Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell ; 160(4): 745-758, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25662011

ABSTRACT

Impaired insulin-mediated suppression of hepatic glucose production (HGP) plays a major role in the pathogenesis of type 2 diabetes (T2D), yet the molecular mechanism by which this occurs remains unknown. Using a novel in vivo metabolomics approach, we show that the major mechanism by which insulin suppresses HGP is through reductions in hepatic acetyl CoA by suppression of lipolysis in white adipose tissue (WAT) leading to reductions in pyruvate carboxylase flux. This mechanism was confirmed in mice and rats with genetic ablation of insulin signaling and mice lacking adipose triglyceride lipase. Insulin's ability to suppress hepatic acetyl CoA, PC activity, and lipolysis was lost in high-fat-fed rats, a phenomenon reversible by IL-6 neutralization and inducible by IL-6 infusion. Taken together, these data identify WAT-derived hepatic acetyl CoA as the main regulator of HGP by insulin and link it to inflammation-induced hepatic insulin resistance associated with obesity and T2D.


Subject(s)
Acetyl Coenzyme A/metabolism , Insulin Resistance , Liver/metabolism , Panniculitis/metabolism , Adipose Tissue, White/chemistry , Adolescent , Animals , Diabetes Mellitus, Type 2 , Diet, High-Fat , Glucose/metabolism , Humans , Hyperglycemia , Interleukin-6/analysis , Lipolysis , Male , Mice , Obesity/metabolism , Rats, Sprague-Dawley
2.
Cell ; 156(1-2): 304-16, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439384

ABSTRACT

A clear relationship exists between visceral obesity and type 2 diabetes, whereas subcutaneous obesity is comparatively benign. Here, we show that adipocyte-specific deletion of the coregulatory protein PRDM16 caused minimal effects on classical brown fat but markedly inhibited beige adipocyte function in subcutaneous fat following cold exposure or ß3-agonist treatment. These animals developed obesity on a high-fat diet, with severe insulin resistance and hepatic steatosis. They also showed altered fat distribution with markedly increased subcutaneous adiposity. Subcutaneous adipose tissue in mutant mice acquired many key properties of visceral fat, including decreased thermogenic and increased inflammatory gene expression and increased macrophage accumulation. Transplantation of subcutaneous fat into mice with diet-induced obesity showed a loss of metabolic benefit when tissues were derived from PRDM16 mutant animals. These findings indicate that PRDM16 and beige adipocytes are required for the "browning" of white fat and the healthful effects of subcutaneous adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue/metabolism , DNA-Binding Proteins/metabolism , Obesity/metabolism , Transcription Factors/metabolism , Adipocytes/metabolism , Animals , DNA-Binding Proteins/genetics , Diet, High-Fat , Insulin Resistance , Mice , Mice, Knockout , Transcription Factors/genetics
3.
Immunity ; 45(3): 583-596, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27566941

ABSTRACT

Mononuclear phagocytes (MNPs) are a highly heterogeneous group of cells that play important roles in maintaining the body's homeostasis. Here, we found CD301b (also known as MGL2), a lectin commonly used as a marker for alternatively activated macrophages, was selectively expressed by a subset of CD11b(+)CD11c(+)MHCII(+) MNPs in multiple organs including adipose tissues. Depleting CD301b(+) MNPs in vivo led to a significant weight loss with increased insulin sensitivity and a marked reduction in serum Resistin-like molecule (RELM) α, a multifunctional cytokine produced by MNPs. Reconstituting RELMα in CD301b(+) MNP-depleted animals restored body weight and normoglycemia. Thus, CD301b(+) MNPs play crucial roles in maintaining glucose metabolism and net energy balance.


Subject(s)
Energy Metabolism/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Lectins, C-Type/metabolism , Phagocytes/metabolism , Adipose Tissue/metabolism , Animals , Female , Glucose , Insulin/metabolism , Insulin Resistance/physiology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL
4.
Nature ; 517(7534): 391-5, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25409143

ABSTRACT

Obesity-linked insulin resistance is a major precursor to the development of type 2 diabetes. Previous work has shown that phosphorylation of PPARγ (peroxisome proliferator-activated receptor γ) at serine 273 by cyclin-dependent kinase 5 (Cdk5) stimulates diabetogenic gene expression in adipose tissues. Inhibition of this modification is a key therapeutic mechanism for anti-diabetic drugs that bind PPARγ, such as the thiazolidinediones and PPARγ partial agonists or non-agonists. For a better understanding of the importance of this obesity-linked PPARγ phosphorylation, we created mice that ablated Cdk5 specifically in adipose tissues. These mice have both a paradoxical increase in PPARγ phosphorylation at serine 273 and worsened insulin resistance. Unbiased proteomic studies show that extracellular signal-regulated kinase (ERK) kinases are activated in these knockout animals. Here we show that ERK directly phosphorylates serine 273 of PPARγ in a robust manner and that Cdk5 suppresses ERKs through direct action on a novel site in MAP kinase/ERK kinase (MEK). Importantly, pharmacological inhibition of MEK and ERK markedly improves insulin resistance in both obese wild-type and ob/ob mice, and also completely reverses the deleterious effects of the Cdk5 ablation. These data show that an ERK/Cdk5 axis controls PPARγ function and suggest that MEK/ERK inhibitors may hold promise for the treatment of type 2 diabetes.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Diabetes Mellitus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , PPAR gamma/metabolism , Adipocytes/enzymology , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Diet, High-Fat , Humans , Insulin Resistance , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Obese , PPAR gamma/chemistry , Phosphorylation
5.
Biochem J ; 475(6): 1063-1074, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29483297

ABSTRACT

Exposure to the toxins methylene cyclopropyl acetic acid (MCPA) and methylene cyclopropyl glycine (MCPG) of unripe ackee and litchi fruit can lead to hypoglycemia and death; however, the molecular mechanisms by which MCPA and MCPG cause hypoglycemia have not been established in vivo To determine the in vivo mechanisms of action of these toxins, we infused them into conscious rodents and assessed rates of hepatic gluconeogenesis and ketogenesis, hepatic acyl-CoA and hepatic acetyl-CoA content, and hepatocellular energy charge. MCPG suppressed rates of hepatic ß-oxidation as reflected by reductions in hepatic ketogenesis, reducing both short- and medium-chain hepatic acyl-CoA concentrations. Hepatic acetyl-CoA content decreased, and hepatic glucose production was inhibited. MCPA also suppressed ß-oxidation of short-chain acyl-CoAs, rapidly inhibiting hepatic ketogenesis and hepatic glucose production, depleting hepatic acetyl-CoA content and ATP content, while increasing other short-chain acyl-CoAs. Utilizing a recently developed positional isotopomer NMR tracer analysis method, we demonstrated that MCPA-induced reductions in hepatic acetyl-CoA content were associated with a marked reduction of hepatic pyruvate carboxylase (PC) flux. Taken together, these data reveal the in vivo mechanisms of action of MCPA and MCPG: the hypoglycemia associated with ingestion of these toxins can be ascribed mostly to MCPA- or MCPG-induced reductions in hepatic PC flux due to inhibition of ß-oxidation of short-chain acyl-CoAs by MCPA or inhibition of both short- and medium-chain acyl-CoAs by MCPG with resultant reductions in hepatic acetyl-CoA content, with an additional contribution to hypoglycemia through reduced hepatic ATP stores by MCPA.


Subject(s)
Cyclopropanes , Glycine/analogs & derivatives , Hypoglycemia/chemically induced , Animals , Gluconeogenesis/drug effects , Glucose/metabolism , Hypoglycemia/pathology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
6.
Proc Natl Acad Sci U S A ; 113(8): 2212-7, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858428

ABSTRACT

Sarcopenia, or skeletal muscle atrophy, is a debilitating comorbidity of many physiological and pathophysiological processes, including normal aging. There are no approved therapies for sarcopenia, but the antihypertrophic myokine myostatin is a potential therapeutic target. Here, we show that treatment of young and old mice with an anti-myostatin antibody (ATA 842) for 4 wk increased muscle mass and muscle strength in both groups. Furthermore, ATA 842 treatment also increased insulin-stimulated whole body glucose metabolism in old mice, which could be attributed to increased insulin-stimulated skeletal muscle glucose uptake as measured by a hyperinsulinemic-euglycemic clamp. Taken together, these studies provide support for pharmacological inhibition of myostatin as a potential therapeutic approach for age-related sarcopenia and metabolic disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Insulin Resistance/physiology , Muscle Strength/physiology , Muscle, Skeletal/pathology , Myostatin/antagonists & inhibitors , Sarcopenia/therapy , Aging/immunology , Aging/pathology , Aging/physiology , Animals , Disease Models, Animal , Energy Metabolism , Humans , Male , Mice , Myostatin/immunology , Myostatin/physiology , Sarcopenia/pathology , Sarcopenia/physiopathology
7.
Diabetologia ; 61(6): 1435-1446, 2018 06.
Article in English | MEDLINE | ID: mdl-29497783

ABSTRACT

AIMS/HYPOTHESIS: Targeting regulators of adipose tissue lipoprotein lipase could enhance adipose lipid clearance, prevent ectopic lipid accumulation and consequently ameliorate insulin resistance and type 2 diabetes. Angiopoietin-like 8 (ANGPTL8) is an insulin-regulated lipoprotein lipase inhibitor strongly expressed in murine adipose tissue. However, Angptl8 knockout mice do not have improved insulin resistance. We hypothesised that pharmacological inhibition, using a second-generation antisense oligonucleotide (ASO) against Angptl8 in adult high-fat-fed rodents, would prevent ectopic lipid accumulation and insulin resistance by promoting adipose lipid uptake. METHODS: ANGPTL8 expression was assessed by quantitative PCR in omental adipose tissue of bariatric surgery patients. High-fat-fed Sprague Dawley rats and C57BL/6 mice were treated with ASO against Angptl8 and insulin sensitivity was assessed by hyperinsulinaemic-euglycaemic clamps in rats and glucose tolerance tests in mice. Factors mediating lipid-induced hepatic insulin resistance were assessed, including lipid content, protein kinase Cε (PKCε) activation and insulin-stimulated Akt phosphorylation. Rat adipose lipid uptake was assessed by mixed meal tolerance tests. Murine energy balance was assessed by indirect calorimetry. RESULTS: Omental fat ANGPTL8 mRNA expression is higher in obese individuals with fatty liver and insulin resistance compared with BMI-matched insulin-sensitive individuals. Angptl8 ASO prevented hepatic steatosis, PKCε activation and hepatic insulin resistance in high-fat-fed rats. Postprandial triacylglycerol uptake in white adipose tissue was increased in Angptl8 ASO-treated rats. Angptl8 ASO protected high-fat-fed mice from glucose intolerance. Although there was no change in net energy balance, Angptl8 ASO increased fat mass in high-fat-fed mice. CONCLUSIONS/INTERPRETATION: Disinhibition of adipose tissue lipoprotein lipase is a novel therapeutic modality to enhance adipose lipid uptake and treat non-alcoholic fatty liver disease and insulin resistance. In line with this, adipose ANGPTL8 is a candidate therapeutic target for these conditions.


Subject(s)
Adipose Tissue/metabolism , Angiopoietin-like Proteins/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Oligonucleotides, Antisense/genetics , Peptide Hormones/genetics , Angiopoietin-Like Protein 8 , Animals , Body Composition , Calorimetry, Indirect , Diet, High-Fat , Glucose Tolerance Test , Insulin Resistance , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
8.
FASEB J ; 31(7): 2916-2924, 2017 07.
Article in English | MEDLINE | ID: mdl-28330852

ABSTRACT

Lipodystrophy is a rare disorder characterized by complete or partial loss of adipose tissue. Patients with lipodystrophy exhibit hypertriglyceridemia, severe insulin resistance, type 2 diabetes, and nonalcoholic steatohepatitis (NASH). Efforts to ameliorate NASH in lipodystrophies with pharmacologic agents have met with limited success. We examined whether a controlled-release mitochondrial protonophore (CRMP) that produces mild liver-targeted mitochondrial uncoupling could decrease hypertriglyceridemia and reverse NASH and diabetes in a mouse model (fatless AZIP/F-1 mice) of severe lipodystrophy and diabetes. After 4 wk of oral CRMP (2 mg/kg body weight per day) or vehicle treatment, mice underwent hyperinsulinemic-euglycemic clamps combined with radiolabeled glucose to assess liver and muscle insulin responsiveness and tissue lipid measurements. CRMP treatment reversed hypertriglyceridemia and insulin resistance in liver and skeletal muscle. Reversal of insulin resistance could be attributed to reductions in diacylglycerol content and reduced PKC-ε and PKC-θ activity in liver and muscle respectively. CRMP treatment also reversed NASH as reflected by reductions in plasma aspartate aminotransferase and alanine aminotransferase concentrations; hepatic steatosis; and hepatic expression of IL-1α, -ß, -2, -4, -6, -10, -12, CD69, and caspase 3 and attenuated activation of the IRE-1α branch of the unfolded protein response. Taken together, these results provide proof of concept for the development of liver-targeted mitochondrial uncoupling agents as a potential novel therapy for lipodystrophy-associated hypertriglyceridemia, NASH and diabetes.-Abulizi, A., Perry, R. J., Camporez, J. P. G., Jurczak, M. J., Petersen, K. F., Aspichueta, P., Shulman, G. I. A controlled-release mitochondrial protonophore reverses hypertriglyceridemia, nonalcoholic steatohepatitis, and diabetes in lipodystrophic mice.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypertriglyceridemia/drug therapy , Lipodystrophy/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Proton Ionophores/therapeutic use , Animals , Delayed-Action Preparations , Eating/drug effects , Energy Metabolism/drug effects , Insulin Resistance , Male , Mice , Mitochondria, Liver/drug effects , Proton Ionophores/administration & dosage , Random Allocation
9.
Proc Natl Acad Sci U S A ; 112(4): 1143-8, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25564660

ABSTRACT

A central paradox in type 2 diabetes is the apparent selective nature of hepatic insulin resistance--wherein insulin fails to suppress hepatic glucose production yet continues to stimulate lipogenesis, resulting in hyperglycemia, hyperlipidemia, and hepatic steatosis. Although efforts to explain this have focused on finding a branch point in insulin signaling where hepatic glucose and lipid metabolism diverge, we hypothesized that hepatic triglyceride synthesis could be driven by substrate, independent of changes in hepatic insulin signaling. We tested this hypothesis in rats by infusing [U-(13)C] palmitate to measure rates of fatty acid esterification into hepatic triglyceride while varying plasma fatty acid and insulin concentrations independently. These experiments were performed in normal rats, high fat-fed insulin-resistant rats, and insulin receptor 2'-O-methoxyethyl chimeric antisense oligonucleotide-treated rats. Rates of fatty acid esterification into hepatic triglyceride were found to be dependent on plasma fatty acid infusion rates, independent of changes in plasma insulin concentrations and independent of hepatocellular insulin signaling. Taken together, these results obviate a paradox of selective insulin resistance, because the major source of hepatic lipid synthesis, esterification of preformed fatty acids, is primarily dependent on substrate delivery and largely independent of hepatic insulin action.


Subject(s)
Insulin Resistance , Insulin/metabolism , Liver/metabolism , Palmitic Acid/metabolism , Signal Transduction , Triglycerides/biosynthesis , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Palmitic Acid/pharmacology , Rats , Receptor, Insulin/metabolism
10.
Proc Natl Acad Sci U S A ; 111(46): 16508-13, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368185

ABSTRACT

The pyruvate dehydrogenase complex (PDH) has been hypothesized to link lipid exposure to skeletal muscle insulin resistance through a glucose-fatty acid cycle in which increased fatty acid oxidation increases acetyl-CoA concentrations, thereby inactivating PDH and decreasing glucose oxidation. However, whether fatty acids induce insulin resistance by decreasing PDH flux remains unknown. To genetically examine this hypothesis we assessed relative rates of pyruvate dehydrogenase flux/mitochondrial oxidative flux and insulin-stimulated rates of muscle glucose metabolism in awake mice lacking pyruvate dehydrogenase kinase 2 and 4 [double knockout (DKO)], which results in constitutively activated PDH. Surprisingly, increased glucose oxidation in DKO muscle was accompanied by reduced insulin-stimulated muscle glucose uptake. Preferential myocellular glucose utilization in DKO mice decreased fatty acid oxidation, resulting in increased reesterification of acyl-CoAs into diacylglycerol and triacylglycerol, with subsequent activation of PKC-θ and inhibition of insulin signaling in muscle. In contrast, other putative mediators of muscle insulin resistance, including muscle acylcarnitines, ceramides, reactive oxygen species production, and oxidative stress markers, were not increased. These findings demonstrate that modulation of oxidative substrate selection to increase muscle glucose utilization surprisingly results in muscle insulin resistance, offering genetic evidence against the glucose-fatty acid cycle hypothesis of muscle insulin resistance.


Subject(s)
Insulin Resistance/physiology , Protein Serine-Threonine Kinases/deficiency , Pyruvate Dehydrogenase Complex/metabolism , Animals , Carnitine/analogs & derivatives , Carnitine/metabolism , Citric Acid Cycle , Dietary Fats/pharmacology , Dietary Fats/toxicity , Enzyme Activation , Fatty Acids/metabolism , Glucose/metabolism , Glycogen/metabolism , Hyperinsulinism/metabolism , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Muscle, Skeletal/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C-theta , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/genetics , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Substrate Specificity
11.
Am J Physiol Endocrinol Metab ; 311(1): E105-16, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27166280

ABSTRACT

Mitochondrial dysfunction is associated with many human diseases and results from mismatch of damage and repair over the life of the organelle. PARK2 is a ubiquitin E3 ligase that regulates mitophagy, a repair mechanism that selectively degrades damaged mitochondria. Deletion of PARK2 in multiple in vivo models results in susceptibility to stress-induced mitochondrial and cellular dysfunction. Surprisingly, Park2 knockout (KO) mice are protected from nutritional stress and do not develop obesity, hepatic steatosis or insulin resistance when fed a high-fat diet (HFD). However, these phenomena are casually related and the physiological basis for this phenotype is unknown. We therefore undertook a series of acute HFD studies to more completely understand the physiology of Park2 KO during nutritional stress. We find that intestinal lipid absorption is impaired in Park2 KO mice as evidenced by increased fecal lipids and reduced plasma triglycerides after intragastric fat challenge. Park2 KO mice developed hepatic steatosis in response to intravenous lipid infusion as well as during incubation of primary hepatocytes with fatty acids, suggesting that hepatic protection from nutritional stress was secondary to changes in energy balance due to altered intestinal triglyceride absorption. Park2 KO mice showed reduced adiposity after 1-wk HFD, as well as improved hepatic and peripheral insulin sensitivity. These studies suggest that changes in intestinal lipid absorption may play a primary role in protection from nutritional stress in Park2 KO mice by preventing HFD-induced weight gain and highlight the need for tissue-specific models to address the role of PARK2 during metabolic stress.


Subject(s)
Body Weight/genetics , Diet, High-Fat , Insulin Resistance/genetics , Intestinal Absorption/genetics , Lipid Metabolism/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Energy Metabolism , Fatty Acids/pharmacology , Fatty Liver/genetics , Feces/chemistry , Infusions, Intravenous , Intestinal Mucosa/metabolism , Lipids/analysis , Lipids/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Mitophagy/genetics , Triglycerides/blood , Weight Gain/genetics
12.
Proc Natl Acad Sci U S A ; 110(31): 12780-5, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23840067

ABSTRACT

Hepatic insulin resistance is a principal component of type 2 diabetes, but the cellular and molecular mechanisms responsible for its pathogenesis remain unknown. Recent studies have suggested that saturated fatty acids induce hepatic insulin resistance through activation of the toll-like receptor 4 (TLR-4) receptor in the liver, which in turn transcriptionally activates hepatic ceramide synthesis leading to inhibition of insulin signaling. In this study, we demonstrate that TLR-4 receptor signaling is not directly required for saturated or unsaturated fat-induced hepatic insulin resistance in both TLR-4 antisense oligonucleotide treated and TLR-4 knockout mice, and that ceramide accumulation is not dependent on TLR-4 signaling or a primary event in hepatic steatosis and impairment of insulin signaling. Further, we show that both saturated and unsaturated fats lead to hepatic accumulation of diacylglycerols, activation of PKCε, and impairment of insulin-stimulated IRS-2 signaling. These data demonstrate that saturated fat-induced insulin resistance is independent of TLR-4 activation and ceramides.


Subject(s)
Dietary Fats, Unsaturated/pharmacology , Fatty Liver/metabolism , Insulin Resistance , Liver/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diglycerides/metabolism , Fatty Liver/chemically induced , Fatty Liver/pathology , Insulin Receptor Substrate Proteins/metabolism , Liver/pathology , Mice , Rats , Rats, Sprague-Dawley
13.
Proc Natl Acad Sci U S A ; 110(5): 1869-74, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23302688

ABSTRACT

Comparative gene identification 58 (CGI-58) is a lipid droplet-associated protein that promotes the hydrolysis of triglyceride by activating adipose triglyceride lipase. Loss-of-function mutations in CGI-58 in humans lead to Chanarin-Dorfman syndrome, a condition in which triglyceride accumulates in various tissues, including the skin, liver, muscle, and intestines. Therefore, without adequate CGI-58 expression, lipids are stored rather than used for fuel, signaling intermediates, and membrane biosynthesis. CGI-58 knockdown in mice using antisense oligonucleotide (ASO) treatment also leads to severe hepatic steatosis as well as increased hepatocellular diacylglycerol (DAG) content, a well-documented trigger of insulin resistance. Surprisingly, CGI-58 knockdown mice remain insulin-sensitive, seemingly dissociating DAG from the development of insulin resistance. Therefore, we sought to determine the mechanism responsible for this paradox. Hyperinsulinemic-euglycemic clamp studies reveal that the maintenance of insulin sensitivity with CGI-58 ASO treatment could entirely be attributed to protection from lipid-induced hepatic insulin resistance, despite the apparent lipotoxic conditions. Analysis of the cellular compartmentation of DAG revealed that DAG increased in the membrane fraction of high fat-fed mice, leading to PKCε activation and hepatic insulin resistance. However, DAG increased in lipid droplets or lipid-associated endoplasmic reticulum rather than the membrane of CGI-58 ASO-treated mice, and thus prevented PKCε translocation to the plasma membrane and induction of insulin resistance. Taken together, these results explain the disassociation of hepatic steatosis and DAG accumulation from hepatic insulin resistance in CGI-58 ASO-treated mice, and highlight the importance of intracellular compartmentation of DAG in causing lipotoxicity and hepatic insulin resistance.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Diglycerides/metabolism , Endoplasmic Reticulum/metabolism , Insulin Resistance , Lipids/chemistry , Liver/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Diet, High-Fat , Endoplasmic Reticulum/drug effects , Gene Expression/drug effects , Gene Knockdown Techniques , Humans , Immunoblotting , Injections, Intraperitoneal , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Protein Kinase C-epsilon/metabolism , Protein Transport/drug effects , Reverse Transcriptase Polymerase Chain Reaction
14.
J Lipid Res ; 56(3): 526-536, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25548259

ABSTRACT

ApoA5 has a critical role in the regulation of plasma TG concentrations. In order to determine whether ApoA5 also impacts ectopic lipid deposition in liver and skeletal muscle, as well as tissue insulin sensitivity, we treated mice with an antisense oligonucleotide (ASO) to decrease hepatic expression of ApoA5. ASO treatment reduced ApoA5 protein expression in liver by 60-70%. ApoA5 ASO-treated mice displayed approximately 3-fold higher plasma TG concentrations, which were associated with decreased plasma TG clearance. Furthermore, ApoA5 ASO-treated mice fed a high-fat diet (HFD) exhibited reduced liver and skeletal muscle TG uptake and reduced liver and muscle TG and diacylglycerol (DAG) content. HFD-fed ApoA5 ASO-treated mice were protected from HFD-induced insulin resistance, as assessed by hyperinsulinemic-euglycemic clamps. This protection could be attributed to increases in both hepatic and peripheral insulin responsiveness associated with decreased DAG activation of protein kinase C (PKC)-ε and PKCθ in liver and muscle, respectively, and increased insulin-stimulated AKT2 pho-sphory-lation in these tissues. In summary, these studies demonstrate a novel role for ApoA5 as a modulator of susceptibility to diet-induced liver and muscle insulin resistance through regulation of ectopic lipid accumulation in liver and skeletal muscle.


Subject(s)
Apolipoproteins/metabolism , Dietary Fats/pharmacology , Insulin Resistance , Liver/metabolism , Muscle, Skeletal/metabolism , Triglycerides/metabolism , Animals , Apolipoprotein A-V , Apolipoproteins/genetics , Gene Knockdown Techniques , Male , Mice , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Triglycerides/genetics
15.
Proc Natl Acad Sci U S A ; 109(37): 14966-71, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22912404

ABSTRACT

Fatty acid amide hydrolase (FAAH) knockout mice are prone to excess energy storage and adiposity, whereas mutations in FAAH are associated with obesity in humans. However, the molecular mechanism by which FAAH affects energy expenditure (EE) remains unknown. Here we show that reduced energy expenditure in FAAH(-/-) mice could be attributed to decreased circulating triiodothyronine and thyroxine concentrations secondary to reduced mRNA expression of both pituitary thyroid-stimulating hormone and hypothalamic thyrotropin-releasing hormone. These reductions in the hypothalamic-pituitary-thyroid axis were associated with activation of hypothalamic peroxisome proliferating-activated receptor γ (PPARγ), and increased hypothalamic deiodinase 2 expression. Infusion of NAEs (anandamide and palmitoylethanolamide) recapitulated increases in PPARγ-mediated decreases in EE. FAAH(-/-) mice were also prone to diet-induced hepatic insulin resistance, which could be attributed to increased hepatic diacylglycerol content and protein kinase Cε activation. Our data indicate that FAAH deletion, and the resulting increases in NAEs, predispose mice to ectopic lipid storage and hepatic insulin resistance by promoting centrally mediated hypothyroidism.


Subject(s)
Amidohydrolases/genetics , Energy Metabolism/physiology , Hypothyroidism/complications , Hypothyroidism/genetics , Insulin Resistance/physiology , Amides , Amidohydrolases/deficiency , Analysis of Variance , Animals , Arachidonic Acids/administration & dosage , Chromatography, Liquid , Endocannabinoids/administration & dosage , Energy Metabolism/genetics , Ethanolamines/administration & dosage , Hypothyroidism/enzymology , Immunoblotting , Mice , Mice, Knockout , PPAR gamma , Palmitic Acids/administration & dosage , Polymerase Chain Reaction , Polyunsaturated Alkamides/administration & dosage , Tandem Mass Spectrometry , Thyrotropin/metabolism , Thyrotropin-Releasing Hormone/metabolism , Thyroxine/blood , Triiodothyronine/blood
16.
Diabetologia ; 57(6): 1232-41, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24718953

ABSTRACT

AIMS/HYPOTHESIS: Aerobic exercise increases muscle glucose and improves insulin action through numerous pathways, including activation of Ca(2+)/calmodulin-dependent protein kinases (CAMKs) and peroxisome proliferator γ coactivator 1α (PGC-1α). While overexpression of PGC-1α increases muscle mitochondrial content and oxidative type I fibres, it does not improve insulin action. Activation of CAMK4 also increases the content of type I muscle fibres, PGC-1α level and mitochondrial content. However, it remains unknown whether CAMK4 activation improves insulin action on glucose metabolism in vivo. METHODS: The effects of CAMK4 activation on skeletal muscle insulin action were quantified using transgenic mice with a truncated and constitutively active form of CAMK4 (CAMK4([Symbol: see text])) in skeletal muscle. Tissue-specific insulin sensitivity was assessed in vivo using a hyperinsulinaemic-euglycaemic clamp and isotopic measurements of glucose metabolism. RESULTS: The rate of insulin-stimulated whole-body glucose uptake was increased by ∼25% in CAMK4([Symbol: see text]) mice. This was largely attributed to an increase of ∼60% in insulin-stimulated glucose uptake in the quadriceps, the largest hindlimb muscle. These changes were associated with improvements in insulin signalling, as reflected by increased phosphorylation of Akt and its substrates and an increase in the level of GLUT4 protein. In addition, there were extramuscular effects: CAMK4([Symbol: see text]) mice had improved hepatic and adipose insulin action. These pleiotropic effects were associated with increased levels of PGC-1α-related myokines in CAMK4([Symbol: see text]) skeletal muscle. CONCLUSIONS/INTERPRETATION: Activation of CAMK4 enhances mitochondrial biogenesis in skeletal muscle while also coordinating improvements in whole-body insulin-mediated glucose.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Glucose/metabolism , Insulin/metabolism , Muscle, Skeletal/enzymology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Female , Male , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics
18.
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.

19.
Diabetes ; 67(4): 624-635, 2018 04.
Article in English | MEDLINE | ID: mdl-29317435

ABSTRACT

Stress responses promote obesity and insulin resistance, in part, by activating the stress-responsive mitogen-activated protein kinases (MAPKs), p38 MAPK, and c-Jun NH2-terminal kinase (JNK). Stress also induces expression of MAPK phosphatase-1 (MKP-1), which inactivates both JNK and p38 MAPK. However, the equilibrium between JNK/p38 MAPK and MKP-1 signaling in the development of obesity and insulin resistance is unclear. Skeletal muscle is a major tissue involved in energy expenditure and glucose metabolism. In skeletal muscle, MKP-1 is upregulated in high-fat diet-fed mice and in skeletal muscle of obese humans. Mice lacking skeletal muscle expression of MKP-1 (MKP1-MKO) showed increased skeletal muscle p38 MAPK and JNK activities and were resistant to the development of diet-induced obesity. MKP1-MKO mice exhibited increased whole-body energy expenditure that was associated with elevated levels of myofiber-associated mitochondrial oxygen consumption. miR-21, a negative regulator of PTEN expression, was upregulated in skeletal muscle of MKP1-MKO mice, resulting in increased Akt activity consistent with enhanced insulin sensitivity. Our results demonstrate that skeletal muscle MKP-1 represents a critical signaling node through which inactivation of the p38 MAPK/JNK module promotes obesity and insulin resistance.


Subject(s)
Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Insulin Resistance , MAP Kinase Kinase 4/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Diet, High-Fat , Energy Metabolism , Humans , Mice , Mice, Knockout , MicroRNAs/metabolism , Mitochondria, Muscle/metabolism , Oxygen Consumption , Signal Transduction
20.
Diabetes ; 67(2): 208-221, 2018 02.
Article in English | MEDLINE | ID: mdl-29203511

ABSTRACT

Disruption of hepatocyte growth hormone (GH) signaling through disruption of Jak2 (JAK2L) leads to fatty liver. Previously, we demonstrated that development of fatty liver depends on adipocyte GH signaling. We sought to determine the individual roles of hepatocyte and adipocyte Jak2 on whole-body and tissue insulin sensitivity and liver metabolism. On chow, JAK2L mice had hepatic steatosis and severe whole-body and hepatic insulin resistance. However, concomitant deletion of Jak2 in hepatocytes and adipocytes (JAK2LA) completely normalized insulin sensitivity while reducing liver lipid content. On high-fat diet, JAK2L mice had hepatic steatosis and insulin resistance despite protection from diet-induced obesity. JAK2LA mice had higher liver lipid content and no protection from obesity but retained exquisite hepatic insulin sensitivity. AKT activity was selectively attenuated in JAK2L adipose tissue, whereas hepatic insulin signaling remained intact despite profound hepatic insulin resistance. Therefore, JAK2 in adipose tissue is epistatic to liver with regard to insulin sensitivity and responsiveness, despite fatty liver and obesity. However, hepatocyte autonomous JAK2 signaling regulates liver lipid deposition under conditions of excess dietary fat. This work demonstrates how various tissues integrate JAK2 signals to regulate insulin/glucose and lipid metabolism.


Subject(s)
Adipose Tissue/enzymology , Insulin Resistance , Janus Kinase 2/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Obesity/metabolism , Adipose Tissue/metabolism , Adiposity , Animals , Diet, High-Fat/adverse effects , Janus Kinase 2/genetics , Lipid Metabolism , Liver/enzymology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/etiology , Obesity/physiopathology , Organ Specificity , Phosphoproteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Threonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL