display skeletal muscle atrophy due to increased Mstn levels. Hence, our aims were first to investigate whether Mstn induced muscle atrophy in Smad3(-/-) mice by increasing ROS and second to delineate Smad3-independent signaling mechanism for Mstn-induced ROS. Herein we show that Smad3(-/-) mice have increased ROS levels in skeletal muscle, and inactivation of Mstn in these mice partially ablates the oxidative stress. Furthermore, ROS induction by Mstn in Smad3(-/-) muscle was not via nuclear factor-κB (p65) signaling but due to activated p38, ERK MAPK signaling and enhanced IL-6 levels. Consequently, TNF-α, nicotinamide adenine dinucleotide phosphate oxidase, and xanthine oxidase levels were up-regulated, which led to an increase in ROS production in Smad3(-/-) skeletal muscle. The exaggerated ROS in the Smad3(-/-) muscle potentiated binding of C/EBP homology protein transcription factor to MuRF1 promoter, resulting in enhanced MuRF1 levels leading to muscle atrophy.


Subject(s)
Muscle Proteins/genetics , Myostatin/physiology , Smad3 Protein/genetics , Transcription Factor RelA/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , CHO Cells , Catalase/metabolism , Cricetinae , Cricetulus , Electron Transport Chain Complex Proteins/metabolism , Female , Gene Expression , Glutathione Peroxidase/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Smad3 Protein/deficiency , Transcription Factor CHOP/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
17.
Diabetes ; 63(3): 1093-102, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24353181

ABSTRACT

We previously showed that ethnicity modifies the association between adiposity and insulin resistance. We sought to determine whether differential body fat partitioning or abnormalities in muscle insulin signaling associated with higher levels of adiposity might underlie this observation. We measured the insulin sensitivity index (ISI), percentage of body fat (%body fat), visceral (VAT) and subcutaneous (SAT) adipose tissue, liver fat, and intramyocellular lipids (IMCL) in 101 Chinese, 82 Malays, and 81 South Asians, as well as phosphorylated (p)-Akt levels in cultured myoblasts from Chinese and South Asians. Lean Chinese and Malays had higher ISI than South Asians. Although the ISI was lower in all ethnic groups when %body fat was higher, this association was stronger in Chinese and Malays, such that no ethnic differences were observed in overweight individuals. These ethnic differences were observed even when %body fat was replaced with fat in other depots. Myoblasts obtained from lean South Asians had lower p-Akt levels than those from lean Chinese. Higher adiposity was associated with lower p-Akt levels in Chinese but not in South Asians, and no ethnic differences were observed in overweight individuals. With higher %body fat, Chinese exhibited smaller increases in deep SAT and IMCL compared with Malays and South Asians, which did not explain the ethnic differences observed. Our study suggests that body fat partitioning does not explain interethnic differences in insulin sensitivity among Asian ethnic groups. Although higher adiposity had greater effect on skeletal muscle insulin sensitivity among Chinese, obesity-independent pathways may be more relevant in South Asians.


Subject(s)
Adipose Tissue/metabolism , Insulin Resistance/ethnology , Adiposity , Adult , Asian People , Humans , Intra-Abdominal Fat/metabolism , Male , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Singapore , Subcutaneous Fat/metabolism
18.
Mol Endocrinol ; 27(9): 1518-35, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23927930

ABSTRACT

Obesity is associated with insulin resistance and abnormal peripheral tissue glucose uptake. However, the mechanisms that interfere with insulin signaling and glucose uptake in human skeletal muscle during obesity are not fully characterized. Using microarray, we have identified that the expression of Pid1 gene, which encodes for a protein that contains a phosphotyrosine-interacting domain, is increased in myoblasts established from overweight insulin-resistant individuals. Molecular analysis further validated that both Pid1 mRNA and protein levels are increased in cell culture models of insulin resistance. Consistent with these results, overexpression of phosphotyrosine interaction domain-containing protein 1 (PID1) in human myoblasts resulted in reduced insulin signaling and glucose uptake, whereas knockdown of PID1 enhanced glucose uptake and insulin signaling in human myoblasts and improved the insulin sensitivity following palmitate-, TNF-α-, or myostatin-induced insulin resistance in human myoblasts. Furthermore, the number of mitochondria in myoblasts that ectopically express PID1 was significantly reduced. In addition to overweight humans, we find that Pid1 levels are also increased in all 3 peripheral tissues (liver, skeletal muscle, and adipose tissue) in mouse models of diet-induced obesity and insulin resistance. An in silico search for regulators of Pid1 expression revealed the presence of nuclear factor-κB (NF-κB) binding sites in the Pid1 promoter. Luciferase reporter assays and chromatin immunoprecipitation studies confirmed that NF-κB is sufficient to transcriptionally up-regulate the Pid1 promoter. Furthermore, we find that myostatin up-regulates Pid1 expression via an NF-κB signaling mechanism. Collectively these results indicate that Pid1 is a potent intracellular inhibitor of insulin signaling pathway during obesity in humans and mice.


Subject(s)
Carrier Proteins/metabolism , Insulin Resistance , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/pathology , Adult , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Knockdown Techniques , Humans , Insulin/metabolism , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Models, Biological , Muscle, Skeletal/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Myostatin/metabolism , NF-kappa B/metabolism , Obesity/genetics , Palmitic Acid/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , Young Adult
19.
J Biol Chem ; 288(9): 6663-78, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23297411

ABSTRACT

High doses of dexamethasone (Dex) or myostatin (Mstn) induce severe atrophy of skeletal muscle. Here we show a novel microRNA1 (miR1)-mediated mechanism through which Dex promotes skeletal muscle atrophy. Using both C2C12 myotubes and mouse models of Dex-induced atrophy we show that Dex induces miR1 expression through glucocorticoid receptor (GR). We further show that Mstn treatment facilitates GR nuclear translocation and thereby induces miR1 expression. Inhibition of miR1 in C2C12 myotubes attenuated the Dex-induced increase in atrophy-related proteins confirming a role for miR1 in atrophy. Analysis of miR1 targets revealed that HSP70 is regulated by miR1 during atrophy. Our results demonstrate that increased miR1 during atrophy reduced HSP70 levels, which resulted in decreased phosphorylation of AKT, as HSP70 binds to and protects phosphorylation of AKT. We further show that loss of pAKT leads to decreased phosphorylation, and thus, enhanced activation of FOXO3, up-regulation of MuRF1 and Atrogin-1, and progression of skeletal muscle atrophy. Based on these results, we propose a model whereby Dex- and Mstn-mediated atrophic signals are integrated through miR1, which then either directly or indirectly, inhibits the proteins involved in providing protection against atrophy.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Dexamethasone/adverse effects , HSP70 Heat-Shock Proteins/metabolism , MicroRNAs/metabolism , Models, Biological , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , CHO Cells , Cricetinae , Cricetulus , Dexamethasone/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HSP70 Heat-Shock Proteins/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , SKP Cullin F-Box Protein Ligases/biosynthesis , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
20.
Cell Metab ; 16(5): 613-24, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23140641

ABSTRACT

Recent research reveals that dysfunction and subsequent loss of mitochondria (mitophagy) is a potent inducer of skeletal muscle wasting. However, the molecular mechanisms that govern the deregulation of mitochondrial function during muscle wasting are unclear. In this report, we show that different muscle-wasting stimuli upregulated mitochondrial E3 ubiquitin protein ligase 1 (Mul1), through a mechanism involving FoxO1/3 transcription factors. Overexpression of Mul1 in skeletal muscles and myoblast cultures was sufficient for the induction of mitophagy. Consistently, Mul1 suppression not only protected against mitophagy but also partially rescued the muscle wasting observed in response to muscle-wasting stimuli. In addition, upregulation of Mul1, while increasing mitochondrial fission, resulted in ubiquitination and degradation of the mitochondrial fusion protein Mfn2. Collectively, these data explain the molecular basis for the loss of mitochondrial number during muscle wasting.


Subject(s)
Mitochondrial Proteins/metabolism , Mitophagy/drug effects , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Dexamethasone/pharmacology , Forkhead Transcription Factors/metabolism , GTP Phosphohydrolases/metabolism , Glucocorticoids/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myoblasts/metabolism , Myostatin/pharmacology , Signal Transduction/drug effects , Ubiquitination , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...