Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Article in English | MEDLINE | ID: mdl-32947810

ABSTRACT

A growing number of developing countries have experienced worsening air pollution, which has been shown to cause significant health problems. However, few studies have explored the impact of air pollution on the mental health of university students, particularly in the Chinese context. In order to address this gap, through a large-scale cross-sectional survey, this study aims to examine the effects of air pollution on final-year Chinese university undergraduates' (due to graduate in 2020) mental health by employing multivariable logistic regression. Our findings show that, first, although normal air quality is not strongly associated with lower levels of negative mental health, there is a strong link between poor air quality and higher levels of negative mental health. More specifically, life satisfaction hedonic unhappiness and depression measured by the Centre for Epidemiological Studies' Depression scale (CES-D) are statistically associated with air pollution. In addition, we also found that gender is a significant factor, as males had more than 1.6 times greater odds of increased mental health problems compared to their female counterparts. Place of birth also plays a significant role in participants' mental health. Moreover, undergraduates with urban household registration experienced significant levels of hedonic unhappiness and depression on the CES-D scale. Finally, we found that there is an association between respondents' economic situation and their mental health too. Overall, this study contributes to the research on air pollution management and mental health intervention, particularly in relation to student groups. The undergraduate curriculum should provide more guidance and suggestions on promoting mental health and establishing positive attitudes to life and academic study of the final year students, under the context of air pollution in China.


Subject(s)
Air Pollution , Mental Health , Adolescent , Adult , Air Pollution/adverse effects , Air Pollution/analysis , China/epidemiology , Cross-Sectional Studies , Female , Humans , Male , Students , Universities , Young Adult
2.
Am J Physiol Endocrinol Metab ; 316(5): E931-E939, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30888859

ABSTRACT

The objective of this study is to determine whether AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), or peroxisome proliferator-activated receptor ß (PPARß) can independently mediate the increase of glucose transporter type 4 (GLUT4) expression that occurs in response to exercise training. We found that PPARß can regulate GLUT4 expression without PGC-1α. We also found AMPK and PPARß are important for maintaining normal physiological levels of GLUT4 protein in the sedentary condition as well following exercise training. However, AMPK and PPARß are not essential for the increase in GLUT4 protein expression that occurs in response to exercise training. We discovered that AMPK activation increases PPARß via myocyte enhancer factor 2A (MEF2A), which acted as a transcription factor for PPARß. Furthermore, exercise training increases the cooperation of AMPK and PPARß to regulate glucose uptake. In conclusion, cooperation between AMPK and PPARß via NRF-1/MEF2A pathway enhances the exercise training mediated adaptive increase in GLUT4 expression and subsequent glucose uptake in skeletal muscle.


Subject(s)
Adenylate Kinase/metabolism , Glucose Transporter Type 4/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , PPAR-beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal , Animals , Cell Line , Electroporation , Feedback, Physiological , Glucose/metabolism , MEF2 Transcription Factors/metabolism , Mice , Nuclear Respiratory Factor 1/metabolism , Rats
3.
Cell Metab ; 25(5): 1176-1185.e5, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28467933

ABSTRACT

The objective of this study was to evaluate the specific mechanism(s) by which PPARß regulates mitochondrial content in skeletal muscle. We discovered that PPARß increases PGC-1α by protecting it from degradation by binding to PGC-1α and limiting ubiquitination. PPARß also induces an increase in nuclear respiratory factor 1 (NRF-1) expression, resulting in increases in mitochondrial respiratory chain proteins and MEF2A, for which NRF-1 is a transcription factor. There was also an increase in AMP kinase phosphorylation mediated by an NRF-1-induced increase in CAM kinase kinase-ß (CaMKKß). Knockdown of PPARß resulted in large decreases in the levels of PGC-1α and mitochondrial proteins and a marked attenuation of the exercise-induced increase in mitochondrial biogenesis. In conclusion, PPARß induces an increase in PGC-1α protein, and PPARß is a transcription factor for NRF-1. Thus, PPARß plays essential roles in the maintenance and adaptive increase in mitochondrial enzymes in skeletal muscle by exercise.


Subject(s)
Mitochondria, Muscle/metabolism , PPAR-beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cell Line , Enzyme Activation , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mitochondria, Muscle/genetics , Nuclear Respiratory Factor 1/genetics , PPAR-beta/genetics , Physical Conditioning, Animal , Proteolysis , Rats, Wistar , Transcriptional Activation , Ubiquitination , Up-Regulation
4.
J Physiol ; 593(3): 635-43, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25416622

ABSTRACT

KEY POINTS: Long-term endurance exercise training results in a reduction in the rates of muscle glycogen depletion and lactic acid accumulation during submaximal exercise; this adaptation is mediated by an increase in muscle mitochondria. There is evidence suggesting that short-term training induces adaptations that downregulate glycogenolysis before there is an increase in functional mitochondria. We discovered that a single long bout of exercise induces decreases in expression of glycogenolytic and glycolytic enzymes in rat skeletal muscle; this adaptation results in slower rates of glycogenolysis and lactic acid accumulation in muscle during contractile activity. Two additional days of training amplified the adaptive response, which appears to be mediated by PGC-1α; this adaptation is biologically significant, because glycogen depletion and lactic acid accumulation are major causes of muscle fatigue. ABSTRACT: Endurance exercise training can increase the ability to perform prolonged strenuous exercise. The major adaptation responsible for this increase in endurance is an increase in muscle mitochondria. This adaptation occurs too slowly to provide a survival advantage when there is a sudden change in environment that necessitates prolonged exercise. In the present study, we discovered another, more rapid adaptation, a downregulation of expression of the glycogenolytic and glycolytic enzymes in muscle that mediates a slowing of muscle glycogen depletion and lactic acid accumulation. This adaptation, which appears to be mediated by PGC-1α, occurs in response to a single exercise bout and is further enhanced by two additional daily exercise bouts. It is biologically significant, because glycogen depletion and lactic acid accumulation are two of the major causes of muscle fatigue and exhaustion.


Subject(s)
Down-Regulation , Glycogenolysis , Muscle, Skeletal/metabolism , Physical Exertion , Transcription Factors/metabolism , Animals , Glycogen/metabolism , Lactic Acid/metabolism , Male , Muscle Fatigue , Muscle, Skeletal/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Wistar , Transcription Factors/genetics
5.
PLoS Biol ; 11(7): e1001603, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23874150

ABSTRACT

It has been reported that feeding mice resveratrol activates AMPK and SIRT1 in skeletal muscle leading to deacetylation and activation of PGC-1α, increased mitochondrial biogenesis, and improved running endurance. This study was done to further evaluate the effects of resveratrol, SIRT1, and PGC-1α deacetylation on mitochondrial biogenesis in muscle. Feeding rats or mice a diet containing 4 g resveratrol/kg diet had no effect on mitochondrial protein levels in muscle. High concentrations of resveratrol lowered ATP concentration and activated AMPK in C2C12 myotubes, resulting in an increase in mitochondrial proteins. Knockdown of SIRT1, or suppression of SIRT1 activity with a dominant-negative (DN) SIRT1 construct, increased PGC-1α acetylation, PGC-1α coactivator activity, and mitochondrial proteins in C2C12 cells. Expression of a DN SIRT1 in rat triceps muscle also induced an increase in mitochondrial proteins. Overexpression of SIRT1 decreased PGC-1α acetylation, PGC-1α coactivator activity, and mitochondrial proteins in C2C12 myotubes. Overexpression of SIRT1 also resulted in a decrease in mitochondrial proteins in rat triceps muscle. We conclude that, contrary to some previous reports, the mechanism by which SIRT1 regulates mitochondrial biogenesis is by inhibiting PGC-1α coactivator activity, resulting in a decrease in mitochondria. We also conclude that feeding rodents resveratrol has no effect on mitochondrial biogenesis in muscle.


Subject(s)
Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Sirtuin 1/metabolism , Stilbenes/pharmacology , Transcription Factors/metabolism , Acetylation/drug effects , Animals , Blotting, Western , Cell Line , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Sprague-Dawley , Resveratrol , Sirtuin 1/genetics , Transcription Factors/genetics
6.
Am J Physiol Endocrinol Metab ; 304(8): E844-52, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23443926

ABSTRACT

There are reports that the ß-adrenergic agonist clenbuterol induces a large increase in peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) in skeletal muscle. This has led to the hypothesis that the increases in PGC-1α and mitochondrial biogenesis induced in muscle by endurance exercise are mediated by catecholamines. In the present study, we evaluated this possibility and found that injecting rats with clenbuterol or norepinephrine induced large increases in PGC-1α and mitochondrial proteins in brown adipose tissue but had no effect on PGC-1α expression or mitochondrial biogenesis in skeletal muscle. In brown adipocytes, the increase in PGC-1α expression induced by ß-adrenergic stimulation is mediated by activation of p38 mitogen-activated protein kinase (p38 MAPK), which phosphorylates and activates the cAMP response element binding protein (CREB) family member activating transcription factor 2 (ATF2), which binds to a cyclic AMP response element (CRE) in the PGC-1α promoter and mediates the increase in PGC-1α transcription. Phospho-CREB does not have this effect. Our results show that the reason for the lack of effect of ß-adrenergic stimulation on PGC-1α expression in muscle is that catecholamines do not activate p38 or increase ATF2 phosphorylation in muscle.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Muscle, Skeletal/physiology , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factor 2/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/enzymology , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Clenbuterol/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Gene Expression/drug effects , Gene Expression/physiology , Hypoglycemic Agents/pharmacology , Male , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/enzymology , Norepinephrine/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation/drug effects , Phosphorylation/physiology , Physical Endurance/physiology , Propranolol/pharmacology , RNA-Binding Proteins/genetics , Rats , Rats, Wistar , Ribonucleotides/pharmacology , Transcription Factors/genetics
7.
Metabolism ; 61(11): 1615-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22571876

ABSTRACT

OBJECTIVE: In a previous study, it was found that a ginseng berry extract with a high content of the ginsenoside Re normalized blood glucose in ob/ob mice. The objective of this study was to evaluate the effect of the ginsenoside Re on insulin resistance of glucose transport in muscles of rats made insulin resistant with a high-fat diet. MATERIAL/METHOD: Rats were fed either rat chow or a high-fat diet for 5 weeks. The rats were then euthanized, and insulin stimulated glucose transport activity was measured in epitrochlearis and soleus muscle strips in vitro. RESULTS: Treatment of muscles with Re alone had no effect on glucose transport. The high-fat diet resulted in ~50% decreases in insulin responsiveness of GLUT4 translocation to the cell surface and glucose transport in epitrochlearis and soleus muscles. Treatment of muscles with Re in vitro for 90 min completely reversed the high-fat diet-induced insulin resistance of glucose transport and GLUT4 translocation. This effect of Re is specific for insulin stimulated glucose transport, as Re treatment did not reverse the high-fat diet-induced resistance of skeletal muscle glucose transport to stimulation by contractions or hypoxia. CONCLUSIONS: Our results show that the ginsenoside Re induces a remarkably rapid reversal of high-fat diet-induced insulin resistance of muscle glucose transport by reversing the impairment of insulin-stimulated GLUT4 translocation to the cell surface.


Subject(s)
Ginsenosides/pharmacology , Insulin Resistance , Muscle, Skeletal/physiopathology , Adenylate Kinase/metabolism , Animals , Electric Stimulation , Glucose/metabolism , In Vitro Techniques , Insulin/metabolism , Male , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Signal Transduction
8.
Genes Dev ; 25(24): 2619-30, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22135324

ABSTRACT

To identify new gene regulatory pathways controlling skeletal muscle energy metabolism, comparative studies were conducted on muscle-specific transgenic mouse lines expressing the nuclear receptors peroxisome proliferator-activated receptor α (PPARα; muscle creatine kinase [MCK]-PPARα) or PPARß/δ (MCK-PPARß/δ). MCK-PPARß/δ mice are known to have enhanced exercise performance, whereas MCK-PPARα mice perform at low levels. Transcriptional profiling revealed that the lactate dehydrogenase b (Ldhb)/Ldha gene expression ratio is increased in MCK-PPARß/δ muscle, an isoenzyme shift that diverts pyruvate into the mitochondrion for the final steps of glucose oxidation. PPARß/δ gain- and loss-of-function studies in skeletal myotubes demonstrated that PPARß/δ, but not PPARα, interacts with the exercise-inducible kinase AMP-activated protein kinase (AMPK) to synergistically activate Ldhb gene transcription by cooperating with myocyte enhancer factor 2A (MEF2A) in a PPARß/δ ligand-independent manner. MCK-PPARß/δ muscle was shown to have high glycogen stores, increased levels of GLUT4, and augmented capacity for mitochondrial pyruvate oxidation, suggesting a broad reprogramming of glucose utilization pathways. Lastly, exercise studies demonstrated that MCK-PPARß/δ mice persistently oxidized glucose compared with nontransgenic controls, while exhibiting supranormal performance. These results identify a transcriptional regulatory mechanism that increases capacity for muscle glucose utilization in a pattern that resembles the effects of exercise training.


Subject(s)
Glucose/metabolism , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , PPAR delta/metabolism , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cells, Cultured , Female , Lactate Dehydrogenases/genetics , Lactate Dehydrogenases/metabolism , Male , Mice , Muscle, Skeletal/enzymology , Oxidation-Reduction , PPAR alpha/metabolism , Physical Conditioning, Animal , Transcriptional Activation
9.
Am J Physiol Endocrinol Metab ; 301(5): E779-84, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21750271

ABSTRACT

It has been reported that supplementation with the antioxidant vitamins C and E prevents the adaptive increases in mitochondrial biogenesis and GLUT4 expression induced by endurance exercise. We reevaluated the effects of these antioxidants on the adaptive responses of rat skeletal muscle to swimming in a short-term study consisting of 9 days of vitamins C and E with exercise during the last 3 days and a longer-term study consisting of 8 wk of antioxidant vitamins with exercise during the last 3 wk. The rats in the antioxidant groups were given 750 mg·kg body wt(-1)·day(-1) vitamin C and 150 mg·kg body wt(-1)·day(-1) vitamin E. In rats euthanized immediately after exercise, plasma TBARs were elevated in the control rats but not in the antioxidant-supplemented rats, providing evidence for an antioxidant effect. In rats euthanized 18 h after exercise there were large increases in insulin responsiveness of glucose transport in epitrochlearis muscles mediated by an approximately twofold increase in GLUT4 expression in both the short- and long-term treatment groups. The protein levels of a number of mitochondrial marker enzymes were also increased about twofold. Superoxide dismutases (SOD) 1 and 2 were increased about twofold in triceps muscle after 3 days of exercise, but only SOD2 was increased after 3 wk of exercise. There were no differences in the magnitudes of any of these adaptive responses between the control and antioxidant groups. These results show that very large doses of antioxidant vitamins do not prevent the exercise-induced adaptive responses of muscle mitochondria, GLUT4, and insulin action to exercise and have no effect on the level of these proteins in sedentary rats.


Subject(s)
Adaptation, Physiological/drug effects , Antioxidants/pharmacology , Cytoprotection/drug effects , Oxidative Stress/drug effects , Physical Conditioning, Animal/physiology , Adaptation, Physiological/physiology , Animals , Ascorbic Acid/pharmacology , Cytoprotection/physiology , Dietary Supplements , Male , Oxidative Stress/physiology , Physical Endurance/drug effects , Rats , Rats, Wistar , Swimming/physiology , Vitamin E/pharmacology
10.
PLoS One ; 6(5): e19739, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21589859

ABSTRACT

BACKGROUND: It has been proposed that muscle insulin resistance in type 2 diabetes is due to a selective decrease in the components of the mitochondrial electron transport chain and results from accumulation of toxic products of incomplete fat oxidation. The purpose of the present study was to test this hypothesis. METHODOLOGY/PRINCIPAL FINDINGS: Rats were made severely iron deficient, by means of an iron-deficient diet. Iron deficiency results in decreases of the iron containing mitochondrial respiratory chain proteins without affecting the enzymes of the fatty acid oxidation pathway. Insulin resistance was induced by feeding iron-deficient and control rats a high fat diet. Skeletal muscle insulin resistance was evaluated by measuring glucose transport activity in soleus muscle strips. Mitochondrial proteins were measured by Western blot. Iron deficiency resulted in a decrease in expression of iron containing proteins of the mitochondrial respiratory chain in muscle. Citrate synthase, a non-iron containing citrate cycle enzyme, and long chain acyl-CoA dehydrogenase (LCAD), used as a marker for the fatty acid oxidation pathway, were unaffected by the iron deficiency. Oleate oxidation by muscle homogenates was increased by high fat feeding and decreased by iron deficiency despite high fat feeding. The high fat diet caused severe insulin resistance of muscle glucose transport. Iron deficiency completely protected against the high fat diet-induced muscle insulin resistance. CONCLUSIONS/SIGNIFICANCE: The results of the study argue against the hypothesis that a deficiency of the electron transport chain (ETC), and imbalance between the ETC and ß-oxidation pathways, causes muscle insulin resistance.


Subject(s)
Electron Transport , Insulin Resistance , Mitochondria/metabolism , Animals , Body Weight , Cyclic AMP-Dependent Protein Kinases/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Muscles/metabolism , Oxidation-Reduction , Phosphorylation , Rats , Triglycerides/metabolism
11.
Aging (Albany NY) ; 3(5): 533-42, 2011 May.
Article in English | MEDLINE | ID: mdl-21566261

ABSTRACT

Plasma dehydroepiandrosterone (DHEA) decreases ~80% between ages 25 and 75 yr. In a preliminary study, we found that 6 mo of DHEA replacement improved insulin action in elderly individuals. The purpose of the present larger, randomized double-blind study was to determine whether a longer period of DHEA replacement improves glucose tolerance. Fifty-seven men and 68 women aged 65 to 75 yr were randomly assigned to 50 mg DHEA or placebo once daily. Year one was a randomized, double blind trial. Year 2 was an open label continuation. DHEA replacement improved glucose tolerance in participants who had abnormal GT initially, reduced plasma triglycerides, and the inflammatory cytokines IL6 and TNFα.


Subject(s)
Aging/physiology , Cytokines/immunology , Dehydroepiandrosterone/administration & dosage , Inflammation/immunology , Insulin Resistance/physiology , Aged , Animals , Blood Chemical Analysis , Dehydroepiandrosterone/blood , Energy Intake , Exercise , Female , Glucose Tolerance Test , Humans , Male , Placebos , Surveys and Questionnaires
12.
FASEB J ; 25(2): 785-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21048043

ABSTRACT

It has been reported that 30% calorie restriction (CR) for 3 mo results in large increases in mitochondrial biogenesis in heart, brain, liver, and adipose tissue, with concomitant increases in respiration and ATP synthesis. We found these results surprising, and performed this study to determine whether 30% CR does induce an increase in mitochondria in heart, brain, liver, adipose tissue, and/or skeletal muscle. To this end, we measured the levels of a range of mitochondrial proteins, and mRNAs. With the exception of long-chain acyl-CoA dehydrogenase protein level, which was increased ∼60% in adipose tissue, none of the mitochondrial proteins or mRNAs that we measured were increased in rats subjected to 30% CR for 14 wk. There was also no increase in citrate synthase activity. Because it is not possible to have an increase in mitochondria without any increase in key mitochondrial proteins, we conclude that 30% CR does not induce an increase in mitochondria in heart, brain, liver, adipose tissue, or skeletal muscle in laboratory rodents.


Subject(s)
Caloric Restriction , Gene Expression Regulation/physiology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Adipose Tissue/metabolism , Animals , Brain/metabolism , Liver/metabolism , Male , Mitochondria/classification , Mitochondrial Proteins/genetics , Muscle, Skeletal/metabolism , Myocardium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
13.
Am J Physiol Endocrinol Metab ; 297(1): E236-41, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19435855

ABSTRACT

Elevated plasma free fatty acids (FFA) cause insulin resistance and are thought to play a key role in mediating insulin resistance in patients with the metabolic syndrome (MTS) and type 2 diabetes mellitus (DM). Two experimental models used to study the mechanisms responsible for insulin resistance in patients are high-fat diet-fed rodents and administration of triglycerides and heparin to raise plasma FFA. As evidence that insulin resistance in high-fat diet-fed rats is due to high FFA, it has been reported that the insulin resistance is rapidly reversed by an overnight fast, a high-glucose meal, and an exercise bout. If true, these findings would invalidate the high-fat diet-fed rodent as a model for MTS or type 2 DM, because insulin resistance is not rapidly reversed by these treatments in patients. The purpose of this study was to determine whether diet-induced insulin resistance is, in fact, rapidly reversible. Incubation of muscles in vitro rapidly reversed insulin resistance induced by administration of triglycerides and heparin, but not by a high-fat diet. An overnight fast and a high-glucose meal were followed by a large increase in insulin-stimulated muscle glucose transport. However, these are adaptive responses, rather than reversals of insulin resistance, because they also occurred in muscles of insulin-sensitive, chow-fed control rats. Our results show that insulin resistance induced by high FFA, i.e., Randle glucose-fatty acid cycle, is transient. In contrast, the insulin resistance induced by a high-fat diet does not reverse rapidly.


Subject(s)
Dietary Fats/pharmacology , Insulin Resistance , Muscle, Skeletal/drug effects , Animals , Biological Transport , Blood Glucose/metabolism , Deoxyglucose/pharmacokinetics , Deoxyglucose/pharmacology , Diet , Fasting/metabolism , Fasting/physiology , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Male , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Time Factors
14.
Proc Natl Acad Sci U S A ; 105(22): 7815-20, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18509063

ABSTRACT

It has been hypothesized that insulin resistance is mediated by a deficiency of mitochondria in skeletal muscle. In keeping with this hypothesis, high-fat diets that cause insulin resistance have been reported to result in a decrease in muscle mitochondria. In contrast, we found that feeding rats high-fat diets that cause muscle insulin resistance results in a concomitant gradual increase in muscle mitochondria. This adaptation appears to be mediated by activation of peroxisome proliferator-activated receptor (PPAR)delta by fatty acids, which results in a gradual, posttranscriptionally regulated increase in PPAR gamma coactivator 1alpha (PGC-1alpha) protein expression. Similarly, overexpression of PPARdelta results in a large increase in PGC-1alpha protein in the absence of any increase in PGC-1alpha mRNA. We interpret our findings as evidence that raising free fatty acids results in an increase in mitochondria by activating PPARdelta, which mediates a posttranscriptional increase in PGC-1alpha. Our findings argue against the concept that insulin resistance is mediated by a deficiency of muscle mitochondria.


Subject(s)
Dietary Fats/administration & dosage , Fatty Acids, Nonesterified/metabolism , Insulin Resistance , Mitochondria, Muscle/drug effects , Muscle, Skeletal/metabolism , Abdominal Fat/drug effects , Animals , Body Weight , Diet/adverse effects , Fatty Acids, Nonesterified/blood , Male , Mitochondria, Muscle/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/ultrastructure , Oxidation-Reduction , PPAR delta/agonists , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Wistar , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors
15.
J Biol Chem ; 282(50): 36642-51, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17932032

ABSTRACT

The transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) has been identified as an inducible regulator of mitochondrial function. Skeletal muscle PGC-1alpha expression is induced post-exercise. Therefore, we sought to determine its role in the regulation of muscle fuel metabolism. Studies were performed using conditional, muscle-specific, PGC-1alpha gain-of-function and constitutive, generalized, loss-of-function mice. Forced expression of PGC-1alpha increased muscle glucose uptake concomitant with augmentation of glycogen stores, a metabolic response similar to post-exercise recovery. Induction of muscle PGC-1alpha expression prevented muscle glycogen depletion during exercise. Conversely, PGC-1alpha-deficient animals exhibited reduced rates of muscle glycogen repletion post-exercise. PGC-1alpha was shown to increase muscle glycogen stores via several mechanisms including stimulation of glucose import, suppression of glycolytic flux, and by down-regulation of the expression of glycogen phosphorylase and its activating kinase, phosphorylase kinase alpha. These findings identify PGC-1alpha as a critical regulator of skeletal muscle fuel stores.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Mitochondria, Muscle/metabolism , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Trans-Activators/biosynthesis , Animals , Glucose/genetics , Glycogen/genetics , Glycogen Phosphorylase/genetics , Glycogen Phosphorylase/metabolism , Mice , Mice, Transgenic , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylase Kinase/genetics , Phosphorylase Kinase/metabolism , Trans-Activators/genetics , Transcription Factors
16.
Proc Natl Acad Sci U S A ; 104(25): 10709-13, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17548828

ABSTRACT

A number of studies have reported that a high-fat diet induces increases in mitochondrial fatty acid oxidation enzymes in muscle. In contrast, in two recent studies raising plasma free fatty acids (FFA) resulted in a decrease in mitochondria. In this work, we reevaluated the effects of raising FFA on muscle mitochondrial biogenesis and capacity for fat oxidation. Rats were fed a high-fat diet and given daily injections of heparin to raise FFA. This treatment induced an increase in mitochondrial biogenesis in muscle, as evidenced by increases in mitochondrial enzymes of the fatty acid oxidation pathway, citrate cycle, and respiratory chain, with an increase in the capacity to oxidize fat, as well as an increase in mitochondrial DNA copy number. Raising FFA also resulted in an increase in binding of peroxisome proliferator-activated receptor (PPAR) delta to the PPAR response element on the carnitine palmitoyltransferase 1 promoter. We interpret our results as evidence that raising FFA induces an increase in mitochondrial biogenesis in muscle by activating PPARdelta.


Subject(s)
Fatty Acids/blood , Mitochondria, Muscle/physiology , Muscle, Skeletal/physiology , Organelle Biogenesis , Animals , DNA, Mitochondrial/genetics , Gene Dosage , Male , Mitochondria/enzymology , Mitochondria/metabolism , Oxidation-Reduction , PPAR gamma/metabolism , Palmitates/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar
17.
J Biol Chem ; 282(26): 18793-9, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17488713

ABSTRACT

Previous studies have shown that raising cytosolic calcium in myotubes induces increases in peroxisome proliferator-activated receptor gamma coactivator-1alpha expression and mitochondrial biogenesis. This finding suggests that the increases in cytosolic calcium in skeletal muscle during exercise may mediate the exercise-induced increase in mitochondria. The initial aim of this study was to determine whether raising calcium in skeletal muscle induces the same adaptations as in myotubes. We found that treatment of rat epitrochlearis muscles with a concentration of caffeine that raises cytosolic calcium to a concentration too low to cause contraction induces increases in peroxisome proliferator-activated receptor gamma coactivator-1alpha expression and mitochondrial biogenesis. Our second aim was to elucidate the pathway by which calcium induces these adaptations. Raising cytosolic calcium has been shown to activate calcium/calmodulin-dependent protein kinase in muscle. In the present study raising cytosolic calcium resulted in increases in phosphorylation of p38 mitogen-activated protein kinase and activating transcription factor-2, which were blocked by the calcium/calmodulin-dependent protein kinase inhibitor KN93 and by the p38 mitogen-activated protein kinase inhibitor SB202190. The increases in peroxisome proliferator-activated receptor gamma coactivator-1alpha expression and mitochondrial biogenesis were also prevented by inhibiting p38 activation. We interpret these findings as evidence that p38 mitogen-activated protein kinase is downstream of calcium/calmodulin-dependent protein kinase in a signaling pathway by which increases in cytosolic calcium lead to increases in peroxisome proliferator-activated receptor gamma coactivator-1alpha expression and mitochondrial biogenesis in muscle.


Subject(s)
Calcium/metabolism , Heat-Shock Proteins/metabolism , Mitochondria/metabolism , PPAR gamma/metabolism , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Actins/metabolism , Activating Transcription Factor 2/metabolism , Adaptation, Physiological/physiology , Animals , Biomarkers/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Male , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Pyridines/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
18.
J Nucl Med ; 48(6): 955-64, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17536107

ABSTRACT

UNLABELLED: The aim of this study was to investigate whether compartmental modeling of 1-(11)C-glucose PET kinetics can be used for noninvasive measurements of myocardial glucose metabolism beyond its initial extraction. METHODS: 1-(11)C-Glucose and U-(13)C-glucose were injected simultaneously into 22 mongrel dogs under a wide range of metabolic states; this was followed by 1 h of PET data acquisition. Heart tissue samples were analyzed for (13)C-glycogen content (nmol/g). Arterial and coronary sinus blood samples (ART/CS) were analyzed for glucose (mumol/mL), (11)C-glucose, (11)CO(2), and (11)C-total acidic metabolites ((11)C-lactate [LA] + (11)CO(2)) (counts/min/mL) and were used to calculate myocardial fractions of (a) glucose and 1-(11)C-glucose extractions, EF(GLU) and EF((11)C-GLU); (b) (11)C-GLU and (11)C-LA oxidation, OF((11)C-GLU) and OF((11)C-LA); (c) (11)C-glycolsysis, GCF((11)C-GLU); and (d) (11)C-glycogen content, GNF((11)C-GLU). On the basis of these measurements, a compartmental model (M) that accounts for the contribution of exogenous (11)C-LA to myocardial (11)C activity was implemented to measure M-EF(GLU), M-GCF(GLU), M-OF(GLU), M-GNF(GLU), and the fraction of myocardial glucose stored as glycogen M-GNF(GLU)/M-EF(GLU)). RESULTS: ART/CS data showed the following: (a) A strong correlation was found between EF((11)C-GLU) and EF(GLU) (r = 0.92, P < 0.0001; slope = 0.95, P = not significantly different from 1). (b) In interventions with high glucose extraction and oxidation, the contribution of OF((11)C-GLU) to total oxidation was higher than that of OF((11)C-LA) (P < 0.01). In contrast, in interventions in which glucose uptake and oxidation were inhibited, OF((11)C-LA) was higher than OF((11)C-GLU) (P < 0.05). (c) A strong correlation was found between GNF((11)C-GLU)/EF(GLU) and direct measurements of fractional (13)C-glycogen content, (r = 0.96, P < 0.0001). Model-derived PET measurements of M-EF(GLU), M-GCF(GLU), and M-OF(GLU) strongly correlated with EF(GLU) (slope = 0.92, r = 0.95, P < 0.0001), GCF((11)C-GLU) (slope = 0.79, r = 0.97, P < 0.0001), and OF((11)C-GLU) (slope = 0.70, r = 0.96, P < 0.0001), respectively. M-GNF(GLU)/M-EF(GLU) strongly correlated with fractional (13)C-content (r = 0.92, P < 0.0001). CONCLUSION: Under nonischemic conditions, it is feasible to measure myocardial glucose metabolism noninvasively beyond its initial extraction with PET using 1-(11)C-glucose and a compartmental modeling approach that takes into account uptake and oxidation of secondarily labeled exogenous (11)C-lactate.


Subject(s)
Glucose/pharmacokinetics , Models, Biological , Myocardium/metabolism , Radiopharmaceuticals/pharmacokinetics , Animals , Blood Glucose/analysis , Carbon Radioisotopes , Dogs , Glycogen/metabolism , Insulin/blood , Positron-Emission Tomography/methods
19.
Am J Physiol Endocrinol Metab ; 292(6): E1842-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17327367

ABSTRACT

Exercise induces an increase in glucose transport in muscle. As the acute increase in glucose transport reverses, it is replaced by an increase in insulin sensitivity. Interleukin-6 (IL-6) increases with exercise and has been reported to activate AMP-activated protein kinase (AMPK). Based on this information, we hypothesized that IL-6 would result in an increase in muscle insulin sensitivity. Rat epitrochlearis and soleus muscles were incubated with 120 ng/ml IL-6. Exposure to IL-6 induced a modest acute increase in glucose transport and was followed 3.5 h later by an increase in insulin sensitivity in epitrochlearis but not soleus muscles. IL-6 also brought about an increase in AMPK phosphorylation in epitrochlearis muscles. We conclude that exposure of fast-twitch muscle to 120 ng/ml IL-6 increases insulin sensitivity by activating AMPK. However, exposure of epitrochlearis muscles to 10 ng/ml IL-6, a concentration >100-fold higher than that attained in plasma during exercise, had no effect on glucose transport or insulin sensitivity. These findings provide evidence that the increases in glucose transport and insulin sensitivity induced by IL-6 are pharmacological rather than physiological effects. We interpret our results as evidence that the increase in IL-6 during exercise does not play a role in the exercise-induced increases in muscle glucose uptake and insulin sensitivity.


Subject(s)
Enzyme Activation/drug effects , Insulin/physiology , Interleukin-6/administration & dosage , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , AMP-Activated Protein Kinases , Animals , Biological Transport/drug effects , Dose-Response Relationship, Drug , Forelimb , Glucose/metabolism , Hindlimb , Interleukin-6/pharmacology , Male , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Time Factors
20.
J Biol Chem ; 282(1): 194-9, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17099248

ABSTRACT

Exercise results in rapid increases in expression of the transcription coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) and in mitochondrial biogenesis in skeletal muscle. PGC-1alpha regulates and coordinates mitochondrial biogenesis, and overexpression of PGC-1alpha in muscle cells results in increases in mitochondrial content. In this context, it has been proposed that the increase in PGC-1alpha protein expression mediates the exercise-induced increase in mitochondrial biogenesis. However, we found that mitochondrial proteins with a short half-life increase as rapidly as, or more rapidly than, PGC-1alpha protein. This finding led us to hypothesize that activation, rather than increased expression, of PGC-1alpha mediates the initial phase of the exercise-induced increase in mitochondria. In this study, we found that most of the PGC-1alpha in resting skeletal muscle is in the cytosol. Exercise resulted in activation of p38 MAPK and movement of PGC-1alpha into the nucleus. In support of our hypothesis, binding of the transcription factor nuclear respiratory factor 1 (NRF-1) to the cytochrome c promoter and NRF-2 to the cytochrome oxidase subunit 4 promoter increased in response to exercise prior to an increase in PGC-1alpha protein. Furthermore, exercise-induced increases in the mRNAs of cytochrome c, delta-aminolevulinate synthase, and citrate synthase also occurred before an increase in PGC-1 protein. Thus, it appears that activation of PGC-1alpha may mediate the initial phase of the exercise-induced adaptive increase in muscle mitochondria, whereas the subsequent increase in PGC-1alpha protein sustains and enhances the increase in mitochondrial biogenesis.


Subject(s)
Muscles/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Nucleus/metabolism , Chromatin/metabolism , Cytosol/metabolism , DNA Primers/chemistry , Male , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL