Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Mol Sci ; 22(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573178

ABSTRACT

Phenolic compounds have emerged in recent years as an option to face insulin resistance and diabetes. The central aim of this study was: (1) to demonstrate that physiological doses of resveratrol (RSV) or quercetin (Q) can influence glucose metabolism in human myotubes, (2) to establish whether AMP-activated protein kinase (AMPK) and protein kinase B -PKB- (Akt) pathways are involved in this effect. In addition, the effects of these polyphenols on mitochondrial biogenesis and fatty acid oxidation were analysed. Myotubes from healthy donors were cultured for 24 h with either 0.1 µM of RSV or with 10 µM of Q. Glucose metabolism, such as glycogen synthesis, glucose oxidation, and lactate production, were measured with D[U-14C]glucose. ß-oxidation using [1-14C]palmitate as well as the expression of key metabolic genes and proteins by Real Time PCR and Western blot were also assessed. Although RSV and Q increased pgc1α expression, they did not significantly change either glucose oxidation or ß-oxidation. Q increased AMPK, insulin receptor substrate 1 (IRS-1), and AS160 phosphorylation in basal conditions and glycogen synthase kinase 3 (GSK3ß) in insulin-stimulated conditions. RSV tended to increase the phosphorylation rates of AMPK and GSK3ß. Both of the polyphenols increased insulin-stimulated glycogen synthesis and reduced lactate production in human myotubes. Thus, physiological doses of RSV or Q may exhibit anti-diabetic actions in human myotubes.


Subject(s)
Diabetes Mellitus/drug therapy , Glucose/metabolism , Muscle Fibers, Skeletal/drug effects , Quercetin/pharmacology , Resveratrol/pharmacology , AMP-Activated Protein Kinases , Adult , Diabetes Mellitus/metabolism , Drug Evaluation, Preclinical , Fatty Acids/metabolism , Healthy Volunteers , Humans , Insulin Resistance , Lactic Acid/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/metabolism , Oxidation-Reduction/drug effects , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins c-akt , Quercetin/therapeutic use , Resveratrol/therapeutic use , Signal Transduction
2.
Endocrine ; 60(1): 112-121, 2018 04.
Article in English | MEDLINE | ID: mdl-29392617

ABSTRACT

PURPOSE: Apelin treatment has been shown to improve insulin sensitivity in insulin resistant mice by acting in skeletal muscles. However, the effects of systemic apelin on the hepatic energy metabolism have not been addressed. We thus aimed to determine the effect of chronic apelin treatment on the hepatic lipid metabolism in insulin resistant mice. The apelin receptor (APJ) expression was also studied in this context since its regulation has only been reported in severe liver pathologies. METHODS: Mice were fed a high-fat diet (HFD) in order to become obese and insulin resistant compared to chow fed mice (CD). HFD mice then received a daily intraperitoneal injection of apelin (0.1 µmol/kg) or PBS during 28 days. RESULTS: Triglycerides content and the expression of different lipogenesis-related genes were significantly decreased in the liver of HFD apelin-treated compared to PBS-treated mice. Moreover, at this stage of insulin resistance, the beta-oxidation was increased in liver homogenates of HFD PBS-treated mice compared to CD mice and reduced in HFD apelin-treated mice. Finally, APJ expression was not up-regulated in the liver of insulin resistant mice. In isolated hepatocytes from chow and HFD fed mice, apelin did not induce significant effect. CONCLUSIONS: Altogether, these results suggest that systemic apelin treatment decreases steatosis in insulin resistant mice without directly targeting hepatocytes.


Subject(s)
Apelin/pharmacology , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Liver/drug effects , Obesity/metabolism , Animals , Energy Metabolism/drug effects , Fatty Liver/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/metabolism , Male , Mice , Triglycerides/metabolism
3.
Sci Rep ; 8(1): 1097, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348496

ABSTRACT

Robust associations between low plasma level of natriuretic peptides (NP) and increased risk of type 2 diabetes (T2D) have been recently reported in humans. Adipose tissue (AT) is a known target of NP. However it is unknown whether NP signalling in human AT relates to insulin sensitivity and modulates glucose metabolism. We here show in two European cohorts that the NP receptor guanylyl cyclase-A (GC-A) expression in subcutaneous AT was down-regulated as a function of obesity grade while adipose NP clearance receptor (NPRC) was up-regulated. Adipose GC-A mRNA level was down-regulated in prediabetes and T2D, and negatively correlated with HOMA-IR and fasting blood glucose. We show for the first time that NP promote glucose uptake in a dose-dependent manner. This effect is reduced in adipocytes of obese individuals. NP activate mammalian target of rapamycin complex 1/2 (mTORC1/2) and Akt signalling. These effects were totally abrogated by inhibition of cGMP-dependent protein kinase and mTORC1/2 by rapamycin. We further show that NP treatment favoured glucose oxidation and de novo lipogenesis independently of significant gene regulation. Collectively, our data support a role for NP in blood glucose control and insulin sensitivity by increasing glucose uptake in human adipocytes. This effect is partly blunted in obesity.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Cyclic GMP/metabolism , Glucose/metabolism , Natriuretic Peptides/pharmacology , Adipose Tissue/metabolism , Biomarkers , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Humans , Insulin Resistance , Models, Biological , Obesity/genetics , Obesity/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
4.
Mol Nutr Food Res ; 61(9)2017 09.
Article in English | MEDLINE | ID: mdl-28544394

ABSTRACT

SCOPE: In recent years, several studies reported the role of eIF4E-binding proteins (4E-BPs) on the development of diet-induced obesity and insulin resistance. Our aim was to investigate the effect of 4E-BP protein deletion on lipid accumulation and metabolism in skeletal muscle in response to a high-fat diet induced obesity in 4E-BP1/2 DKO mice. METHODS AND RESULTS: Diet-induced obesity engendered increased ectopic accumulation of lipotoxic species in skeletal muscle of 4E-BP1 and 4E-BP2 double knockout mice (4E-BP1/2 DKO), namely diacylglycerols and ceramides. Increased lipid accumulation was associated with alterations in the expression of genes involved in fatty acid transport (FATP, CD36), diacylglycerol/triacylglycerol biosynthesis (GPAT1, AGPAT1, DGAT1), and ß-oxidation (CPT1b, MCAD). Diet-induced obesity resulted in increased lean mass and muscle in 4E-BP1/2 DKO mice despite the development of a more severe systemic insulin resistance. Since increased expression of genes of several proteolytic systems (MuRF1, atrogin/MAFbx, and cathepsin-l) in 4EBP1/2 DKO skeletal muscle was reported, the increase of skeletal muscle mass in 4E-BP1/2 DKO mice suggests that ablation of 4E-BPs compensate with activation of muscle anabolism. CONCLUSIONS: These findings indicate that 4E-BP proteins may prevent excess lipid accumulation in skeletal muscle and suggest that 4E-BPs are key regulators of muscle homeostasis regardless of insulin sensitivity.


Subject(s)
Carrier Proteins/physiology , Eukaryotic Initiation Factors/physiology , Muscle, Skeletal/metabolism , Obesity/metabolism , Phosphoproteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Diet, High-Fat , Insulin Resistance , Lipid Metabolism , Male , Mechanistic Target of Rapamycin Complex 1/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Proteostasis
5.
Diabetes ; 66(2): 314-324, 2017 02.
Article in English | MEDLINE | ID: mdl-27899482

ABSTRACT

The mediobasal hypothalamus (MBH) contains neurons capable of directly detecting metabolic signals such as glucose to control energy homeostasis. Among them, glucose-excited (GE) neurons increase their electrical activity when glucose rises. In view of previous work, we hypothesized that transient receptor potential canonical type 3 (TRPC3) channels are involved in hypothalamic glucose detection and the control of energy homeostasis. To investigate the role of TRPC3, we used constitutive and conditional TRPC3-deficient mouse models. Hypothalamic glucose detection was studied in vivo by measuring food intake and insulin secretion in response to increased brain glucose level. The role of TRPC3 in GE neuron response to glucose was studied by using in vitro calcium imaging on freshly dissociated MBH neurons. We found that whole-body and MBH TRPC3-deficient mice have increased body weight and food intake. The anorectic effect of intracerebroventricular glucose and the insulin secretory response to intracarotid glucose injection are blunted in TRPC3-deficient mice. TRPC3 loss of function or pharmacological inhibition blunts calcium responses to glucose in MBH neurons in vitro. Together, the results demonstrate that TRPC3 channels are required for the response to glucose of MBH GE neurons and the central effect of glucose on insulin secretion and food intake.


Subject(s)
Body Weight/genetics , Eating/genetics , Energy Metabolism/genetics , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Neurons/metabolism , TRPC Cation Channels/genetics , Animals , Blotting, Western , Fasting , Glucose Tolerance Test , Homeostasis , Hypothalamus/cytology , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , TRPC Cation Channels/metabolism
6.
Sci Rep ; 6: 38310, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27922115

ABSTRACT

Lipid droplets (LD) play a central role in lipid homeostasis by controlling transient fatty acid (FA) storage and release from triacylglycerols stores, while preventing high levels of cellular toxic lipids. This crucial function in oxidative tissues is altered in obesity and type 2 diabetes. Perilipin 5 (PLIN5) is a LD protein whose mechanistic and causal link with lipotoxicity and insulin resistance has raised controversies. We investigated here the physiological role of PLIN5 in skeletal muscle upon various metabolic challenges. We show that PLIN5 protein is elevated in endurance-trained (ET) subjects and correlates with muscle oxidative capacity and whole-body insulin sensitivity. When overexpressed in human skeletal muscle cells to recapitulate the ET phenotype, PLIN5 diminishes lipolysis and FA oxidation under basal condition, but paradoxically enhances FA oxidation during forskolin- and contraction- mediated lipolysis. Moreover, PLIN5 partly protects muscle cells against lipid-induced lipotoxicity. In addition, we demonstrate that down-regulation of PLIN5 in skeletal muscle inhibits insulin-mediated glucose uptake under normal chow feeding condition, while paradoxically improving insulin sensitivity upon high-fat feeding. These data highlight a key role of PLIN5 in LD function, first by finely adjusting LD FA supply to mitochondrial oxidation, and second acting as a protective factor against lipotoxicity in skeletal muscle.


Subject(s)
Lipid Droplets/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Perilipin-5/genetics , Satellite Cells, Skeletal Muscle/metabolism , Animals , Body Weight , Diglycerides/metabolism , Gene Expression , Humans , Insulin Resistance , Lipid Droplets/chemistry , Lipid Droplets/drug effects , Lipolysis/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Oleic Acid/metabolism , Oleic Acid/pharmacology , Oxidation-Reduction , Perilipin-5/metabolism , Physical Endurance/physiology , Primary Cell Culture , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Sedentary Behavior , Triglycerides/metabolism
7.
Sci Rep ; 6: 31849, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27549402

ABSTRACT

Apelin is a bioactive peptide involved in the control of energy metabolism. In the hypothalamus, chronic exposure to high levels of apelin is associated with an increase in hepatic glucose production, and then contributes to the onset of type 2 diabetes. However, the molecular mechanisms behind deleterious effects of chronic apelin in the brain and consequences on energy expenditure and thermogenesis are currently unknown. We aimed to evaluate the effects of chronic intracerebroventricular (icv) infusion of apelin in normal mice on hypothalamic inflammatory gene expression, energy expenditure, thermogenesis and brown adipose tissue functions. We have shown that chronic icv infusion of apelin increases the expression of pro-inflammatory factors in the hypothalamus associated with an increase in plasma interleukin-1 beta. In parallel, mice infused with icv apelin exhibit a significant lower energy expenditure coupled to a decrease in PGC1alpha, PRDM16 and UCP1 expression in brown adipose tissue which could explain the alteration of thermogenesis in these mice. These data provide compelling evidence that central apelin contributes to the development of type 2 diabetes by altering energy expenditure, thermogenesis and fat browning.


Subject(s)
Adipose Tissue, Brown/drug effects , Apelin/pharmacology , Energy Metabolism/drug effects , Thermogenesis/drug effects , Adipose Tissue, Brown/metabolism , Animals , Apelin/administration & dosage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Infusions, Intraventricular , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
8.
Mol Metab ; 5(7): 527-537, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27408777

ABSTRACT

OBJECTIVE: Recent data suggest that adipose triglyceride lipase (ATGL) plays a key role in providing energy substrate from triglyceride pools and that alterations of its expression/activity relate to metabolic disturbances in skeletal muscle. Yet little is known about its regulation. We here investigated the role of the protein G0/G1 Switch Gene 2 (G0S2), recently described as an inhibitor of ATGL in white adipose tissue, in the regulation of lipolysis and oxidative metabolism in skeletal muscle. METHODS: We first examined G0S2 protein expression in relation to metabolic status and muscle characteristics in humans. We next overexpressed and knocked down G0S2 in human primary myotubes to assess its impact on ATGL activity, lipid turnover and oxidative metabolism, and further knocked down G0S2 in vivo in mouse skeletal muscle. RESULTS: G0S2 protein is increased in skeletal muscle of endurance-trained individuals and correlates with markers of oxidative capacity and lipid content. Recombinant G0S2 protein inhibits ATGL activity by about 40% in lysates of mouse and human skeletal muscle. G0S2 overexpression augments (+49%, p < 0.05) while G0S2 knockdown strongly reduces (-68%, p < 0.001) triglyceride content in human primary myotubes and mouse skeletal muscle. We further show that G0S2 controls lipolysis and fatty acid oxidation in a strictly ATGL-dependent manner. These metabolic adaptations mediated by G0S2 are paralleled by concomitant changes in glucose metabolism through the modulation of Pyruvate Dehydrogenase Kinase 4 (PDK4) expression (5.4 fold, p < 0.001). Importantly, downregulation of G0S2 in vivo in mouse skeletal muscle recapitulates changes in lipid metabolism observed in vitro. CONCLUSION: Collectively, these data indicate that G0S2 plays a key role in the regulation of skeletal muscle ATGL activity, lipid content and oxidative metabolism.

9.
Sci Rep ; 6: 26442, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27220353

ABSTRACT

Dissecting exercise-mimicking pathways that can replicate the benefits of exercise in obesity and diabetes may lead to promising treatments for metabolic disorders. Muscle estrogen-related receptor gamma (ERRγ) is induced by exercise, and when over-expressed in the skeletal muscle mimics exercise by stimulating glycolytic-to-oxidative myofiber switch, mitochondrial biogenesis and angiogenesis in lean mice. The objective of this study was to test whether muscle ERRγ in obese mice mitigates weight gain and insulin resistance. To do so, ERRγ was selectively over-expressed in the skeletal muscle of obese and diabetic db/db mice. Muscle ERRγ over-expression successfully triggered glycolytic-to-oxidative myofiber switch, increased functional mitochondrial content and boosted vascular supply in the db/db mice. Despite aerobic remodeling, ERRγ surprisingly failed to improve whole-body energy expenditure, block muscle accumulation of triglycerides, toxic diacylglycerols (DAG) and ceramides or suppress muscle PKCε sarcolemmal translocation in db/db mice. Consequently, muscle ERRγ did not mitigate impaired muscle insulin signaling or insulin resistance in these mice. In conclusion, obesity and diabetes in db/db mice are not amenable to selective ERRγ-directed programming of classic exercise-like effects in the skeletal muscle. Other biochemical pathways or integrated whole-body effects of exercise may be critical for resisting diabetes and obesity.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Muscle, Skeletal/metabolism , Receptors, Estrogen/physiology , Animals , Diabetes Mellitus, Type 2/pathology , Glycolysis , Lipid Metabolism , Mice, Obese , Mice, Transgenic , Mitochondria, Muscle/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Oxidation-Reduction , Physical Conditioning, Animal
10.
Diabetes ; 64(12): 4033-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26253614

ABSTRACT

Circulating natriuretic peptide (NP) levels are reduced in obesity and predict the risk of type 2 diabetes (T2D). Since skeletal muscle was recently shown as a key target tissue of NP, we aimed to investigate muscle NP receptor (NPR) expression in the context of obesity and T2D. Muscle NPRA correlated positively with whole-body insulin sensitivity in humans and was strikingly downregulated in obese subjects and recovered in response to diet-induced weight loss. In addition, muscle NP clearance receptor (NPRC) increased in individuals with impaired glucose tolerance and T2D. Similar results were found in obese diabetic mice. Although no acute effect of brain NP (BNP) on insulin sensitivity was observed in lean mice, chronic BNP infusion improved blood glucose control and insulin sensitivity in skeletal muscle of obese and diabetic mice. This occurred in parallel with a reduced lipotoxic pressure in skeletal muscle due to an upregulation of lipid oxidative capacity. In addition, chronic NP treatment in human primary myotubes increased lipid oxidation in a PGC1α-dependent manner and reduced palmitate-induced lipotoxicity. Collectively, our data show that activation of NPRA signaling in skeletal muscle is important for the maintenance of long-term insulin sensitivity and has the potential to treat obesity-related metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Glucose Intolerance/etiology , Insulin Resistance , Muscle, Skeletal/metabolism , Obesity/physiopathology , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction , Adult , Animals , Body Mass Index , Cells, Cultured , Diabetes Mellitus, Type 2/prevention & control , Diet, Reducing , Disease Progression , Glucose Intolerance/prevention & control , Humans , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Middle Aged , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology , Obesity/diet therapy , Obesity/metabolism , Obesity/pathology , Random Allocation , Receptors, Atrial Natriuretic Factor/agonists , Receptors, Atrial Natriuretic Factor/genetics , Specific Pathogen-Free Organisms , Weight Loss
11.
Cell Rep ; 7(4): 1116-29, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24794440

ABSTRACT

Adipose tissue fibrosis development blocks adipocyte hypertrophy and favors ectopic lipid accumulation. Here, we show that adipose tissue fibrosis is associated with obesity and insulin resistance in humans and mice. Kinetic studies in C3H mice fed a high-fat diet show activation of macrophages and progression of fibrosis along with adipocyte metabolic dysfunction and death. Adipose tissue fibrosis is attenuated by macrophage depletion. Impairment of Toll-like receptor 4 signaling protects mice from obesity-induced fibrosis. The presence of a functional Toll-like receptor 4 on adipose tissue hematopoietic cells is necessary for the initiation of adipose tissue fibrosis. Continuous low-dose infusion of the Toll-like receptor 4 ligand, lipopolysaccharide, promotes adipose tissue fibrosis. Ex vivo, lipopolysaccharide-mediated induction of fibrosis is prevented by antibodies against the profibrotic factor TGFß1. Together, these results indicate that obesity and endotoxemia favor the development of adipose tissue fibrosis, a condition associated with insulin resistance, through immune cell Toll-like receptor 4.


Subject(s)
Adipose Tissue/pathology , Endotoxemia/metabolism , Obesity/metabolism , Toll-Like Receptor 4/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Endotoxemia/pathology , Fibrosis , Humans , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/physiology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Obesity/pathology , Signal Transduction , Toll-Like Receptor 4/genetics
12.
Diabetes ; 63(10): 3253-65, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24789919

ABSTRACT

The presence of brown adipose tissue (BAT) in human adults opens attractive perspectives to treat metabolic disorders. Indeed, BAT dissipates energy as heat via uncoupling protein (UCP)1. Brown adipocytes are located in specific deposits or can emerge among white fat through the so-called browning process. Although numerous inducers have been shown to drive this process, no study has investigated whether it could be controlled by specific metabolites. Here, we show that lactate, an important metabolic intermediate, induces browning of murine white adipose cells with expression of functional UCP1. Lactate-induced browning also occurs in human cells and in vivo. Lactate controls Ucp1 expression independently of hypoxia-inducible factor-1α and PPARα pathways but requires active PPARγ signaling. We demonstrate that the lactate effect on Ucp1 is mediated by intracellular redox modifications as a result of lactate transport through monocarboxylate transporters. Further, the ketone body ß-hydroxybutyrate, another metabolite that impacts redox state, is also a strong browning inducer. Because this redox-dependent increase in Ucp1 expression promotes an oxidative phenotype with mitochondria, browning appears as an adaptive mechanism to alleviate redox pressure. Our findings open new perspectives for the control of adipose tissue browning and its physiological relevance.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipogenesis/physiology , Animals , Energy Metabolism/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidation-Reduction , Oxygen Consumption/physiology , PPAR gamma/metabolism , Stem Cells
13.
J Clin Endocrinol Metab ; 98(12): 4863-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24178794

ABSTRACT

CONTEXT: Skeletal muscle lipase and intramyocellular triglyceride (IMTG) play a role in obesity-related metabolic disorders. OBJECTIVES: The aim of the present study was to investigate the impact of 8 weeks of endurance exercise training on IMTG content and lipolytic proteins in obese male subjects. DESIGN AND VOLUNTEERS: Ten obese subjects completed an 8-week supervised endurance exercise training intervention in which vastus lateralis muscle biopsy samples were collected before and after training. MAIN OUTCOME MEASURES: Clinical characteristics and ex vivo substrate oxidation rates were measured pre- and posttraining. Skeletal muscle lipid content and lipolytic protein expression were also investigated. RESULTS: Our data show that exercise training reduced IMTG content by 42% (P < .01) and increased skeletal muscle oxidative capacity, whereas no change in total diacylglycerol content and glucose oxidation was found. Exercise training up-regulated adipose triglyceride lipase, perilipin (PLIN) 3 protein, and PLIN5 protein contents in skeletal muscle despite no change in mRNA levels. Training also increased hormone sensitive-lipase Ser660 phosphorylation. No significant changes in comparative gene identification 58, G0/G1 switch gene 2, and PLIN2 protein and mRNA levels were observed in response to training. Interestingly, we noted a strong relationship between skeletal muscle comparative gene identification 58 and mitochondrial respiratory chain complex I protein contents at baseline (r = 0.87, P < .0001). CONCLUSIONS: Endurance exercise training coordinately up-regulates fat oxidative capacity and lipolytic protein expression in skeletal muscle of obese subjects. This physiological adaptation probably favors fat oxidation and may alleviate the lipotoxic lipid pressure in skeletal muscle. Enhancement of IMTG turnover may be required for the beneficial metabolic effects of exercise in obesity.


Subject(s)
Exercise , Lipolysis , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Obesity/metabolism , Physical Endurance , Triglycerides/metabolism , Adult , Body Mass Index , Cohort Studies , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipase/biosynthesis , Male , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Obesity/pathology , Obesity/therapy , Oxidative Phosphorylation , Perilipin-3 , Perilipin-5 , Phosphorylation , Protein Processing, Post-Translational , Proteins/genetics , Proteins/metabolism , Quadriceps Muscle/enzymology , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Sterol Esterase/metabolism , Up-Regulation , Vesicular Transport Proteins/biosynthesis
14.
J Clin Endocrinol Metab ; 98(9): 3739-47, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23884778

ABSTRACT

CONTEXT: It was suggested that human cultured primary myotubes retain the metabolic characteristics of their donor in vitro. OBJECTIVES: The aim of the present study was to investigate whether the metabolic responses to endurance training are also conserved in culture. DESIGN AND VOLUNTEERS: Middle-aged obese subjects completed an 8-week supervised aerobic exercise training program in which vastus lateralis muscle biopsies were collected before and after training. MAIN OUTCOME MEASURES: Anthropometric and blood parameters, as well as aerobic capacity, were assessed before and after training. Muscle biopsies were either used for Western blot analysis or digested to harvest myogenic progenitors that were differentiated into myotubes. Glucose oxidation, palmitate oxidation, and glycogen synthesis assays were performed on myotubes before and after training. Gene expression was assessed by real-time quantitative PCR. RESULTS: Our data indicate that in parallel of in vivo improvement of whole-body aerobic capacity and glucose metabolism, biopsy-derived primary myotubes showed similar patterns in vitro. Indeed, glucose oxidation, glycogen synthesis, and inhibition of palmitate oxidation by glucose were enhanced in myotubes after training. This was associated with consistent changes in the expression of metabolism-linked genes such as GLUT1, PDK4, and PDHA1. Interestingly, no difference in myogenic differentiation capacity was observed before and after training. CONCLUSION: Aerobic exercise training is associated with metabolic adaptations in vivo that are preserved in human cultured primary myotubes. It can be hypothesized that skeletal muscle microenvironmental changes induced by endurance training lead to metabolic imprinting on myogenic progenitor cells.


Subject(s)
Exercise Therapy , Exercise , Glucose/metabolism , Muscle Fibers, Skeletal/metabolism , Obesity/metabolism , Quadriceps Muscle/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycogen/biosynthesis , Humans , Male , Middle Aged , Muscle Fibers, Skeletal/pathology , Obesity/pathology , Obesity/therapy , Oxidation-Reduction , Palmitic Acid/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Quadriceps Muscle/pathology
15.
Endocrinology ; 154(4): 1444-53, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23471217

ABSTRACT

Elevated expression/activity of adipose triglyceride lipase (ATGL) and/or reduced activity of hormone-sensitive lipase (HSL) in skeletal muscle are causally linked to insulin resistance in vitro. We investigated here the effect of high-fat feeding on skeletal muscle lipolytic proteins, lipotoxicity, and insulin signaling in vivo. Five-week-old C3H mice were fed normal chow diet (NCD) or 45% kcal high-fat diet (HFD) for 4 weeks. Wild-type and HSL knockout mice fed NCD were also studied. Whole-body and muscle insulin sensitivity, as well as lipolytic protein expression, lipid levels, and insulin signaling in skeletal muscle, were measured. HFD induced whole-body insulin resistance and glucose intolerance and reduced skeletal muscle glucose uptake compared with NCD. HFD increased skeletal muscle total diacylglycerol (DAG) content, protein kinase Cθ and protein kinase Cε membrane translocation, and impaired insulin signaling as reflected by a robust increase of basal Ser1101 insulin receptor substrate 1 phosphorylation (2.8-fold, P < .05) and a decrease of insulin-stimulated v-Akt murine thymoma viral oncogene homolog Ser473 (-37%, P < .05) and AS160 Thr642 (-47%, P <.01) phosphorylation. We next showed that HFD strongly reduced HSL phosphorylation at Ser660. HFD significantly up-regulated the muscle protein content of the ATGL coactivator comparative gene identification 58 and triacylglycerol hydrolase activity, despite a lower ATGL protein content. We further show a defective skeletal muscle insulin signaling and DAG accumulation in HSL knockout compared with wild-type mice. Together, these data suggest a pathophysiological link between altered skeletal muscle lipase expression and DAG-mediated insulin resistance in mice.


Subject(s)
Diet, High-Fat , Insulin Resistance , Lipase/metabolism , Muscle, Skeletal/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Animals , Carrier Proteins/metabolism , Diglycerides/metabolism , Glucose/metabolism , Glucose Tolerance Test , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Muscle Proteins/metabolism , Perilipin-2 , Perilipin-3 , Phosphorylation , Weight Gain
16.
PLoS Biol ; 11(2): e1001485, 2013.
Article in English | MEDLINE | ID: mdl-23431266

ABSTRACT

When energy is needed, white adipose tissue (WAT) provides fatty acids (FAs) for use in peripheral tissues via stimulation of fat cell lipolysis. FAs have been postulated to play a critical role in the development of obesity-induced insulin resistance, a major risk factor for diabetes and cardiovascular disease. However, whether and how chronic inhibition of fat mobilization from WAT modulates insulin sensitivity remains elusive. Hormone-sensitive lipase (HSL) participates in the breakdown of WAT triacylglycerol into FAs. HSL haploinsufficiency and treatment with a HSL inhibitor resulted in improvement of insulin tolerance without impact on body weight, fat mass, and WAT inflammation in high-fat-diet-fed mice. In vivo palmitate turnover analysis revealed that blunted lipolytic capacity is associated with diminution in FA uptake and storage in peripheral tissues of obese HSL haploinsufficient mice. The reduction in FA turnover was accompanied by an improvement of glucose metabolism with a shift in respiratory quotient, increase of glucose uptake in WAT and skeletal muscle, and enhancement of de novo lipogenesis and insulin signalling in liver. In human adipocytes, HSL gene silencing led to improved insulin-stimulated glucose uptake, resulting in increased de novo lipogenesis and activation of cognate gene expression. In clinical studies, WAT lipolytic rate was positively and negatively correlated with indexes of insulin resistance and WAT de novo lipogenesis gene expression, respectively. In obese individuals, chronic inhibition of lipolysis resulted in induction of WAT de novo lipogenesis gene expression. Thus, reduction in WAT lipolysis reshapes FA fluxes without increase of fat mass and improves glucose metabolism through cell-autonomous induction of fat cell de novo lipogenesis, which contributes to improved insulin sensitivity.


Subject(s)
Adipose Tissue, White/drug effects , Adipose Tissue/metabolism , Lipid Metabolism/drug effects , Adipose Tissue/drug effects , Adipose Tissue, White/metabolism , Adolescent , Adult , Aged , Animals , Glucose , Humans , Lipolysis/drug effects , Male , Mice , Middle Aged , Niacin/pharmacology , Sterol Esterase/metabolism , Young Adult
17.
J Clin Invest ; 122(12): 4675-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23114600

ABSTRACT

Cardiac natriuretic peptides (NP) are major activators of human fat cell lipolysis and have recently been shown to control brown fat thermogenesis. Here, we investigated the physiological role of NP on the oxidative metabolism of human skeletal muscle. NP receptor type A (NPRA) gene expression was positively correlated to mRNA levels of PPARγ coactivator-1α (PGC1A) and several oxidative phosphorylation (OXPHOS) genes in human skeletal muscle. Further, the expression of NPRA, PGC1A, and OXPHOS genes was coordinately upregulated in response to aerobic exercise training in human skeletal muscle. In human myotubes, NP induced PGC-1α and mitochondrial OXPHOS gene expression in a cyclic GMP-dependent manner. NP treatment increased OXPHOS protein expression, fat oxidation, and maximal respiration independent of substantial changes in mitochondrial proliferation and mass. Treatment of myotubes with NP recapitulated the effect of exercise training on muscle fat oxidative capacity in vivo. Collectively, these data show that activation of NP signaling in human skeletal muscle enhances mitochondrial oxidative metabolism and fat oxidation. We propose that NP could contribute to exercise training-induced improvement in skeletal muscle fat oxidative capacity in humans.


Subject(s)
Atrial Natriuretic Factor/physiology , Muscle, Skeletal/metabolism , Natriuretic Peptide, Brain/physiology , Oxidative Phosphorylation , Receptors, Atrial Natriuretic Factor/metabolism , Adaptation, Physiological , Adult , Cells, Cultured , Gene Expression Regulation , Genes, Mitochondrial , Heat-Shock Proteins/metabolism , Humans , Lipid Metabolism , Male , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Obesity , Oxidation-Reduction , Oxygen Consumption , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Primary Cell Culture , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Transcription Factors/metabolism , Up-Regulation
18.
J Lipid Res ; 53(5): 839-848, 2012 May.
Article in English | MEDLINE | ID: mdl-22383684

ABSTRACT

We investigated here the specific role of CGI-58 in the regulation of energy metabolism in skeletal muscle. We first examined CGI-58 protein expression in various muscle types in mice, and next modulated CGI-58 expression during overexpression and knockdown studies in human primary myotubes and evaluated the consequences on oxidative metabolism. We observed a preferential expression of CGI-58 in oxidative muscles in mice consistent with triacylglycerol hydrolase activity. We next showed by pulse-chase that CGI-58 overexpression increased by more than 2-fold the rate of triacylglycerol (TAG) hydrolysis, as well as TAG-derived fatty acid (FA) release and oxidation. Oppositely, CGI-58 silencing reduced TAG hydrolysis and TAG-derived FA release and oxidation (-77%, P < 0.001), whereas it increased glucose oxidation and glycogen synthesis. Interestingly, modulations of CGI-58 expression and FA release are reflected by changes in pyruvate dehydrogenase kinase 4 gene expression. This regulation involves the activation of the peroxisome proliferator activating receptor-δ (PPARδ) by lipolysis products. Altogether, these data reveal that CGI-58 plays a limiting role in the control of oxidative metabolism by modulating FA availability and the expression of PPARδ-target genes, and highlight an important metabolic function of CGI-58 in skeletal muscle.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Energy Metabolism , Lipase/metabolism , Lipolysis , Muscle, Skeletal/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/deficiency , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Adolescent , Animals , Cells, Cultured , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Glucose/metabolism , Humans , Hydrolases/metabolism , Mice , Mitochondria/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/enzymology , Oxidation-Reduction , PPAR delta/metabolism , Triglycerides/metabolism , Young Adult
19.
Diabetes ; 60(6): 1734-42, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21498783

ABSTRACT

OBJECTIVE: Insulin resistance is associated with elevated content of skeletal muscle lipids, including triacylglycerols (TAGs) and diacylglycerols (DAGs). DAGs are by-products of lipolysis consecutive to TAG hydrolysis by adipose triglyceride lipase (ATGL) and are subsequently hydrolyzed by hormone-sensitive lipase (HSL). We hypothesized that an imbalance of ATGL relative to HSL (expression or activity) may contribute to DAG accumulation and insulin resistance. RESEARCH DESIGN AND METHODS: We first measured lipase expression in vastus lateralis biopsies of young lean (n = 9), young obese (n = 9), and obese-matched type 2 diabetic (n = 8) subjects. We next investigated in vitro in human primary myotubes the impact of altered lipase expression/activity on lipid content and insulin signaling. RESULTS: Muscle ATGL protein was negatively associated with whole-body insulin sensitivity in our population (r = -0.55, P = 0.005), whereas muscle HSL protein was reduced in obese subjects. We next showed that adenovirus-mediated ATGL overexpression in human primary myotubes induced DAG and ceramide accumulation. ATGL overexpression reduced insulin-stimulated glycogen synthesis (-30%, P < 0.05) and disrupted insulin signaling at Ser1101 of the insulin receptor substrate-1 and downstream Akt activation at Ser473. These defects were fully rescued by nonselective protein kinase C inhibition or concomitant HSL overexpression to restore a proper lipolytic balance. We show that selective HSL inhibition induces DAG accumulation and insulin resistance. CONCLUSIONS: Altogether, the data indicate that altered ATGL and HSL expression in skeletal muscle could promote DAG accumulation and disrupt insulin signaling and action. Targeting skeletal muscle lipases may constitute an interesting strategy to improve insulin sensitivity in obesity and type 2 diabetes.


Subject(s)
Insulin Resistance/physiology , Lipase/metabolism , Muscle, Skeletal/enzymology , Sterol Esterase/metabolism , Adult , Chromatography, Gas , Diglycerides/metabolism , Female , Glucose Clamp Technique , Humans , Insulin Resistance/genetics , Lipase/genetics , Male , Mass Spectrometry , Middle Aged , Spectrometry, Mass, Electrospray Ionization , Sterol Esterase/genetics , Tandem Mass Spectrometry , Triglycerides/metabolism , Young Adult
20.
Diabetes ; 56(11): 2797-808, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17704298

ABSTRACT

OBJECTIVE: Rosiglitazone displays powerful antidiabetes benefits but is associated with increased body weight and adipogenesis. Keeping in mind the concept of selective peroxisome proliferator-activated receptor (PPAR)gamma modulator, the aim of this study was to characterize the properties of a new PPARgamma ligand, S 26948, with special attention in body-weight gain. RESEARCH DESIGN AND METHODS: We used transient transfection and binding assays to characterized the binding characteristics of S 26948 and GST pull-down experiments to investigate its pattern of coactivator recruitment compared with rosiglitazone. We also assessed its adipogenic capacity in vitro using the 3T3-F442A cell line and its in vivo effects in ob/ob mice (for antidiabetes and antiobesity properties), as well as the homozygous human apolipoprotein E2 knocking mice (E2-KI) (for antiatherogenic capacity). RESULTS: S 26948 displayed pharmacological features of a high selective ligand for PPARgamma with low potency in promoting adipocyte differentiation. It also displayed a different coactivator recruitment profile compared with rosiglitazone, being unable to recruit DRIP205 or PPARgamma coactivator-1 alpha. In vivo experiments showed that S 26948 was as efficient in ameliorating glucose and lipid homeostasis as rosiglitazone, but it did not increase body and white adipose tissue weights and improved lipid oxidation in liver. In addition, S 26948 represented one of the few molecules of the PPARgamma ligand class able to decrease atherosclerotic lesions. CONCLUSIONS: These findings establish S 26948 as a selective PPARgamma ligand with distinctive coactivator recruitment and gene expression profile, reduced adipogenic effect, and improved biological responses in vivo.


Subject(s)
Atherosclerosis/prevention & control , Diabetic Angiopathies/prevention & control , Hypoglycemic Agents/pharmacology , PPAR gamma/physiology , Animals , COS Cells , Cell Membrane/physiology , Chlorocebus aethiops , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Haplorhini , Humans , Ligands , PPAR gamma/drug effects , PPAR gamma/genetics , Retinoid X Receptor alpha/drug effects , Retinoid X Receptor alpha/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL