Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Physiol ; 9: 781, 2018.
Article in English | MEDLINE | ID: mdl-30008672

ABSTRACT

Genetic variation is associated with a number of lifestyle behaviours; it may be associated with adherence and individual responses to exercise training. We tested single nucleotide polymorphisms (SNPs) in the acid ceramidase gene (ASAH1) for association with subject adherence and physiologic benefit with exercise training in two well-characterised randomised, controlled 8-month exercise interventions: STRRIDE I (n = 239) and STRRIDE II (n = 246). Three ASAH1 non-coding SNPs in a linkage disequilibrium block were associated with non-completion: rs2898458(G/T), rs7508(A/G), and rs3810(A/G) were associated with non-completion in both additive (OR = 1.8, 1.8, 2.0; P < 0.05 all) and dominant (OR = 2.5, 2.6, 3.5; P < 0.05 all) models; with less skeletal muscle ASAH expression (p < 0.01) in a subset (N = 60); and poorer training response in cardiorespiratory fitness (peak VO2 change rs3810 r2 = 0.29, P = 0.04; rs2898458 r2 = 0.29, P = 0.08; rs7508 r2 = 0.28, p = 0.09); and similar in direction and magnitude in both independent exploratory and replication studies. Adherence to exercise may be partly biologically and genetically moderated through metabolic regulatory pathways participating in skeletal muscle adaptation to exercise training.

2.
Am J Physiol Regul Integr Comp Physiol ; 310(8): R707-10, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26864813

ABSTRACT

Respiratory dysfunction is prevalent in critically ill patients and can lead to adverse clinical outcomes, including respiratory failure and increased mortality. Respiratory muscles, which normally sustain respiration through inspiratory muscle contractions, become weakened during critical illness, and recent studies suggest that respiratory muscle weakness is related to systemic inflammation. Here, we investigate the pathophysiological role of the inflammatory JAK1/3 signaling pathway in diaphragm weakness in two distinct experimental models of critical illness. In the first experiment, mice received subcutaneous injections of PBS or C26 cancer cells and were fed chow formulated with or without the JAK1/3 inhibitor R548 for 26 days. Diaphragm specific force was significantly reduced in tumor-bearing mice receiving standard chow; however, treatment with the JAK1/3 inhibitor completely prevented diaphragm weakness. Diaphragm cross-sectional area was diminished by ∼25% in tumor-bearing mice but was similar to healthy mice in tumor-bearing animals treated with R548. In the second study, mice received sham surgery or coronary artery ligation, leading to myocardial infarction (MI), and were treated with R548 or vehicle 1 h postsurgery, and once daily for 3 days. Diaphragm specific force was comparable between sham surgery/vehicle, sham surgery/R548 and MI/R548 groups, but significantly decreased in the MI/vehicle group. Markers of oxidative damage and activated caspase-3, mechanisms previously identified to reduce muscle contractility, were not elevated in diaphragm extracts. These experiments implicate JAK1/3 signaling in cancer- and MI-mediated diaphragm weakness in mice, and provide a compelling case for further investigation.


Subject(s)
Colonic Neoplasms/drug therapy , Diaphragm/drug effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Muscle Strength/drug effects , Muscle Weakness/prevention & control , Myocardial Infarction/drug therapy , Protein Kinase Inhibitors/pharmacology , Respiration Disorders/prevention & control , Animals , Cachexia/enzymology , Cachexia/etiology , Cachexia/physiopathology , Colonic Neoplasms/complications , Colonic Neoplasms/enzymology , Colonic Neoplasms/physiopathology , Diaphragm/enzymology , Diaphragm/physiopathology , Disease Models, Animal , Janus Kinase 1/metabolism , Janus Kinase 3/metabolism , Male , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Weakness/enzymology , Muscle Weakness/etiology , Muscle Weakness/physiopathology , Myocardial Infarction/complications , Myocardial Infarction/enzymology , Myocardial Infarction/physiopathology , Respiration/drug effects , Respiration Disorders/enzymology , Respiration Disorders/etiology , Respiration Disorders/physiopathology , Signal Transduction/drug effects
3.
Mol Med ; 20: 579-89, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25286450

ABSTRACT

Mechanical ventilation (MV) is one of the lynchpins of modern intensive-care medicine and is life saving in many critically ill patients. Continuous ventilator support, however, results in ventilation-induced diaphragm dysfunction (VIDD) that likely prolongs patients' need for MV and thereby leads to major associated complications and avoidable intensive care unit (ICU) deaths. Oxidative stress is a key pathogenic event in the development of VIDD, but its regulation remains largely undefined. We report here that the JAK-STAT pathway is activated in MV in the human diaphragm, as evidenced by significantly increased phosphorylation of JAK and STAT. Blockage of the JAK-STAT pathway by a JAK inhibitor in a rat MV model prevents diaphragm muscle contractile dysfunction (by ~85%, p < 0.01). We further demonstrate that activated STAT3 compromises mitochondrial function and induces oxidative stress in vivo, and, interestingly, that oxidative stress also activates JAK-STAT. Inhibition of JAK-STAT prevents oxidative stress-induced protein oxidation and polyubiquitination and recovers mitochondrial function in cultured muscle cells. Therefore, in ventilated diaphragm muscle, activation of JAK-STAT is critical in regulating oxidative stress and is thereby central to the downstream pathogenesis of clinical VIDD. These findings establish the molecular basis for the therapeutic promise of JAK-STAT inhibitors in ventilated ICU patients.


Subject(s)
Diaphragm/metabolism , Janus Kinases/metabolism , Respiration, Artificial/adverse effects , STAT Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Adult , Aged , Aged, 80 and over , Animals , Diaphragm/physiopathology , Gene Expression Profiling , HEK293 Cells , Humans , Membrane Potential, Mitochondrial , Middle Aged , Oxidative Stress , Rats, Sprague-Dawley , Signal Transduction
4.
PLoS One ; 9(4): e94032, 2014.
Article in English | MEDLINE | ID: mdl-24710205

ABSTRACT

Numerous human diseases can lead to atrophy of skeletal muscle, and loss of this tissue has been correlated with increased mortality and morbidity rates. Clinically addressing muscle atrophy remains an unmet medical need, and the development of preclinical tools to assist drug discovery and basic research in this effort is important for advancing this goal. In this report, we describe the development of a bioluminescent gene reporter rat, based on the zinc finger nuclease-targeted insertion of a bicistronic luciferase reporter into the 3' untranslated region of a muscle specific E3 ubiquitin ligase gene, MuRF1 (Trim63). In longitudinal studies, we noninvasively assess atrophy-related expression of this reporter in three distinct models of muscle loss (sciatic denervation, hindlimb unloading and dexamethasone-treatment) and show that these animals are capable of generating refined detail on in vivo MuRF1 expression with high temporal and anatomical resolution.


Subject(s)
Luminescent Measurements/methods , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Female , Genes, Reporter , Hindlimb Suspension , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Rats , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
5.
FASEB J ; 28(7): 2790-803, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24671708

ABSTRACT

Controlled mechanical ventilation (CMV) is associated with the development of diaphragm atrophy and contractile dysfunction, and respiratory muscle weakness is thought to contribute significantly to delayed weaning of patients. Therefore, therapeutic strategies for preventing these processes may have clinical benefit. The aim of the current study was to investigate the role of the Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in CMV-mediated diaphragm wasting and weakness in rats. CMV-induced diaphragm atrophy and contractile dysfunction coincided with marked increases in STAT3 phosphorylation on both tyrosine 705 (Tyr705) and serine 727 (Ser727). STAT3 activation was accompanied by its translocation into mitochondria within diaphragm muscle and mitochondrial dysfunction. Inhibition of JAK signaling during CMV prevented phosphorylation of both target sites on STAT3, eliminated the accumulation of phosphorylated STAT3 within the mitochondria, and reversed the pathologic alterations in mitochondrial function, reduced oxidative stress in the diaphragm, and maintained normal diaphragm contractility. In addition, JAK inhibition during CMV blunted the activation of key proteolytic pathways in the diaphragm, as well as diaphragm atrophy. These findings implicate JAK/STAT3 signaling in the development of diaphragm muscle atrophy and dysfunction during CMV and suggest that the delayed extubation times associated with CMV can be prevented by inhibition of Janus kinase signaling.-Smith, I. J., Godinez, G. L., Singh, B. K., McCaughey, K. M., Alcantara, R. R., Gururaja, T., Ho, M. S., Nguyen, H. N., Friera, A. M., White, K. A., McLaughlin, J. R., Hansen, D., Romero, J. M., Baltgalvis, K. A., Claypool, M. D., Li, W., Lang, W., Yam, G. C., Gelman, M. S., Ding, R., Yung, S. L., Creger, D. P., Chen, Y., Singh, R., Smuder, A. J., Wiggs, M. P., Kwon, O.-S., Sollanek, K. J., Powers, S. K., Masuda, E. S., Taylor, V. C., Payan, D. G., Kinoshita, T., Kinsella, T. M. Inhibition of Janus kinase signaling during controlled mechanical ventilation prevents ventilation-induced diaphragm dysfunction.


Subject(s)
Diaphragm/metabolism , Janus Kinases/metabolism , Respiration, Artificial/adverse effects , Signal Transduction/physiology , Animals , Interleukin-6/metabolism , Male , Mitochondria/metabolism , Muscle Weakness/metabolism , Muscular Atrophy/metabolism , Oxidative Stress/physiology , Phosphorylation/physiology , Proteolysis , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Serine/metabolism , Tyrosine/metabolism
6.
Am J Physiol Heart Circ Physiol ; 306(8): H1128-45, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24561866

ABSTRACT

Intermittent claudication is a form of exercise intolerance characterized by muscle pain during walking in patients with peripheral artery disease (PAD). Endothelial cell and muscle dysfunction are thought to be important contributors to the etiology of this disease, but a lack of preclinical models that incorporate these elements and measure exercise performance as a primary end point has slowed progress in finding new treatment options for these patients. We sought to develop an animal model of peripheral vascular insufficiency in which microvascular dysfunction and exercise intolerance were defining features. We further set out to determine if pharmacological activation of 5'-AMP-activated protein kinase (AMPK) might counteract any of these functional deficits. Mice aged on a high-fat diet demonstrate many functional and molecular characteristics of PAD, including the sequential development of peripheral vascular insufficiency, increased muscle fatigability, and progressive exercise intolerance. These changes occur gradually and are associated with alterations in nitric oxide bioavailability. Treatment of animals with an AMPK activator, R118, increased voluntary wheel running activity, decreased muscle fatigability, and prevented the progressive decrease in treadmill exercise capacity. These functional performance benefits were accompanied by improved mitochondrial function, the normalization of perfusion in exercising muscle, increased nitric oxide bioavailability, and decreased circulating levels of the endogenous endothelial nitric oxide synthase inhibitor asymmetric dimethylarginine. These data suggest that aged, obese mice represent a novel model for studying exercise intolerance associated with peripheral vascular insufficiency, and pharmacological activation of AMPK may be a suitable treatment for intermittent claudication associated with PAD.


Subject(s)
AMP-Activated Protein Kinases/physiology , Diet, High-Fat , Enzyme Activators/administration & dosage , Obesity/complications , Peripheral Vascular Diseases/physiopathology , Physical Exertion/physiology , Aging , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Arginine/analogs & derivatives , Arginine/blood , Cilostazol , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Intermittent Claudication/complications , Intermittent Claudication/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fatigue/drug effects , Muscle, Skeletal/blood supply , Nitric Oxide Synthase Type III/metabolism , Peripheral Vascular Diseases/etiology , Phosphodiesterase 3 Inhibitors/administration & dosage , Tetrazoles/administration & dosage , Vasodilator Agents
7.
PLoS One ; 8(12): e81870, 2013.
Article in English | MEDLINE | ID: mdl-24339975

ABSTRACT

Modulation of mitochondrial function through inhibiting respiratory complex I activates a key sensor of cellular energy status, the 5'-AMP-activated protein kinase (AMPK). Activation of AMPK results in the mobilization of nutrient uptake and catabolism for mitochondrial ATP generation to restore energy homeostasis. How these nutrient pathways are affected in the presence of a potent modulator of mitochondrial function and the role of AMPK activation in these effects remain unclear. We have identified a molecule, named R419, that activates AMPK in vitro via complex I inhibition at much lower concentrations than metformin (IC50 100 nM vs 27 mM, respectively). R419 potently increased myocyte glucose uptake that was dependent on AMPK activation, while its ability to suppress hepatic glucose production in vitro was not. In addition, R419 treatment of mouse primary hepatocytes increased fatty acid oxidation and inhibited lipogenesis in an AMPK-dependent fashion. We have performed an extensive metabolic characterization of its effects in the db/db mouse diabetes model. In vivo metabolite profiling of R419-treated db/db mice showed a clear upregulation of fatty acid oxidation and catabolism of branched chain amino acids. Additionally, analyses performed using both (13)C-palmitate and (13)C-glucose tracers revealed that R419 induces complete oxidation of both glucose and palmitate to CO2 in skeletal muscle, liver, and adipose tissue, confirming that the compound increases mitochondrial function in vivo. Taken together, our results show that R419 is a potent inhibitor of complex I and modulates mitochondrial function in vitro and in diabetic animals in vivo. R419 may serve as a valuable molecular tool for investigating the impact of modulating mitochondrial function on nutrient metabolism in multiple tissues and on glucose and lipid homeostasis in diabetic animal models.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Experimental/metabolism , Mitochondria, Liver/metabolism , Muscle Cells/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Enzyme Activation/drug effects , Fatty Acids/metabolism , Glucose/metabolism , Hep G2 Cells , Humans , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Mice , Mitochondria, Liver/pathology , Muscle Cells/pathology , Oxidation-Reduction/drug effects , Palmitates/pharmacology , Protein Kinase Inhibitors/pharmacology
8.
Muscle Nerve ; 43(3): 410-4, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21305563

ABSTRACT

The influence of cancer on skeletal muscle calpain expression and activity in humans is poorly understood. We tested the hypothesis that calpain activity is increased in skeletal muscle from gastric cancer patients with no or <5% weight loss. Muscle biopsies were obtained from rectus abdominis muscle in 15 patients who underwent surgery for gastric cancer and had <5% weight loss and also in 15 control patients. Calpain activity was determined using a calpain-specific substrate in the absence or presence of calcium. The expression of µ- and m-calpain, calpastatin, atrogin-1, and MuRF1 was determined by real-time polymerase chain reaction. Calpain activity was increased by 70% in cancer patients compared with controls. There were no differences in mRNA levels for µ- and m-calpain, calpastatin, atrogin-1, or MuRF1 between control and cancer patients. Calpain activity may be increased in muscle from gastric cancer patients even before changes in molecular markers of muscle wasting and significant weight loss occur.


Subject(s)
Calpain/metabolism , Muscle, Skeletal/enzymology , Stomach Neoplasms/enzymology , Weight Loss/physiology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Aged , Enzyme Activation/physiology , Female , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/metabolism , Signal Transduction/physiology , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , Ubiquitin/metabolism
9.
J Cell Biochem ; 111(4): 1059-73, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20677217

ABSTRACT

Myostatin is a negative regulator of muscle mass and has been reported to be upregulated in several conditions characterized by muscle atrophy. The influence of sepsis on myostatin expression and activity is poorly understood. Here, we tested the hypothesis that sepsis upregulates the expression and downstream signaling of myostatin in skeletal muscle. Because sepsis-induced muscle wasting is at least in part regulated by glucocorticoids, we also determined the influence of glucocorticoids on myostatin expression. Sepsis was induced in rats by cecal ligation and puncture and control rats were sham-operated. In other experiments, rats were injected intraperitoneally with dexamethasone (10 mg/kg) or corresponding volume of vehicle. Surprisingly, myostatin mRNA levels were reduced and myostatin protein levels were unchanged in muscles from septic rats. Muscle levels of activin A, follistatin, and total and phosphorylated Smad2 (p-Smad2) were not influenced by sepsis, suggesting that myostatin downstream signaling was not altered during sepsis. Interestingly, total and p-Smad3 levels were increased in septic muscle, possibly reflecting altered signaling through pathways other than myostatin. Similar to sepsis, treatment of rats with dexamethasone reduced myostatin mRNA levels and did not alter myostatin protein levels. Fasting, an additional condition characterized by muscle wasting, reduced myostatin mRNA and activin A protein levels, increased myostatin protein, and did not influence follistatin and p-Smad2 levels. Of note, total and p-Smad3 levels were reduced in muscle during fasting. The results suggest that sepsis and glucocorticoids do not upregulate the expression and activity of myostatin in skeletal muscle. The role of myostatin may vary between different conditions characterized by muscle wasting. Downstream signaling through Smad2 and 3 is probably regulated not only by myostatin but by other mechanisms as well.


Subject(s)
Down-Regulation/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myostatin/genetics , Sepsis/metabolism , Activins/genetics , Activins/metabolism , Animals , Dexamethasone/pharmacology , Down-Regulation/drug effects , Fasting/metabolism , Follistatin/genetics , Follistatin/metabolism , Male , Mifepristone/pharmacology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Myostatin/blood , Organ Size/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Sepsis/genetics , Smad Proteins/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Am J Physiol Endocrinol Metab ; 299(4): E533-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20647557

ABSTRACT

Muscle wasting during sepsis is at least in part regulated by glucocorticoids and is associated with increased transcription of genes encoding the ubiquitin ligases atrogin-1 and muscle-specific RING-finger protein-1 (MuRF1). Recent studies suggest that muscle atrophy caused by denervation is associated with reduced expression of the nuclear cofactor peroxisome proliferator-activated receptor-γ coactivator (PGC)-1ß and that PGC-1ß may be a repressor of the atrogin-1 and MuRF1 genes. The influence of other muscle-wasting conditions on the expression of PGC-1ß is not known. We tested the influence of sepsis and glucocorticoids on PGC-1ß and examined the potential link between downregulated PGC-1ß expression and upregulated atrogin-1 and MuRF1 expression in skeletal muscle. Sepsis in rats and mice and treatment with dexamethasone resulted in downregulated expression of PGC-1ß and increased expression of atrogin-1 and MuRF1 in the fast-twitch extensor digitorum longus muscle, with less pronounced changes in the slow-twitch soleus muscle. In additional experiments, adenoviral gene transfer of PGC-1ß into cultured C2C12 myotubes resulted in a dose-dependent decrease in atrogin-1 and MuRF1 mRNA levels. Treatment of cultured C2C12 myotubes with dexamethasone or PGC-1ß small interfering RNA (siRNA) resulted in downregulated PGC-1ß expression and increased protein degradation. Taken together, our results suggest that sepsis- and glucocorticoid-induced muscle wasting may, at least in part, be regulated by decreased expression of the nuclear cofactor PGC-1ß.


Subject(s)
Glucocorticoids/pharmacology , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , RNA-Binding Proteins/biosynthesis , Sepsis/metabolism , Trans-Activators/biosynthesis , Transcription Factors/biosynthesis , Animals , Down-Regulation/drug effects , Male , Mice , Muscle Fibers, Skeletal/physiology , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscular Atrophy/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/genetics , Sepsis/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
11.
Am J Physiol Regul Integr Comp Physiol ; 299(2): R509-20, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538901

ABSTRACT

Muscle wasting during sepsis is in part regulated by glucocorticoids. In recent studies, treatment of cultured muscle cells in vitro with dexamethasone upregulated expression and activity of p300, a histone acetyl transferase (HAT), and reduced expression and activity of the histone deacetylases-3 (HDAC3) and -6, changes that favor hyperacetylation. Here, we tested the hypothesis that sepsis and glucocorticoids regulate p300 and HDAC3 and -6 in skeletal muscle in vivo. Because sepsis-induced metabolic changes are particularly pronounced in white, fast-twitch skeletal muscle, most experiments were performed in extensor digitorum longus muscles. Sepsis in rats upregulated p300 mRNA and protein levels, stimulated HAT activity, and reduced HDAC6 expression and HDAC activity. The sepsis-induced changes in p300 and HDAC expression were prevented by the glucocorticoid receptor antagonist RU38486. Treatment of rats with dexamethasone increased expression of p300 and HAT activity, reduced expression of HDAC3 and -6, and inhibited HDAC activity. Finally, treatment with the HDAC inhibitor trichostatin A resulted in increased muscle proteolysis and expression of the ubiquitin ligase atrogin-1. Taken together, our results suggest for the first time that sepsis-induced muscle wasting may be regulated by glucocorticoid-dependent hyperacetylation caused by increased p300 and reduced HDAC expression and activity. The recent development of pharmacological HDAC activators may provide a novel avenue to prevent and treat muscle wasting in sepsis and other catabolic conditions.


Subject(s)
Dexamethasone/toxicity , E1A-Associated p300 Protein/metabolism , Glucocorticoids/toxicity , Histone Deacetylases/metabolism , Muscle, Skeletal/drug effects , Muscular Atrophy/enzymology , Sepsis/enzymology , Animals , Disease Models, Animal , Down-Regulation , E1A-Associated p300 Protein/genetics , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Hormone Antagonists/pharmacology , Hydroxamic Acids/pharmacology , Male , Mifepristone/pharmacology , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Muscular Atrophy/etiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Sepsis/complications , Sirtuin 1/metabolism , Time Factors , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
12.
Curr Opin Clin Nutr Metab Care ; 13(4): 423-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20473154

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to discuss novel insight into mechanisms of glucocorticoid-regulated muscle wasting, in particular the role of transcription factors and nuclear cofactors. In addition, novel strategies that may become useful in the treatment or prevention of glucocorticoid-induced muscle wasting are reviewed. RECENT FINDINGS: Studies suggest that glucocorticoid-induced upregulation of the transcription factors Forkhead box O 1 and CCAAT/enhancer-binding protein beta and downregulation of MyoD and myogenin are involved in glucocorticoid-induced muscle wasting. In addition, glucocorticoid-induced hyperacetylation caused by increased expression of the nuclear cofactor p300 and its histone acetyl transferase activity and decreased expression and activity of histone deacetylases plays an important role in glucocorticoid-induced muscle proteolysis and wasting. Other mechanisms may also be involved in glucocorticoid-induced muscle wasting, including insulin resistance and store-operated calcium entry. Novel potential strategies to prevent or treat glucocorticoid-induced muscle wasting include the use of small molecule histone deacetylase activators, dissociated glucocorticoid receptor agonists, and 11beta-hydroxysteroid dehydrogenase type 1 inhibitors. SUMMARY: An increased understanding of molecular mechanisms regulating glucocorticoid-induced muscle wasting will help develop new strategies to prevent and treat this debilitating condition.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/adverse effects , Muscular Atrophy/genetics , Transcription Factors/metabolism , Acetylation , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , E1A-Associated p300 Protein/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Histone Deacetylases/metabolism , Humans , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenin/genetics , Myogenin/metabolism
13.
Int J Biochem Cell Biol ; 42(5): 701-11, 2010 May.
Article in English | MEDLINE | ID: mdl-20079455

ABSTRACT

Sepsis-induced muscle wasting has severe clinical consequences, including muscle weakness, need for prolonged ventilatory support and stay in the intensive care unit, and delayed ambulation with risk for pulmonary and thromboembolic complications. Understanding molecular mechanisms regulating loss of muscle mass in septic patients therefore has significant clinical implications. Forkhead Box O (FOXO) transcription factors have been implicated in muscle wasting, partly reflecting upregulation of the ubiquitin ligases atrogin-1 and MuRF1. The influence of sepsis on FOXO transcription factors in skeletal muscle is poorly understood. We tested the hypothesis that sepsis upregulates expression and activity of FOXO transcription factors in skeletal muscle by a glucocorticoid-dependent mechanism. Sepsis in rats increased muscle FOXO1 and 3a mRNA and protein levels but did not influence FOXO4 expression. Nuclear FOXO1 levels and DNA binding activity were increased in septic muscle whereas FOXO3a nuclear levels were not increased during sepsis. Sepsis-induced expression of FOXO1 was reduced by the glucocorticoid receptor antagonist RU38486 and treatment of rats with dexamethasone increased FOXO1 mRNA levels suggesting that the expression of FOXO1 is regulated by glucocorticoids. Reducing FOXO1, but not FOXO3a, expression by siRNA in cultured L6 myotubes inhibited dexamethasone-induced atrogin-1 and MuRF1 expression, further supporting a role of FOXO1 in glucocorticoid-regulated muscle wasting. Results suggest that sepsis increases FOXO1 expression and activity in skeletal muscle by a glucocorticoid-dependent mechanism and that glucocorticoid-dependent upregulation of atrogin-1 and MuRF1 in skeletal muscle is regulated by FOXO1. The study is significant because it provides novel information about molecular mechanisms involved in sepsis-induced muscle wasting.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Sepsis/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Male , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscular Atrophy/etiology , Muscular Atrophy/physiopathology , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Transport , RNA, Messenger/metabolism , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Sepsis/complications , Time Factors , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Physiol Genomics ; 36(3): 149-57, 2009 Feb 02.
Article in English | MEDLINE | ID: mdl-19033545

ABSTRACT

Endurance exercise (EE) leads to beneficial alterations in skeletal muscle lipid metabolism in overweight and obese individuals; however, the mechanisms of these improvements are poorly understood. The primary goal of the current investigation was to test the hypothesis that long-term EE training (6 mo) leads to alterations in the mRNA abundance of key lipid metabolism enzymes in skeletal muscle of overweight and obese middle-aged women and men. A secondary aim of this study was to investigate the hypothesis that exercise-mediated adaptations in mRNA levels differ between women and men. The mRNA abundance of representative lipogenic and lipolytic genes from major lipid metabolism pathways, as well as representative lipogenic and lipolytic transcription factors, were determined by real-time PCR from skeletal muscle biopsies collected before and approximately 24 h after the final bout of 6 mo of EE. Six months of EE led to increases in muscle lipoprotein lipase, peroxisome proliferator-activated receptor-gamma coactivator-1alpha, carnitine palmitoyltransferase-1 beta, diacylglycerol acyltransferase-1, and acid ceramidase mRNA in women, but not men. In contrast, in men, EE led to reductions in the mRNA content of the lipogenic factors sterol regulatory element binding protein-1c and serine palmitoyl transferase. These data suggest that EE-mediated alterations in the abundance of the lipid metabolism genes studied here are fundamentally different between overweight and obese middle-aged women and men. Future studies should determine whether these adaptations in mRNA levels translate into changes in protein function.


Subject(s)
Enzymes/genetics , Exercise/physiology , Muscle, Skeletal/metabolism , Obesity/therapy , Overweight/therapy , Adult , Aged , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Enzymes/metabolism , Female , Gene Expression Profiling , Humans , Lipid Metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Male , Middle Aged , Obesity/genetics , Obesity/metabolism , Overweight/genetics , Overweight/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
15.
Am J Physiol Endocrinol Metab ; 295(4): E762-71, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18492780

ABSTRACT

Muscle wasting in sepsis reflects activation of multiple proteolytic mechanisms, including lyosomal and ubiquitin-proteasome-dependent protein breakdown. Recent studies suggest that activation of the calpain system also plays an important role in sepsis-induced muscle wasting. Perhaps the most important consequence of calpain activation in skeletal muscle during sepsis is disruption of the sarcomere, allowing for the release of myofilaments (including actin and myosin) that are subsequently ubiquitinated and degraded by the 26S proteasome. Other important consequences of calpain activation that may contribute to muscle wasting during sepsis include degradation of certain transcription factors and nuclear cofactors, activation of the 26S proteasome, and inhibition of Akt activity, allowing for downstream activation of Foxo transcription factors and GSK-3beta. The role of calpain activation in sepsis-induced muscle wasting suggests that the calpain system may be a therapeutic target in the prevention and treatment of muscle wasting during sepsis. Furthermore, because calpain activation may also be involved in muscle wasting caused by other conditions, including different muscular dystrophies and cancer, calpain inhibitors may be beneficial not only in the treatment of sepsis-induced muscle wasting but in other conditions causing muscle atrophy as well.


Subject(s)
Calpain/physiology , Muscle, Skeletal/pathology , Sepsis/complications , Sepsis/pathology , Wasting Syndrome/etiology , Wasting Syndrome/pathology , Animals , Calcium/metabolism , Calpain/antagonists & inhibitors , Calpain/genetics , Humans , Muscle, Skeletal/physiopathology , Proto-Oncogene Proteins c-akt/physiology , Sarcomeres/physiology , Sepsis/drug therapy , Transcription Factors , Wasting Syndrome/drug therapy
16.
Exp Physiol ; 92(3): 561-73, 2007 May.
Article in English | MEDLINE | ID: mdl-17272355

ABSTRACT

The role of the calpain proteases in skeletal muscle atrophy is poorly understood. One goal of these experiments was to clarify whether calpains act upstream of the ubiquitin-proteasome pathway (UPP). Calpain activation may also inhibit the anabolic signalling of Akt, since a molecular chaperone previously shown to mediate Akt activity, heat shock protein 90 (HSP 90), is a calpain substrate. Thus, an additional objective was to determine whether calpain activation affects the Akt signalling pathway. Ex vivo experiments were conducted using isolated rat diaphragm muscle. Calpain activation increased total protein degradation by 65%. Proteasome inhibition prevented this large rise in proteolysis, demonstrating that the proteasome was necessary for calpain-activated protein degradation. In addition, calpain activation increased proteasome-dependent proteolysis by 144%, further supporting the idea of sequential proteolytic pathways. Calpain reduced Akt and mammalian target of rapamycin (mTOR) phosphorylation by 35 and 50%, respectively, and activated glycogen synthase kinase-3 beta (GSK-3beta) by 40%. Additionally, calpain activation reduced HSP 90beta and mTOR protein content by 33 and 50%, respectively. These data suggest that calpains play a dual role in protein metabolism by concomitantly activating proteasome-dependent proteolysis and inhibiting the Akt pathway of protein synthesis.


Subject(s)
Calpain/physiology , Diaphragm/physiology , Muscle Proteins/metabolism , Proteasome Endopeptidase Complex/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Animals , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HSP90 Heat-Shock Proteins/metabolism , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL