Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Diabetologia ; 66(3): 567-578, 2023 03.
Article in English | MEDLINE | ID: mdl-36456864

ABSTRACT

AIMS/HYPOTHESIS: Athletes exhibit increased muscle insulin sensitivity, despite increased intramuscular triacylglycerol content. This phenomenon has been coined the 'athlete's paradox' and is poorly understood. Recent findings suggest that the subcellular distribution of sn-1,2-diacylglycerols (DAGs) in the plasma membrane leading to activation of novel protein kinase Cs (PKCs) is a crucial pathway to inducing insulin resistance. Here, we hypothesised that regular aerobic exercise would preserve muscle insulin sensitivity by preventing increases in plasma membrane sn-1,2-DAGs and activation of PKCε and PKCθ despite promoting increases in muscle triacylglycerol content. METHODS: C57BL/6J mice were allocated to three groups (regular chow feeding [RC]; high-fat diet feeding [HFD]; RC feeding and running wheel exercise [RC-EXE]). We used a novel LC-MS/MS/cellular fractionation method to assess DAG stereoisomers in five subcellular compartments (plasma membrane [PM], endoplasmic reticulum, mitochondria, lipid droplets and cytosol) in the skeletal muscle. RESULTS: We found that the HFD group had a greater content of sn-DAGs and ceramides in multiple subcellular compartments compared with the RC mice, which was associated with an increase in PKCε and PKCθ translocation. However, the RC-EXE mice showed, of particular note, a reduction in PM sn-1,2-DAG and ceramide content when compared with HFD mice. Consistent with the PM sn-1,2-DAG-novel PKC hypothesis, we observed an increase in phosphorylation of threonine1150 on the insulin receptor kinase (IRKT1150), and reductions in insulin-stimulated IRKY1162 phosphorylation and IRS-1-associated phosphoinositide 3-kinase activity in HFD compared with RC and RC-EXE mice, which are sites of PKCε and PKCθ action, respectively. CONCLUSIONS/INTERPRETATION: These results demonstrate that lower PKCθ/PKCε activity and sn-1,2-DAG content, especially in the PM compartment, can explain the preserved muscle insulin sensitivity in RC-EXE mice.


Subject(s)
Insulin Resistance , Mice , Animals , Insulin Resistance/physiology , Protein Kinase C-theta/metabolism , Protein Kinase C-epsilon/metabolism , Chromatography, Liquid , Phosphatidylinositol 3-Kinases/metabolism , Mice, Inbred C57BL , Tandem Mass Spectrometry , Insulin/metabolism , Muscle, Skeletal/metabolism , Triglycerides/metabolism , Ceramides/metabolism
2.
Proc Natl Acad Sci U S A ; 117(14): 8166-8176, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32188779

ABSTRACT

Multiple insulin-regulated enzymes participate in hepatic glycogen synthesis, and the rate-controlling step responsible for insulin stimulation of glycogen synthesis is unknown. We demonstrate that glucokinase (GCK)-mediated glucose phosphorylation is the rate-controlling step in insulin-stimulated hepatic glycogen synthesis in vivo, by use of the somatostatin pancreatic clamp technique using [13C6]glucose with metabolic control analysis (MCA) in three rat models: 1) regular chow (RC)-fed male rats (control), 2) high fat diet (HFD)-fed rats, and 3) RC-fed rats with portal vein glucose delivery at a glucose infusion rate matched to the control. During hyperinsulinemia, hyperglycemia dose-dependently increased hepatic glycogen synthesis. At similar levels of hyperinsulinemia and hyperglycemia, HFD-fed rats exhibited a decrease and portal delivery rats exhibited an increase in hepatic glycogen synthesis via the direct pathway compared with controls. However, the strong correlation between liver glucose-6-phosphate concentration and net hepatic glycogen synthetic rate was nearly identical in these three groups, suggesting that the main difference between models is the activation of GCK. MCA yielded a high control coefficient for GCK in all three groups. We confirmed these findings in studies of hepatic GCK knockdown using an antisense oligonucleotide. Reduced liver glycogen synthesis in lipid-induced hepatic insulin resistance and increased glycogen synthesis during portal glucose infusion were explained by concordant changes in translocation of GCK. Taken together, these data indicate that the rate of insulin-stimulated hepatic glycogen synthesis is controlled chiefly through GCK translocation.


Subject(s)
Fatty Liver/pathology , Glucokinase/metabolism , Glucose/metabolism , Liver Glycogen/biosynthesis , Liver/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Gene Knockdown Techniques , Glucokinase/genetics , Glucose/administration & dosage , Glucose-6-Phosphate/analysis , Glucose-6-Phosphate/metabolism , Humans , Hyperglycemia/etiology , Hyperglycemia/pathology , Hyperinsulinism/etiology , Hyperinsulinism/pathology , Insulin/metabolism , Insulin Resistance , Liver/pathology , Male , Metabolomics , Phosphorylation , Rats
3.
Nat Med ; 25(3): 526-528, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30733621

ABSTRACT

In the version of this article originally published, the VPC and VCS flux data shown in Fig. 6e,f were inadvertently duplicated from Fig. 5j,k. The correct data are now shown in Fig. 6e,f. In these corrected data, VPC flux in response to chronic oral metformin treatment was still significantly decreased (Fig. 6e), and there was still no impact of metformin on VCS flux (Fig. 6f). Therefore, the text describing these data remains the same and this correction does not change the conclusion of this study.

4.
Nat Med ; 24(9): 1384-1394, 2018 09.
Article in English | MEDLINE | ID: mdl-30038219

ABSTRACT

Metformin, the universal first-line treatment for type 2 diabetes, exerts its therapeutic glucose-lowering effects by inhibiting hepatic gluconeogenesis. However, the primary molecular mechanism of this biguanide remains unclear, though it has been suggested to act, at least partially, by mitochondrial complex I inhibition. Here we show that clinically relevant concentrations of plasma metformin achieved by acute intravenous, acute intraportal or chronic oral administration in awake normal and diabetic rats inhibit gluconeogenesis from lactate and glycerol but not from pyruvate and alanine, implicating an increased cytosolic redox state in mediating metformin's antihyperglycemic effect. All of these effects occurred independently of complex I inhibition, evidenced by unaltered hepatic energy charge and citrate synthase flux. Normalizing the cytosolic redox state by infusion of methylene blue or substrates that contribute to gluconeogenesis independently of the cytosolic redox state abrogated metformin-mediated inhibition of gluconeogenesis in vivo. Additionally, in mice expressing constitutively active acetyl-CoA carboxylase, metformin acutely decreased hepatic glucose production and increased the hepatic cytosolic redox state without altering hepatic triglyceride content or gluconeogenic enzyme expression. These studies demonstrate that metformin, at clinically relevant plasma concentrations, inhibits hepatic gluconeogenesis in a redox-dependent manner independently of reductions in citrate synthase flux, hepatic nucleotide concentrations, acetyl-CoA carboxylase activity, or gluconeogenic enzyme protein expression.


Subject(s)
Gluconeogenesis/drug effects , Metformin/pharmacology , Acetyl-CoA Carboxylase/metabolism , Adenylate Kinase/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Dihydroxyacetone/metabolism , Disease Models, Animal , Injections, Intravenous , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Metformin/administration & dosage , Mice , Oxidation-Reduction , Phosphorylation/drug effects , Pyruvic Acid/metabolism , Rats, Sprague-Dawley , Streptozocin
5.
Cell Rep ; 19(10): 1997-2004, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28591572

ABSTRACT

Hepatic lipid accumulation has been implicated in the development of insulin resistance, but translational evidence in humans is limited. We investigated the relationship between liver fat and tissue-specific insulin sensitivity in 133 obese subjects. Although the presence of hepatic steatosis in obese subjects was associated with hepatic, adipose tissue, and peripheral insulin resistance, we found that intrahepatic triglycerides were not strictly sufficient or essential for hepatic insulin resistance. Thus, to examine the molecular mechanisms that link hepatic steatosis to hepatic insulin resistance, we comprehensively analyzed liver biopsies from a subset of 29 subjects. Here, hepatic cytosolic diacylglycerol content, but not hepatic ceramide content, was increased in subjects with hepatic insulin resistance. Moreover, cytosolic diacylglycerols were strongly associated with hepatic PKCε activation, as reflected by PKCε translocation to the plasma membrane. These results demonstrate the relevance of hepatic diacylglycerol-induced PKCε activation in the pathogenesis of NAFLD-associated hepatic insulin resistance in humans.


Subject(s)
Ceramides/metabolism , Diglycerides/metabolism , Insulin Resistance , Non-alcoholic Fatty Liver Disease/metabolism , Protein Kinase C-epsilon/metabolism , Enzyme Activation , Female , Humans , Male , Non-alcoholic Fatty Liver Disease/pathology , Protein Transport
6.
Nature ; 510(7503): 84-91, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24899308

ABSTRACT

Non-alcoholic fatty liver disease and its downstream sequelae, hepatic insulin resistance and type 2 diabetes, are rapidly growing epidemics, which lead to increased morbidity and mortality rates, and soaring health-care costs. Developing interventions requires a comprehensive understanding of the mechanisms by which excess hepatic lipid develops and causes hepatic insulin resistance and type 2 diabetes. Proposed mechanisms implicate various lipid species, inflammatory signalling and other cellular modifications. Studies in mice and humans have elucidated a key role for hepatic diacylglycerol activation of protein kinase Cε in triggering hepatic insulin resistance. Therapeutic approaches based on this mechanism could alleviate the related epidemics of non-alcoholic fatty liver disease and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Lipid Metabolism , Lipids , Liver/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Diglycerides/metabolism , Fatty Liver/drug therapy , Fatty Liver/metabolism , Humans , Hyperglycemia/metabolism , Lipids/biosynthesis , Lipodystrophy/metabolism , Lipogenesis , Muscle, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease , Triglycerides/biosynthesis
7.
Hepatology ; 57(5): 1763-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23175050

ABSTRACT

UNLABELLED: Genome-wide array studies have associated the patatin-like phospholipase domain-containing 3 (PNPLA3) gene polymorphisms with hepatic steatosis. However, it is unclear whether PNPLA3 functions as a lipase or a lipogenic enzyme and whether PNPLA3 is involved in the pathogenesis of hepatic insulin resistance. To address these questions we treated high-fat-fed rats with specific antisense oligonucleotides to decrease hepatic and adipose pnpla3 expression. Reducing pnpla3 expression prevented hepatic steatosis, which could be attributed to decreased fatty acid esterification measured by the incorporation of [U-(13) C]-palmitate into hepatic triglyceride. While the precursors for phosphatidic acid (PA) (long-chain fatty acyl-CoAs and lysophosphatidic acid [LPA]) were not decreased, we did observe an ∼20% reduction in the hepatic PA content, ∼35% reduction in the PA/LPA ratio, and ∼60%-70% reduction in transacylation activity at the level of acyl-CoA:1-acylglycerol-sn-3-phosphate acyltransferase. These changes were associated with an ∼50% reduction in hepatic diacylglycerol (DAG) content, an ∼80% reduction in hepatic protein kinase Cε activation, and increased hepatic insulin sensitivity, as reflected by a 2-fold greater suppression of endogenous glucose production during the hyperinsulinemic-euglycemic clamp. Finally, in humans, hepatic PNPLA3 messenger RNA (mRNA) expression was strongly correlated with hepatic triglyceride and DAG content, supporting a potential lipogenic role of PNPLA3 in humans. CONCLUSION: PNPLA3 may function primarily in a lipogenic capacity and inhibition of PNPLA3 may be a novel therapeutic approach for treatment of nonalcoholic fatty liver disease-associated hepatic insulin resistance.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/chemically induced , Fatty Liver/physiopathology , Insulin Resistance/physiology , Lipids/adverse effects , Membrane Proteins/physiology , Phospholipases A2/physiology , Animals , Biopsy , Diglycerides/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Membrane Proteins/drug effects , Membrane Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Phospholipases A2/drug effects , Phospholipases A2/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Triglycerides/metabolism
8.
J Nucl Med ; 53(6): 908-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22573821

ABSTRACT

UNLABELLED: The ability to noninvasively measure endogenous pancreatic ß-cell mass (BCM) would accelerate research on the pathophysiology of diabetes and revolutionize the preclinical development of new treatments, the clinical assessment of therapeutic efficacy, and the early diagnosis and subsequent monitoring of disease progression. The vesicular monoamine transporter type 2 (VMAT2) is coexpressed with insulin in ß-cells and represents a promising target for BCM imaging. METHODS: We evaluated the VMAT2 radiotracer (18)F-fluoropropyl-dihydrotetrabenazine ((18)F-FP-(+)-DTBZ, also known as (18)F-AV-133) for quantitative PET of BCM in healthy control subjects and patients with type 1 diabetes mellitus. Standardized uptake value was calculated as the net tracer uptake in the pancreas normalized by injected dose and body weight. Total volume of distribution, the equilibrium ratio of tracer concentration in tissue relative to plasma, was estimated by kinetic modeling with arterial input functions. Binding potential, the steady-state ratio of specific binding to nondisplaceable uptake, was calculated using the renal cortex as a reference tissue devoid of specific VMAT2 binding. RESULTS: Mean pancreatic standardized uptake value, total volume of distribution, and binding potential were reduced by 38%, 20%, and 40%, respectively, in type 1 diabetes mellitus. The radiotracer binding parameters correlated with insulin secretion capacity as determined by arginine-stimulus tests. Group differences and correlations with ß-cell function were enhanced for total pancreas binding parameters that accounted for tracer binding density and organ volume. CONCLUSION: These findings demonstrate that quantitative evaluation of islet ß-cell density and aggregate BCM can be performed clinically with (18)F-FP-(+)-DTBZ PET.


Subject(s)
Diabetes Mellitus, Type 1/diagnostic imaging , Insulin-Secreting Cells/diagnostic imaging , Positron-Emission Tomography/methods , Tetrabenazine/analogs & derivatives , Adolescent , Adult , Animals , Female , Fluorine Radioisotopes , Humans , Insulin-Secreting Cells/physiology , Macaca mulatta , Magnetic Resonance Imaging , Male , Middle Aged
9.
Hepatology ; 54(5): 1650-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21793029

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) and insulin resistance have recently been found to be associated with increased plasma concentrations of apolipoprotein CIII (APOC3) in humans carrying single nucleotide polymorphisms within the insulin response element of the APOC3 gene. To examine whether increased expression of APOC3 would predispose mice to NAFLD and hepatic insulin resistance, human APOC3 overexpressing (ApoC3Tg) mice were metabolically phenotyped following either a regular chow or high-fat diet (HFD). After HFD feeding, ApoC3Tg mice had increased hepatic triglyceride accumulation, which was associated with cellular ballooning and inflammatory changes. ApoC3Tg mice also manifested severe hepatic insulin resistance assessed by a hyperinsulinemic-euglycemic clamp, which could mostly be attributed to increased hepatic diacylglycerol content, protein kinase C-ϵ activation, and decreased insulin-stimulated Akt2 activity. Increased hepatic triglyceride content in the HFD-fed ApoC3Tg mice could be attributed to a ≈ 70% increase in hepatic triglyceride uptake and ≈ 50% reduction hepatic triglyceride secretion. CONCLUSION: These data demonstrate that increase plasma APOC3 concentrations predispose mice to diet-induced NAFLD and hepatic insulin resistance.


Subject(s)
Apolipoprotein C-III/blood , Apolipoprotein C-III/genetics , Fatty Liver/genetics , Fatty Liver/metabolism , Insulin Resistance/genetics , Animal Feed , Animals , Apolipoprotein B-100/metabolism , Blood Glucose/metabolism , Cholesterol, VLDL/metabolism , Dietary Fats/pharmacology , Diglycerides/metabolism , Female , Genetic Predisposition to Disease/genetics , Hyperinsulinism/metabolism , Male , Mice , Mice, Mutant Strains , Non-alcoholic Fatty Liver Disease , Postprandial Period/physiology , Protein Kinase C/metabolism , Triglycerides/metabolism , Triglycerides/pharmacokinetics
10.
Cell Metab ; 12(6): 668-74, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21109199

ABSTRACT

Aging-associated muscle insulin resistance has been hypothesized to be due to decreased mitochondrial function, secondary to cumulative free radical damage, leading to increased intramyocellular lipid content. To directly test this hypothesis, we examined both in vivo and in vitro mitochondrial function, intramyocellular lipid content, and insulin action in lean healthy mice with targeted overexpression of the human catalase gene to mitochondria (MCAT mice). Here, we show that MCAT mice are protected from age-induced decrease in muscle mitochondrial function (∼30%), energy metabolism (∼7%), and lipid-induced muscle insulin resistance. This protection from age-induced reduction in mitochondrial function was associated with reduced mitochondrial oxidative damage, preserved mitochondrial respiration and muscle ATP synthesis, and AMP-activated protein kinase-induced mitochondrial biogenesis. Taken together, these data suggest that the preserved mitochondrial function maintained by reducing mitochondrial oxidative damage may prevent age-associated whole-body energy imbalance and muscle insulin resistance.


Subject(s)
Aging/metabolism , Catalase/metabolism , Energy Metabolism/physiology , Insulin Resistance/physiology , Mitochondria/metabolism , Mitochondria/physiology , Muscle, Skeletal/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Catalase/genetics , DNA Damage , Humans , Insulin/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Oxidative Stress/physiology , Oxygen Consumption/physiology , Reactive Oxygen Species/metabolism
11.
J Neurosci ; 30(42): 13983-91, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20962220

ABSTRACT

To determine whether plasma lactate can be a significant fuel for human brain energy metabolism, infusions of [3-(13)C]lactate and (1)H-(13)C polarization transfer spectroscopy were used to detect the entry and utilization of lactate. During the 2 h infusion study, (13)C incorporation in the amino acid pools of glutamate and glutamine were measured with a 5 min time resolution. With a plasma concentration ([Lac](P)) being in the 0.8-2.8 mmol/L range, the tissue lactate concentration ([Lac](B)) was assessed as well as the fractional contribution of lactate to brain energy metabolism (CMRlac). From the measured relationship between unidirectional lactate influx (V(in)) and plasma and brain lactate concentrations, lactate transport constants were calculated using a reversible Michaelis-Menten model. The results show that (1) in the physiological range, plasma lactate unidirectional transport (V(in)) and concentration in tissue increase close to linearly with the lactate concentration in plasma; (2) the maximum potential contribution of plasma lactate to brain metabolism is 10% under basal plasma lactate conditions of ∼1.0 mmol/L and as much as 60% at supraphysiological plasma lactate concentrations when the transporters are saturated; (3) the half-saturation constant K(T) is 5.1 ± 2.7 mmol/L and V(MAX) is 0.40 ± 0.13 µmol · g(-1) · min(-1) (68% confidence interval); and (4) the majority of plasma lactate is metabolized in neurons similar to glucose.


Subject(s)
Brain Chemistry/physiology , Energy Metabolism/physiology , Lactic Acid/blood , Adult , Algorithms , Astrocytes/metabolism , Carbon Radioisotopes , Data Interpretation, Statistical , Female , Glucose/metabolism , Humans , Kinetics , Magnetic Resonance Spectroscopy , Male , Neuroglia/metabolism , Neurons/metabolism , Occipital Lobe/cytology , Occipital Lobe/metabolism , Parietal Lobe/metabolism , Tricarboxylic Acids/metabolism , Young Adult
12.
J Cereb Blood Flow Metab ; 30(1): 211-21, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19794401

ABSTRACT

A decline in brain function is a characteristic feature of healthy aging; however, little is known about the biologic basis of this phenomenon. To determine whether there are alterations in brain mitochondrial metabolism associated with healthy aging, we combined (13)C/(1)H magnetic resonance spectroscopy with infusions of [1-(13)C]glucose and [2-(13)C]acetate to quantitatively characterize rates of neuronal and astroglial tricarboxylic acid cycles, as well as neuroglial glutamate-glutamine cycling, in healthy elderly and young volunteers. Compared with young subjects, neuronal mitochondrial metabolism and glutamate-glutamine cycle flux was approximately 30% lower in elderly subjects. The reduction in individual subjects correlated strongly with reductions in N-acetylaspartate and glutamate concentrations consistent with chronic reductions in brain mitochondrial function. In elderly subjects infused with [2-(13)C]acetate labeling of glutamine, C4 and C3 differed from that of the young subjects, indicating age-related changes in glial mitochondrial metabolism. Taken together, these studies show that healthy aging is associated with reduced neuronal mitochondrial metabolism and altered glial mitochondrial metabolism, which may in part be responsible for declines in brain function.


Subject(s)
Aging/physiology , Brain Chemistry/physiology , Mitochondria/metabolism , Adult , Aged , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Brain/anatomy & histology , Female , Glucose Tolerance Test , Glutamic Acid/metabolism , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male
13.
Proc Natl Acad Sci U S A ; 105(50): 19926-31, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19066218

ABSTRACT

Peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1alpha has been shown to play critical roles in regulating mitochondria biogenesis, respiration, and muscle oxidative phenotype. Furthermore, reductions in the expression of PGC-1alpha in muscle have been implicated in the pathogenesis of type 2 diabetes. To determine the effect of increased muscle-specific PGC-1alpha expression on muscle mitochondrial function and glucose and lipid metabolism in vivo, we examined body composition, energy balance, and liver and muscle insulin sensitivity by hyperinsulinemic-euglycemic clamp studies and muscle energetics by using (31)P magnetic resonance spectroscopy in transgenic mice. Increased expression of PGC-1alpha in muscle resulted in a 2.4-fold increase in mitochondrial density, which was associated with an approximately 60% increase in the unidirectional rate of ATP synthesis. Surprisingly, there was no effect of increased muscle PGC-1alpha expression on whole-body energy expenditure, and PGC-1alpha transgenic mice were more prone to fat-induced insulin resistance because of decreased insulin-stimulated muscle glucose uptake. The reduced insulin-stimulated muscle glucose uptake could most likely be attributed to a relative increase in fatty acid delivery/triglyceride reesterfication, as reflected by increased expression of CD36, acyl-CoA:diacylglycerol acyltransferase1, and mitochondrial acyl-CoA:glycerol-sn-3-phosphate acyltransferase, that may have exceeded mitochondrial fatty acid oxidation, resulting in increased intracellular lipid accumulation and an increase in the membrane to cytosol diacylglycerol content. This, in turn, caused activation of PKC, decreased insulin signaling at the level of insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation, and skeletal muscle insulin resistance.


Subject(s)
Glucose/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Trans-Activators/biosynthesis , Animals , Diet , Energy Metabolism , Fats/administration & dosage , Fats/metabolism , Fatty Acids/metabolism , Gene Expression , Insulin/pharmacology , Insulin Resistance , Mice , Mice, Transgenic , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/ultrastructure , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors
14.
Diabetes ; 55(4): 929-34, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16567513

ABSTRACT

We hypothesized that increased capacity for brain utilization of nonglucose substrates (monocarboxylic acids [MCAs]) by upregulation of the MCA transporters may contribute metabolic substrates during hypoglycemia. To test this hypothesis, we assessed brain acetate metabolism in five well-controlled type 1 diabetic subjects and six nondiabetic control subjects using 13C magnetic resonance spectroscopy during infusions of [2-(13)C]acetate during hypoglycemia (approximately 55 mg/dl). Acetate is transported into the brain through MCA transporters that are also used for lactate and ketones. Brain acetate concentrations were over twofold higher in the subjects with diabetes than the control subjects (P = 0.01). The fraction of oxidative metabolism from acetate (P = 0.015) and the rate of MCA transport (P = 0.01) were also approximately twofold higher in the diabetic subjects. We conclude that during hypoglycemia MCA transport in the brain was increased by approximately twofold in patients with well-controlled type 1 diabetes, as reflected by higher brain acetate concentrations and rates of acetate oxidation. This upregulation would potentially allow a similar twofold increase in the transport of other MCAs, including lactate, during insulin-induced hypoglycemia. These data are consistent with the hypothesis that upregulation of MCA transport may contribute to the maintenance of brain energetics during hypoglycemia in patients with type 1 diabetes.


Subject(s)
Acetates/metabolism , Brain/metabolism , Diabetes Mellitus, Type 1/metabolism , Glutamic Acid/metabolism , gamma-Aminobutyric Acid/metabolism , Adult , Biological Transport , Diabetes Mellitus, Type 1/blood , Female , Humans , Insulin/blood , Kinetics , Male , Reference Values
15.
PLoS Med ; 2(9): e233, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16089501

ABSTRACT

BACKGROUND: Insulin resistance is the best predictor for the development of type 2 diabetes. Recent studies have shown that young, lean, insulin-resistant (IR) offspring of parents with type 2 diabetes have reduced basal rates of muscle mitochondrial phosphorylation activity associated with increased intramyocellular lipid (IMCL) content, which in turn blocks insulin signaling and insulin action in muscle. In order to further characterize mitochondrial activity in these individuals, we examined insulin-stimulated rates of adenosine triphosphate (ATP) synthesis and phosphate transport in skeletal muscle in a similar cohort of participants. METHODS AND FINDINGS: Rates of insulin-stimulated muscle mitochondrial ATP synthase flux and insulin-stimulated increases in concentrations of intramyocellular inorganic phosphate (Pi) were assessed by 31P magnetic resonance spectroscopy (MRS) in healthy, lean, IR offspring of parents with type 2 diabetes and healthy, lean control participants with normal insulin sensitivity. IMCL content in the soleus muscle of all participants was assessed by 1H MRS. During a hyperinsulinemic-euglycemic clamp, rates of insulin-stimulated glucose uptake were decreased by approximately 50% in the IR offspring compared to the control participants (p = 0.007 versus controls) and were associated with an approximately 2-fold increase in IMCL content (p < 0.006 versus controls). In the control participants rates of ATP synthesis increased by approximately 90% during the hyperinsulinemic-euglycemic clamp. In contrast, insulin-stimulated rates of muscle mitochondrial ATP synthesis increased by only 5% in the IR offspring (p = 0.001 versus controls) and was associated with a severe reduction of insulin-stimulated increases in the intramyocellular Pi concentrations (IR offspring: 4.7% +/- 1.9% versus controls: 19.3% +/- 5.7%; p = 0.03). Insulin-induced increases in intramyocellular Pi concentrations correlated well with insulin-stimulated increases in rates of ATP synthesis (r = 0.67; p = 0.008). CONCLUSIONS: These data demonstrate that insulin-stimulated rates of mitochondrial ATP synthesis are reduced in IR offspring of parents with type 2 diabetes. Furthermore, these IR offspring also have impaired insulin-stimulated phosphate transport in muscle, which may contribute to their defects in insulin-stimulated rates of mitochondrial ATP synthesis.


Subject(s)
Adenosine Triphosphate/metabolism , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Insulin/administration & dosage , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Adenosine Triphosphate/biosynthesis , Adult , Calorimetry, Indirect , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/metabolism , Female , Genetic Predisposition to Disease , Glucose/metabolism , Glucose Clamp Technique , Humans , Lipid Metabolism , Magnetic Resonance Spectroscopy , Male , Oxidative Phosphorylation , Phosphates/metabolism
16.
Hypertension ; 45(5): 828-33, 2005 May.
Article in English | MEDLINE | ID: mdl-15824195

ABSTRACT

Insulin resistance is a major player in the pathogenesis of the metabolic syndrome and type 2 diabetes, and yet, the mechanisms responsible for it remain poorly understood. Magnetic resonance spectroscopy studies in humans suggest that a defect in insulin-stimulated glucose transport in skeletal muscle is the primary metabolic abnormality in insulin-resistant type 2 diabetics. Fatty acids appear to cause this defect in glucose transport by inhibiting insulin-stimulated tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1) and IRS-1 associated phosphatidylinositol 3-kinase activity. A number of different metabolic abnormalities may increase intramyocellular/intrahepatic fatty acid metabolites; these include increased fat delivery to muscle/liver as a consequence of either excess energy intake or defects in adipocyte fat metabolism and acquired or inherited defects in mitochondrial fatty acid oxidation. Understanding the molecular/biochemical defects responsible for insulin resistance is beginning to unveil novel therapeutic targets for treatment of the metabolic syndrome and type 2 diabetes.


Subject(s)
Inflammation/etiology , Insulin Resistance/physiology , Animals , Fatty Acids/metabolism , Glucose/metabolism , Humans , Lipid Metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Obesity/complications , Obesity/physiopathology
17.
Lancet ; 362(9388): 951-7, 2003 Sep 20.
Article in English | MEDLINE | ID: mdl-14511928

ABSTRACT

BACKGROUND: Impaired glucose tolerance is common among obese adolescents, but the changes in insulin sensitivity and secretion that lead to this prediabetic state are unknown. We investigated whether altered partitioning of myocellular and abdominal fat relates to abnormalities in glucose homoeostasis in obese adolescents with prediabetes. METHODS: We studied 14 obese children with impaired glucose tolerance and 14 with normal glucose tolerance, of similar ages, sex distribution, and degree of obesity. Insulin sensitivity and secretion were assessed by the euglycaemic-hyperinsulinaemic clamp and the hyperglycaemic clamp. Intramyocellular lipid was assessed by proton nuclear magnetic resonance spectroscopy and abdominal fat distribution by magnetic resonance imaging. FINDINGS: Peripheral glucose disposal was significantly lower in individuals with impaired than in those with normal glucose tolerance (mean 35.4 [SE 4.0] vs 60.6 [7.2] micromoles per kg lean body mass per min; p=0.023) owing to a reduction in non-oxidative glucose disposal metabolism (storage). Individuals with impaired glucose tolerance had higher intramyocellular lipid content (3.04 [0.43] vs 1.99 [0.19]%, p=0.03), lower abdominal subcutaneous fat (460 [47] vs 626 [39] cm2, p=0.04), and slightly higher visceral fat than the controls (70 [11] vs 47 [6] cm2, p=0.065), resulting in a higher ratio of visceral to subcutaneous fat (0.15 [0.02] vs 0.07 [0.01], p=0.002). Intramyocellular and visceral lipid contents were inversely related to the glucose disposal and non-oxidative glucose metabolism and positively related to the 2 h plasma glucose concentration. INTERPRETATION: In obese children and adolescents with prediabetes, intramyocellular and intra-abdominal lipid accumulation is closely linked to the development of severe peripheral insulin resistance.


Subject(s)
Abdomen/anatomy & histology , Adipose Tissue/anatomy & histology , Glucose Tolerance Test , Insulin Resistance , Muscle, Skeletal/anatomy & histology , Prediabetic State/diagnosis , Adolescent , Child , Female , Humans , Insulin/blood , Male , Obesity , Prediabetic State/blood
19.
Am J Cardiol ; 90(5A): 11G-18G, 2002 Sep 05.
Article in English | MEDLINE | ID: mdl-12231074

ABSTRACT

Insulin resistance is a principal feature of type 2 diabetes and precedes the clinical development of the disease by 10 to 20 years. Insulin resistance is caused by the decreased ability of peripheral target tissues (especially muscle) to respond properly to normal circulating concentrations of insulin. Defects in muscle glycogen synthesis play a significant role in insulin resistance, and 3 potentially rate-controlling steps in muscle glucose metabolism have been implicated in its pathogenesis: glycogen synthase, hexokinase, and GLUT4 (the major insulin-stimulated glucose transporter). Results from recent studies using nuclear magnetic resonance (NMR) spectroscopy implicate intracellular defects in glucose transport as the rate-controlling step for insulin-mediated glucose uptake in muscle. These alterations in glucose transport activity are likely the result of dysregulation of intramyocellular fatty acid metabolism, whereby fatty acids cause insulin resistance by activation of a serine kinase cascade, leading to decreased insulin-stimulated insulin receptor substrate (IRS)-1 tyrosine phosphorylation and decreased IRS-1-associated phosphatidylinositol 3-kinase activity, a required step in insulin-stimulated glucose transport into muscle. The thiazolidinedione class of antidiabetic agents directly targets insulin resistance in skeletal muscle by improving glucose transport activity and insulin-stimulated muscle glycogen synthesis. Although the precise mechanism of action is not known, recent NMR studies support the hypothesis that these agents improve insulin action in skeletal muscle and liver by promoting a redistribution of fat out of these tissues and into peripheral adipocytes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Diabetes Mellitus, Type 2/drug therapy , Fatty Acids/metabolism , Glycogen/biosynthesis , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Magnetic Resonance Spectroscopy , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Signal Transduction , Thiazoles/pharmacology
20.
J Cereb Blood Flow Metab ; 22(7): 890-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12142574

ABSTRACT

Infusions of [2,4-13C2]-beta-hydroxybutyrate and 1H-13C polarization transfer spectroscopy were used in normal human subjects to detect the entry and metabolism of beta-hydroxybutyrate in the brain. During the 2-hour infusion study, 13C label was detectable in the beta-hydroxybutyrate resonance positions and in the amino acid pools of glutamate, glutamine, and aspartate. With a plasma concentration of 2.25 +/- 0.24 mmol/L (four volunteers), the apparent tissue beta-hydroxybutyrate concentration reached 0.18 +/- 0.06 mmol/L during the last 20 minutes of the study. The relative fractional enrichment of 13C-4-glutamate labeling was 6.78 +/- 1.71%, whereas 13C-4-glutamine was 5.68 +/- 1.84%. Steady-state modeling of the 13C label distribution in glutamate and glutamine suggests that, under these conditions, the consumption of the beta-hydroxybutyrate is predominantly neuronal, used at a rate of 0.032 +/- 0.009 mmol. kg-1. min-1, and accounts for 6.4 +/- 1.6% of total acetyl coenzyme A oxidation. These results are consistent with minimal accumulation of cerebral ketones with rapid utilization, implying blood-brain barrier control of ketone oxidation in the nonfasted adult human brain.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Brain/metabolism , 3-Hydroxybutyric Acid/administration & dosage , Acetyl Coenzyme A/metabolism , Aspartic Acid/metabolism , Blood-Brain Barrier , Carbon Isotopes , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Ketones/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Neurons/metabolism , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...