Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Nutrients ; 12(3)2020 Feb 28.
Article in English | MEDLINE | ID: mdl-32121211

ABSTRACT

This study aimed to investigate the long-term effects of training intervention and resting on protein expression and stability of peroxisome proliferator-activated receptor ß/δ (PPARß), peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC1α), glucose transporter type 4 (GLUT4), and mitochondrial proteins, and determine whether glucose homeostasis can be regulated through stable expression of these proteins after training. Rats swam daily for 3, 6, 9, 14, or 28 days, and then allowed to rest for 5 days post-training. Protein and mRNA levels were measured in the skeletal muscles of these rats. PPARß was overexpressed and knocked down in myotubes in the skeletal muscle to investigate the effects of swimming training on various signaling cascades of PGC-1α transcription, insulin signaling, and glucose uptake. Exercise training (Ext) upregulated PPARß, PGC-1α, GLUT4, and mitochondrial enzymes, including NADH-ubiquinone oxidoreductase (NUO), cytochrome c oxidase subunit I (COX1), citrate synthase (CS), and cytochrome c (Cyto C) in a time-dependent manner and promoted the protein stability of PPARß, PGC-1α, GLUT4, NUO, CS, and Cyto C, such that they were significantly upregulated 5 days after training cessation. PPARß overexpression increased the PGC-1α protein levels post-translation and improved insulin-induced signaling responsiveness and glucose uptake. The present results indicate that Ext promotes the protein stability of key mitochondria enzymes GLUT4, PGC-1α, and PPARß even after Ext cessation.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Muscle, Skeletal/metabolism , PPAR-beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal , Animals , Glucose Transporter Type 4/metabolism , Half-Life , Male , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , PPAR-beta/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Protein Biosynthesis/drug effects , Protein Stability/drug effects , Rats, Wistar , Transcription, Genetic/drug effects , Up-Regulation/drug effects
2.
Lancet Diabetes Endocrinol ; 7(9): 673-683, 2019 09.
Article in English | MEDLINE | ID: mdl-31303390

ABSTRACT

BACKGROUND: For several cardiometabolic risk factors, values considered within normal range are associated with an increased risk of cardiovascular morbidity and mortality. We aimed to investigate the short-term and long-term effects of calorie restriction with adequate nutrition on these risk factors in healthy, lean, or slightly overweight young and middle-aged individuals. METHODS: CALERIE was a phase 2, multicentre, randomised controlled trial in young and middle-aged (21-50 years), healthy non-obese (BMI 22·0-27·9 kg/m2) men and women done in three clinical centres in the USA. Participants were randomly assigned (2:1) to a 25% calorie restriction diet or an ad libitum control diet. Exploratory cardiometabolic risk factor responses to a prescribed 25% calorie restriction diet for 2 years were evaluated (systolic, diastolic, and mean blood pressure; plasma lipids; high-sensitivity C-reactive protein; metabolic syndrome score; and glucose homoeostasis measures of fasting insulin, glucose, insulin resistance, and 2-h glucose, area-under-the curve for glucose, and insulin from an oral glucose tolerance test) analysed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT00427193. FINDINGS: From May 8, 2007, to Feb 26, 2010, of 238 participants that were assessed, 218 were randomly assigned to and started a 25% calorie restriction diet (n=143, 66%) or an ad libitum control diet (n=75, 34%). Individuals in the calorie restriction group achieved a mean reduction in calorie intake of 11·9% (SE 0·7; from 2467 kcal to 2170 kcal) versus 0·8% (1·0) in the control group, and a sustained mean weight reduction of 7·5 kg (SE 0·4) versus an increase of 0·1 kg (0·5) in the control group, of which 71% (mean change in fat mass 5·3 kg [SE 0·3] divided by mean change in weight 7·5 kg [0·4]) was fat mass loss. Calorie restriction caused a persistent and significant reduction from baseline to 2 years of all measured conventional cardiometabolic risk factors, including change scores for LDL-cholesterol (p<0·0001), total cholesterol to HDL-cholesterol ratio (p<0·0001), and systolic (p<0·0011) and diastolic (p<0·0001) blood pressure. In addition, calorie restriction resulted in a significant improvement at 2 years in C-reactive protein (p=0·012), insulin sensitivity index (p<0·0001), and metabolic syndrome score (p<0·0001) relative to control. A sensitivity analysis revealed the responses to be robust after controlling for relative weight loss changes. INTERPRETATION: 2 years of moderate calorie restriction significantly reduced multiple cardiometabolic risk factors in young, non-obese adults. These findings suggest the potential for a substantial advantage for cardiovascular health of practicing moderate calorie restriction in young and middle-aged healthy individuals, and they offer promise for pronounced long-term population health benefits. FUNDING: National Institute on Aging and National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health.


Subject(s)
Caloric Restriction , Cardiovascular Diseases/diet therapy , Metabolic Diseases/diet therapy , Overweight/prevention & control , Adult , Blood Pressure , Cardiovascular Diseases/blood , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/prevention & control , Cholesterol, LDL/blood , Female , Glucose Tolerance Test , Humans , Male , Metabolic Diseases/blood , Metabolic Diseases/physiopathology , Metabolic Diseases/prevention & control , Middle Aged , Overweight/blood , Overweight/complications , Overweight/physiopathology , Prospective Studies , Time Factors , Treatment Outcome , Weight Loss , Young Adult
3.
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
4.
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
5.
Aging (Albany NY) ; 8(7): 1416-31, 2016 07.
Article in English | MEDLINE | ID: mdl-27410480

ABSTRACT

Calorie restriction (CR) inhibits inflammation and slows aging in many animal species, but in rodents housed in pathogen-free facilities, CR impairs immunity against certain pathogens. However, little is known about the effects of long-term moderate CR on immune function in humans. In this multi-center, randomized clinical trial to determine CR's effect on inflammation and cell-mediated immunity, 218 healthy non-obese adults (20-50 y), were assigned 25% CR (n=143) or an ad-libitum (AL) diet (n=75), and outcomes tested at baseline, 12, and 24 months of CR. CR induced a 10.4% weight loss over the 2-y period. Relative to AL group, CR reduced circulating inflammatory markers, including total WBC and lymphocyte counts, ICAM-1 and leptin. Serum CRP and TNF-α concentrations were about 40% and 50% lower in CR group, respectively. CR had no effect on the delayed-type hypersensitivity skin response or antibody response to vaccines, nor did it cause difference in clinically significant infections. In conclusion, long-term moderate CR without malnutrition induces a significant and persistent inhibition of inflammation without impairing key in vivo indicators of cell-mediated immunity. Given the established role of these pro-inflammatory molecules in the pathogenesis of multiple chronic diseases, these CR-induced adaptations suggest a shift toward a healthy phenotype.


Subject(s)
Body Composition/physiology , Caloric Restriction , Diet , Inflammation/diet therapy , Adult , Biomarkers/blood , C-Reactive Protein/metabolism , Energy Intake , Female , Humans , Inflammation/blood , Intercellular Adhesion Molecule-1/blood , Leptin/blood , Lymphocyte Count , Male , Middle Aged , Treatment Outcome , Tumor Necrosis Factor-alpha/blood
6.
Cell Rep ; 14(3): 422-428, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26774472

ABSTRACT

Calorie restriction (CR) retards aging, acts as a hormetic intervention, and increases serum corticosterone and HSP70 expression in rodents. However, less is known regarding the effects of CR on these factors in humans. Serum cortisol and molecular chaperones and autophagic proteins were measured in the skeletal muscle of subjects on CR diets for 3-15 years and in control volunteers. Serum cortisol was higher in the CR group than in age-matched sedentary and endurance athlete groups (15.6 ± 4.6 ng/dl versus 12.3 ± 3.9 ng/dl and 11.2 ± 2.7 ng/dl, respectively; p ≤ 0.001). HSP70, Grp78, beclin-1, and LC3 mRNA and/or protein levels were higher in the skeletal muscle of the CR group compared to controls. Our data indicate that CR in humans is associated with sustained rises in serum cortisol, reduced inflammation, and increases in key molecular chaperones and autophagic mediators involved in cellular protein quality control and removal of dysfunctional proteins and organelles.


Subject(s)
Caloric Restriction , Muscle, Skeletal/metabolism , Adult , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Body Mass Index , Cluster Analysis , Endoplasmic Reticulum Chaperone BiP , Exercise , Female , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hydrocortisone/blood , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , RNA, Messenger/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Aging Cell ; 15(1): 22-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26443692

ABSTRACT

Young-onset calorie restriction (CR) in rodents decreases serum IGF-1 concentration and increases serum corticosterone levels, which have been hypothesized to play major roles in mediating its anticancer and anti-aging effects. However, little is known on the effects of CR on the IGF-1 system and cortisol in humans. To test the sustained effects of CR on these key hormonal adaptations, we performed a multicenter randomized trial of a 2-year 25% CR intervention in 218 nonobese (body mass index between 22 and 27.8 kg m(-2) ) young and middle-aged (20-50 years age range) men and women. Average CR during the first 6 months was 19.5 ± 0.8% and 9.1 ± 0.7% over the next 18 months of the study. Weight loss averaged 7.6 ± 0.3 kg over the 2-years period of which 71% was fat mass loss (P < 0.0001). Average CR during the CR caused a significant 21% increase in serum IGFBP-1 and a 42% reduction in IGF-1:IGFBP-1 ratio at 2 years (P < 0.008), but did not change IGF-1 and IGF-1:IGFBP-3 ratio levels. Serum cortisol concentrations were slightly but significantly increased by CR at 1 year only (P = 0.003). Calorie restriction had no effect on serum concentrations of PDGF-AB and TGFß-1. We conclude, on the basis of the present and previous findings, that, in contrast to rodents, humans do not respond to CR with a decrease in serum IGF-1 concentration or with a sustained and biological relevant increase in serum cortisol. However, long-term CR in humans significantly and persistently increases serum IGFBP-1 concentration.


Subject(s)
Aging/physiology , Caloric Restriction , Energy Intake/physiology , Hydrocortisone/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Adult , Blood Glucose/metabolism , Body Mass Index , Female , Humans , Insulin-Like Growth Factor Binding Protein 1/blood , Male , Middle Aged , Sex Characteristics , Time Factors
8.
J Gerontol A Biol Sci Med Sci ; 70(9): 1097-104, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26187233

ABSTRACT

BACKGROUND: Caloric restriction (CR), energy intake reduced below ad libitum (AL) intake, increases life span in many species. The implications for humans can be clarified by randomized controlled trials of CR. METHODS: To determine CR's feasibility, safety, and effects on predictors of longevity, disease risk factors, and quality of life in nonobese humans aged 21-51 years, 218 persons were randomized to a 2-year intervention designed to achieve 25% CR or to AL diet. Outcomes were change from baseline resting metabolic rate adjusted for weight change ("RMR residual") and core temperature (primary); plasma triiodothyronine (T3) and tumor necrosis factor-α (secondary); and exploratory physiological and psychological measures. RESULTS: Body mass index averaged 25.1 (range: 21.9-28.0 kg/m(2)). Eighty-two percent of CR and 95% of AL participants completed the protocol. The CR group achieved 11.7±0.7 %CR (mean ± standard error) and maintained 10.4±0.4% weight loss. Weight change in AL was negligible. RMR residual decreased significantly more in CR than AL at 12 months (p = .04) but not 24 months (M24). Core temperature change differed little between groups. T3 decreased more in CR at M12 and M24 (p < .001), while tumor necrosis factor-α decreased significantly more only at M24 (p = .02). CR had larger decreases in cardiometabolic risk factors and in daily energy expenditure adjusted for weight change, without adverse effects on quality of life. CONCLUSIONS: Sustained CR is feasible in nonobese humans. The effects of the achieved CR on correlates of human survival and disease risk factors suggest potential benefits for aging-related outcomes that could be elucidated by further human studies.


Subject(s)
Caloric Restriction , Longevity , Adult , Basal Metabolism , Blood Glucose/analysis , Blood Pressure , Body Temperature , C-Reactive Protein/analysis , Energy Intake , Feasibility Studies , Female , Humans , Lipids/blood , Male , Middle Aged , Time Factors , Triiodothyronine/blood , Tumor Necrosis Factor-alpha/blood , Weight Loss , Young Adult
9.
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
10.
Med Sci Sports Exerc ; 46(6): 1098-103, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24576859

ABSTRACT

INTRODUCTION: After food ingestion, the incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), are secreted by the intestines into circulation where they act on the pancreas to promote insulin secretion. We evaluated the hypothesis that low postprandial plasma insulin levels in lean exercise-trained individuals are associated with low concentrations of incretin hormones. METHODS: A cross-sectional study was performed to compare postprandial incretin hormone levels in lean endurance exercise-trained individuals (EX; n = 14, ≥40 yr) and age- and sex-matched, nonobese, sedentary control subjects (CON, n = 14). The main outcome measures were GLP-1, GIP, insulin, and glucose incremental areas under the curve (AUC) as measured in plasma samples collected during a 2-h,75-g oral glucose tolerance test (OGTT). RESULTS: The EX group had lower body fat percentage (14.6% ± 1.1% vs 23.3% ± 1.7%, P = 0.0002) and higher maximal oxygen uptake (53 ± 2 vs 34 ± 2, P < 0.0001) than CON. Glucose AUC did not differ between groups (P = 0.20). Insulin AUC was lower in EX (2.5 ± 0.5 vs 4.2 ± 1.2 µU·mL·1000 min, P = 0.02). No differences were observed between groups (EX and CON, respectively) for GLP-1 AUC (3.5 ± 0.7 vs 4.1 ± 1.1 pmol·min·100 L, P = 0.61) or GIP AUC (19.2 ± 1.4 vs 18.0 ± 1.4 pg·min·1000 mL; P = 0.56). In CON, insulin AUC was correlated with AUC for GLP-1 (r = 0.53, P = 0.05) and GIP (r = 0.71, P = 0.004), but no such correlations were observed in EX (both P ≥ 0.67). CONCLUSIONS: Low postprandial insulin levels in lean exercise-trained individuals are not attributable to lower incretin hormone concentrations. However, exercise may decrease the dependency of postprandial insulin levels on incretin hormones.


Subject(s)
Incretins/blood , Insulin/blood , Physical Education and Training , Physical Endurance/physiology , Postprandial Period , Anthropometry , Area Under Curve , Blood Glucose/metabolism , Cross-Sectional Studies , Energy Intake , Female , Gastric Inhibitory Polypeptide/blood , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test , Humans , Male , Middle Aged , Oxygen Consumption
11.
Aging (Albany NY) ; 6(2): 149-57, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24659610

ABSTRACT

Dietary supplements are widely used for health purposes. However, little is known about the metabolic and cardiovascular effects of combinations of popular over-the-counter supplements, each of which has been shown to have anti-oxidant, anti-inflammatory and pro-longevity properties in cell culture or animal studies. This study was a 6-month randomized, single-blind controlled trial, in which 56 non-obese (BMI 21.0-29.9 kg/m(2)) men and women, aged 38 to 55 yr, were assigned to a dietary supplement (SUP) group or control (CON) group, with a 6-month follow-up. The SUP group took 10 dietary supplements each day (100 mg of resveratrol, a complex of 800 mg each of green, black, and white tea extract, 250 mg of pomegranate extract, 650 mg of quercetin, 500 mg of acetyl-l-carnitine, 600 mg of lipoic acid, 900 mg of curcumin, 1 g of sesamin, 1.7 g of cinnamon bark extract, and 1.0 g fish oil). Both the SUP and CON groups took a daily multivitamin/mineral supplement. The main outcome measures were arterial stiffness, endothelial function, biomarkers of inflammation and oxidative stress, and cardiometabolic risk factors. Twenty-four weeks of daily supplementation with 10 dietary supplements did not affect arterial stiffness or endothelial function in nonobese individuals. These compounds also did not alter body fat measured by DEXA, blood pressure, plasma lipids, glucose, insulin, IGF-1, and markers of inflammation and oxidative stress. In summary, supplementation with a combination of popular dietary supplements has no cardiovascular or metabolic effects in non-obese relatively healthy individuals.


Subject(s)
Biomarkers , Cardiovascular Diseases/prevention & control , Dietary Supplements , Metabolic Diseases/prevention & control , Adult , Female , Humans , Male , Middle Aged
12.
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
13.
Diabetes ; 62(4): 1036-40, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23520283

ABSTRACT

Based on evidence that patients with type 2 diabetes (T2DM), obese insulin-resistant individuals, and lean insulin-resistant offspring of parents with T2DM have ~30% less mitochondria in their muscles than lean control subjects, it appears to be widely accepted that mitochondrial "deficiency" is responsible for insulin resistance. The proposed mechanism for this effect is an impaired ability to oxidize fat, resulting in lipid accumulation in muscle. The purpose of this counterpoint article is to review the evidence against the mitochondrial deficiency concept. This evidence includes the findings that 1) development of insulin resistance in laboratory rodents fed high-fat diets occurs despite a concomitant increase in muscle mitochondria; 2) mitochondrial deficiency severe enough to impair fat oxidation in resting muscle causes an increase, not a decrease, in insulin action; and 3) most of the studies comparing fat oxidation in insulin-sensitive and insulin-resistant individuals have shown that fat oxidation is higher in T2DM patients and obese insulin-resistant individuals than in insulin-sensitive control subjects. In conclusion, it seems clear, based on this evidence, that the 30% reduction in muscle content of mitochondria in patients with T2DM is not responsible for insulin resistance.


Subject(s)
Insulin Resistance/physiology , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Humans , Obesity/metabolism , Oxidation-Reduction
14.
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
15.
Diabetes Metab Res Rev ; 29(1): 98-100, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23300148

ABSTRACT

The people afflicted with obesity and its consequences need a menu of low-cost programmes to mitigate the over-nutrition and under-activity that underlie their condition. This editorial commentary points out the multiple merits (and shortcomings) of the Nordic walking - with ski poles - presented in an accompanying article. The Nordic exercise programme is then examined more broadly in the context of the general principles and limitations of exercise programmes - with and without calorie control - as a guide to future innovations.


Subject(s)
Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/therapy , Exercise Therapy/methods , Glucose Intolerance/therapy , Overweight/therapy , Walking/physiology , Female , Humans , Male
16.
Aging Cell ; 11(5): 876-84, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22712469

ABSTRACT

Serum dehydroepiandrosterone (DHEA) concentrations decrease approximately 80% between ages 25 and 75 year. Aging also results in an increase in arterial stiffness, which is an independent predictor of cardiovascular disease (CVD) risk and mortality. Therefore, it is conceivable that DHEA replacement in older adults could reduce arterial stiffness. We sought to determine whether DHEA replacement therapy in older adults reduces carotid augmentation index (AI) and carotid-femoral pulse wave velocity (PWV) as indices of arterial stiffness. A randomized, double-blind trial was conducted to study the effects of 50 mg day(-1) DHEA replacement on AI (n = 92) and PWV (n = 51) in women and men aged 65-75 year. Inflammatory cytokines and sex hormones were measured in fasting serum. AI decreased in the DHEA group, but not in the placebo group (difference between groups, -6 ± 2 AI units, P = 0.002). Pulse wave velocity also decreased (difference between groups, -3.5 ± 1.0 m s(-1), P = 0.001); however, after adjusting for baseline values, the between-group comparison became nonsignificant (P = 0.20). The reductions in AI and PWV were accompanied by decreases in inflammatory cytokines (tumor necrosis factor α and IL-6, P < 0.05) and correlated with increases in serum DHEAS (r = -0.31 and -0.37, respectively, P < 0.05). The reductions in AI also correlated with free testosterone index (r = -0.23, P = 0.03). In conclusion, DHEA replacement in elderly men and women improves indices of arterial stiffness. Arterial stiffness increases with age and is an independent risk factor for CVD. Therefore, the improvements observed in this study suggest that DHEA replacement might partly reverse arterial aging and reduce CVD risk.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Vascular Stiffness/drug effects , Age Factors , Aged , Blood Flow Velocity/drug effects , Dehydroepiandrosterone/blood , Double-Blind Method , Female , Humans , Male , Risk Factors
17.
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
18.
Aging Cell ; 11(4): 644-50, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22510429

ABSTRACT

Caloric restriction (CR) retards aging in laboratory rodents. No information is available on the effects of long-term CR on physiologic markers of aging and longevity in humans. Heart rate variability (HRV) is a marker for cardiac autonomic functioning. The progressive decline in HRV with aging and the association of higher HRV with better health outcomes are well established. Heart rate variability assessment is a reliable tool by which the effects of CR on autonomic function can be assessed. Time- and frequency-domain analyses compared 24-h HRV in 22 CR individuals aged 35-82 years and 20 age-matched controls eating Western diets (WD). The CR group was significantly leaner than the WD group. Heart rate was significantly lower, and virtually, all HRV values were significantly higher in the CR group than in the WD group (P < 0.002). Heart rate variability in the CR individuals was comparable with published norms for healthy individuals 20 years younger. In addition, when differences in heart rate (HR) and HRV between CR and WD were compared with previously published changes in HRV induced in healthy adults given atenolol, percent differences in each measure were generally similar in direction and magnitude and suggested declines in sympathetic and increases in parasympathetic modulation of HR and increased circadian variability associated with CR. These findings provide evidence that CR has direct systemic effects that counter the expected age-associated changes in autonomic function so that HRV indexes in CR individuals are similar to those of individuals 20 years younger eating WDs.


Subject(s)
Aging/physiology , Autonomic Nervous System/physiology , Caloric Restriction , Adult , Aged , Aged, 80 and over , Analysis of Variance , Case-Control Studies , Female , Heart Rate/physiology , Humans , Longevity/physiology , Male , Middle Aged , Risk Factors
19.
Biophys J ; 102(3): 399-406, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22325261

ABSTRACT

Inference of the insulin secretion rate (ISR) from C-peptide measurements as a quantification of pancreatic ß-cell function is clinically important in diseases related to reduced insulin sensitivity and insulin action. ISR derived from C-peptide concentration is an example of nonparametric Bayesian model selection where a proposed ISR time-course is considered to be a "model". An inferred value of inaccessible continuous variables from discrete observable data is often problematic in biology and medicine, because it is a priori unclear how robust the inference is to the deletion of data points, and a closely related question, how much smoothness or continuity the data actually support. Predictions weighted by the posterior distribution can be cast as functional integrals as used in statistical field theory. Functional integrals are generally difficult to evaluate, especially for nonanalytic constraints such as positivity of the estimated parameters. We propose a computationally tractable method that uses the exact solution of an associated likelihood function as a prior probability distribution for a Markov-chain Monte Carlo evaluation of the posterior for the full model. As a concrete application of our method, we calculate the ISR from actual clinical C-peptide measurements in human subjects with varying degrees of insulin sensitivity. Our method demonstrates the feasibility of functional integral Bayesian model selection as a practical method for such data-driven inference, allowing the data to determine the smoothing timescale and the width of the prior probability distribution on the space of models. In particular, our model comparison method determines the discrete time-step for interpolation of the unobservable continuous variable that is supported by the data. Attempts to go to finer discrete time-steps lead to less likely models.


Subject(s)
C-Peptide/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Models, Biological , Bayes Theorem , Female , Humans , Insulin Secretion , Kinetics , Male , Markov Chains , Monte Carlo Method
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...