Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Arch Biochem Biophys ; 623-624: 49-57, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28526426

ABSTRACT

AMP-activated protein kinase (AMPK) is an enzyme crucial in cellular metabolism found to be inhibited in many metabolic diseases including type 2 diabetes. Thiazolidinediones (TZDs) are a class of anti-diabetic drug known to activate AMPK through increased phosphorylation at Thr172, however there has been no research to date on whether they have any effect on inhibition of AMPK's lesser known site of inhibition, Ser485/491. HepG2 cells were treated with troglitazone and phosphorylation of AMPK was found to increase at both Thr172 and Ser485 in a dose- and time-dependent manner. Treatment of HepG2 cells with insulin and PMA led to increases in p-AMPK Ser485 via Akt and PKD1 respectively; however these kinases were not found to be implicated in increases seen from troglitazone. Incubation with the other TZDs, rosiglitazone and pioglitazone, let to a minor increase in p-AMPK Ser485 phosphorylation as well as AMPK activity; however these findings were significantly less than those of troglitazone under equal conditions. These data suggest that the effects of troglitazone on AMPK are more complex than previously thought. Phosphorylation at sites of both activation and inhibition can occur in tandem, although the mechanism by which this occurs has not yet been elucidated.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chromans/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Enzyme Activation/drug effects , Hypoglycemic Agents/pharmacology , Thiazolidinediones/pharmacology , Diabetes Mellitus, Type 2/metabolism , Hep G2 Cells , Humans , Insulin/metabolism , Phosphorylation/drug effects , Pioglitazone , Rosiglitazone , Troglitazone
2.
Diabetologia ; 59(12): 2654-2663, 2016 12.
Article in English | MEDLINE | ID: mdl-27677764

ABSTRACT

AIMS/HYPOTHESIS: To directly assess the role of beta cell lipolysis in insulin secretion and whole-body energy homeostasis, inducible beta cell-specific adipose triglyceride lipase (ATGL)-deficient (B-Atgl-KO) mice were studied under normal diet (ND) and high-fat diet (HFD) conditions. METHODS: Atgl flox/flox mice were cross-bred with Mip-Cre-ERT mice to generate Mip-Cre-ERT/+;Atgl flox/flox mice. At 8 weeks of age, these mice were injected with tamoxifen to induce deletion of beta cell-specific Atgl (also known as Pnpla2), and the mice were fed an ND or HFD. RESULTS: ND-fed male B-Atgl-KO mice showed decreased insulinaemia and glucose-induced insulin secretion (GSIS) in vivo. Changes in GSIS correlated with the islet content of long-chain saturated monoacylglycerol (MAG) species that have been proposed to be metabolic coupling factors for insulin secretion. Exogenous MAGs restored GSIS in B-Atgl-KO islets. B-Atgl-KO male mice fed an HFD showed reduced insulinaemia, glycaemia in the fasted and fed states and after glucose challenge, as well as enhanced insulin sensitivity. Moreover, decreased insulinaemia in B-Atgl-KO mice was associated with increased energy expenditure, and lipid metabolism in brown (BAT) and white (WAT) adipose tissues, leading to reduced fat mass and body weight. CONCLUSIONS/INTERPRETATION: ATGL in beta cells regulates insulin secretion via the production of signalling MAGs. Decreased insulinaemia due to lowered GSIS protects B-Atgl-KO mice from diet-induced obesity, improves insulin sensitivity, increases lipid mobilisation from WAT and causes BAT activation. The results support the concept that fuel excess can drive obesity and diabetes via hyperinsulinaemia, and that an islet beta cell ATGL-lipolysis/adipose tissue axis controls energy homeostasis and body weight via insulin secretion.


Subject(s)
Adipose Tissue/metabolism , Body Weight/physiology , Energy Metabolism/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Adipose Tissue/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Calcium/metabolism , Diet, High-Fat/adverse effects , Female , Homeostasis/drug effects , Homeostasis/physiology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Lipase/metabolism , Lipid Metabolism/drug effects , Lipolysis/drug effects , Lipolysis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology , Tandem Mass Spectrometry
3.
Biosci Rep ; 36(5)2016 10.
Article in English | MEDLINE | ID: mdl-27534430

ABSTRACT

High concentrations of glucose and palmitate increase endothelial cell inflammation and apoptosis, events that often precede atherogenesis. They may do so by decreasing basal autophagy and AMP-activated protein kinase (AMPK) activity, although the mechanisms by which this occurs are not clear. Decreased function of the lysosome, an organelle required for autophagy and AMPK, have been associated with hyperactivity of glycogen synthase kinase 3ß (GSK3ß). To determine whether GSK3ß affects nutrient-induced changes in autophagy and AMPK activity, we used a primary human aortic endothelial cell (HAEC) model of type 2 diabetes that we had previously characterized with impaired AMPK activity and autophagy [Weikel et al. (2015) Am. J. Phys. Cell Physiol. 308: , C249-C263]. Presently, we found that incubation of HAECs with excess nutrients (25 mM glucose and 0.4 mM palmitate) increased GSK3ß activity and impaired lysosome acidification. Suppression of GSK3ß in these cells by treatment with a chemical inhibitor or overexpression of kinase-dead GSK3ß attenuated these lysosomal changes. Under control and excess nutrient conditions, knockdown of GSK3ß increased autophagosome formation, forkhead box protein O1 (FOXO1) activity and AMPK signalling and decreased Akt signalling. Similar changes in autophagy, AMPK and Akt signalling were observed in aortas from mice treated with the GSK3ß inhibitor CHIR 99021. Thus, increasing basal autophagy and AMPK activity by inhibiting GSK3ß may be an effective strategy in the setting of hyperglycaemia and dyslipidaemia for restoring endothelial cell health and reducing atherogenesis.


Subject(s)
Aorta/metabolism , Diabetes Mellitus, Type 2/genetics , Glycogen Synthase Kinase 3 beta/genetics , Hyperglycemia/genetics , Protein Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Aorta/pathology , Apoptosis/genetics , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Autophagy/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Knockdown Techniques , Glucose/metabolism , Glucose/pharmacology , Glycogen Synthase Kinase 3 beta/biosynthesis , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hyperglycemia/pathology , Mice , Palmitates/metabolism , Palmitates/pharmacology , Phosphorylation , Primary Cell Culture , Protein Kinases/biosynthesis , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction
4.
PLoS One ; 11(4): e0153017, 2016.
Article in English | MEDLINE | ID: mdl-27043434

ABSTRACT

Diet induced obese (DIO) mice can be stratified according to their weight gain in response to high fat diet as low responders (LDR) and high responders (HDR). This allows the study of ß-cell failure and the transitions to prediabetes (LDR) and early diabetes (HDR). C57BL/6N mice were fed for 8 weeks with a normal chow diet (ND) or a high fat diet and stratified as LDR and HDR. Freshly isolated islets from ND, LDR and HDR mice were studied ex-vivo for mitochondrial metabolism, AMPK activity and signalling, the expression and activity of key enzymes of energy metabolism, cholesterol synthesis, and mRNA profiling. Severely compromised glucose-induced insulin secretion in HDR islets, as compared to ND and LDR islets, was associated with suppressed AMP-kinase activity. HDR islets also showed reduced acetyl-CoA carboxylase activity and enhanced activity of 3-hydroxy-3-methylglutaryl-CoA reductase, which led respectively to elevated fatty acid oxidation and increased cholesterol biosynthesis. HDR islets also displayed mitochondrial membrane hyperpolarization and reduced ATP turnover in the presence of elevated glucose. Expression of protein kinase Cε, which reduces both lipolysis and production of signals for insulin secretion, was elevated in DIO islets. Genes whose expression increased or decreased by more than 1.2-fold were minor between LDR and ND islets (17 differentially expressed), but were prominent between HDR and ND islets (1508 differentially expressed). In HDR islets, particularly affected genes were related to cell cycle and proliferation, AMPK signaling, mitochondrial metabolism and cholesterol metabolism. In conclusion, chronically reduced AMPK activity, mitochondrial dysfunction, elevated cholesterol biosynthesis in islets, and substantial alterations in gene expression accompany ß-cell failure in HDR islets. The ß-cell compensation process in the prediabetic state (LDR) is largely independent of transcriptional adaptive changes, whereas the transition to early diabetes (HDR) is associated with major alterations in gene expression.


Subject(s)
Diet/adverse effects , Insulin-Secreting Cells/metabolism , Obesity/etiology , Obesity/metabolism , Adenylate Kinase/metabolism , Animals , Cells, Cultured , Cholesterol/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Insulin/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oxygen Consumption , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Transcriptome
5.
Metabolism ; 65(5): 634-645, 2016 May.
Article in English | MEDLINE | ID: mdl-27085772

ABSTRACT

AMP-activated protein kinase (AMPK) plays a critical role both in sensing and regulating cellular energy state. In experimental animals, its activation has been shown to reduce the risk of obesity and diabetes-related co-morbidities such as insulin resistance, the metabolic syndrome and atherosclerotic cardiovascular disease. However, in humans, AMPK activation alone often does not completely resolve these conditions. Thus, an improved understanding of AMPK action and regulation in metabolic and other diseases is needed. Herein, we provide a brief description of the enzymatic regulation of AMPK and review its role in maintaining energy homeostasis. We then discuss tissue-specific actions of AMPK that become distorted during such conditions as obesity, type 2 diabetes and certain cancers. Finally, we explore recent findings regarding the interactions of AMPK with mammalian target of rapamycin complex 1 and the lysosome and discuss how changes in these relationships during overnutrition may lead to AMPK dysfunction. A more thorough understanding of AMPK's molecular interactions during diseases of overnutrition may provide key insights for the development of AMPK-based combinatorial treatments for metabolic disease.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Glucose Metabolism Disorders/enzymology , Insulin Resistance , Models, Biological , Neoplasms/enzymology , Obesity/enzymology , AMP-Activated Protein Kinases/chemistry , Animals , Energy Intake , Glucose Metabolism Disorders/metabolism , Humans , Lysosomes/enzymology , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Obesity/metabolism , Organ Specificity , TOR Serine-Threonine Kinases/metabolism
6.
J Biol Chem ; 291(11): 5664-5675, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26797128

ABSTRACT

AMP-activated protein kinase (AMPK) is an energy-sensing enzyme whose activity is inhibited in settings of insulin resistance. Exposure to a high glucose concentration has recently been shown to increase phosphorylation of AMPK at Ser(485/491) of its α1/α2 subunit; however, the mechanism by which it does so is not known. Diacylglycerol (DAG), which is also increased in muscle exposed to high glucose, activates a number of signaling molecules including protein kinase (PK)C and PKD1. We sought to determine whether PKC or PKD1 is involved in inhibition of AMPK by causing Ser(485/491) phosphorylation in skeletal muscle cells. C2C12 myotubes were treated with the PKC/D1 activator phorbol 12-myristate 13-acetate (PMA), which acts as a DAG mimetic. This caused dose- and time-dependent increases in AMPK Ser(485/491) phosphorylation, which was associated with a ∼60% decrease in AMPKα2 activity. Expression of a phosphodefective AMPKα2 mutant (S491A) prevented the PMA-induced reduction in AMPK activity. Serine phosphorylation and inhibition of AMPK activity were partially prevented by the broad PKC inhibitor Gö6983 and fully prevented by the specific PKD1 inhibitor CRT0066101. Genetic knockdown of PKD1 also prevented Ser(485/491) phosphorylation of AMPK. Inhibition of previously identified kinases that phosphorylate AMPK at this site (Akt, S6K, and ERK) did not prevent these events. PMA treatment also caused impairments in insulin-signaling through Akt, which were prevented by PKD1 inhibition. Finally, recombinant PKD1 phosphorylated AMPKα2 at Ser(491) in cell-free conditions. These results identify PKD1 as a novel upstream kinase of AMPKα2 Ser(491) that plays a negative role in insulin signaling in muscle cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Insulin/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Protein Kinase C/metabolism , Signal Transduction , Animals , Cell Line , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Phosphorylation , Serine/metabolism
8.
PLoS One ; 10(5): e0127388, 2015.
Article in English | MEDLINE | ID: mdl-25996822

ABSTRACT

We have previously shown that incubation for 1h with excess glucose or leucine causes insulin resistance in rat extensor digitorum longus (EDL) muscle by inhibiting AMP-activated protein kinase (AMPK). To examine the events that precede and follow these changes, studies were performed in rat EDL incubated with elevated levels of glucose or leucine for 30min-2h. Incubation in high glucose (25mM) or leucine (100µM) significantly diminished AMPK activity by 50% within 30min, with further decreases occurring at 1 and 2h. The initial decrease in activity at 30min coincided with a significant increase in muscle glycogen. The subsequent decreases at 1h were accompanied by phosphorylation of αAMPK at Ser485/491, and at 2h by decreased SIRT1 expression and increased PP2A activity, all of which have previously been shown to diminish AMPK activity. Glucose infusion in vivo, which caused several fold increases in plasma glucose and insulin, produced similar changes but with different timing. Thus, the initial decrease in AMPK activity observed at 3h was associated with changes in Ser485/491 phosphorylation and SIRT1 expression and increased PP2A activity was a later event. These findings suggest that both ex vivo and in vivo, multiple factors contribute to fuel-induced decreases in AMPK activity in skeletal muscle and the insulin resistance that accompanies it.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Animal Nutritional Physiological Phenomena , Glucose/metabolism , Muscle, Skeletal/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Gene Expression , Glucose/administration & dosage , Glycogen/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidation-Reduction , Phosphorylation , Rats , Sirtuin 1/genetics , Sirtuin 1/metabolism
9.
PLoS One ; 10(3): e0121959, 2015.
Article in English | MEDLINE | ID: mdl-25798922

ABSTRACT

SIRT1 is a NAD+-dependent deacetylase thought to regulate cellular metabolic pathways in response to alterations in nutrient flux. In the current study we investigated whether acute changes in SIRT1 expression affect markers of muscle mitochondrial content and also determined whether SIRT1 influenced muscle insulin resistance induced by acute glucose oversupply. In male Wistar rats either SIRT1 or a deacetylase inactive mutant form (H363Y) was electroprated into the tibialis cranialis (TC) muscle. The other leg was electroporated with an empty control vector. One week later, glucose was infused and hyperglycaemia was maintained at ~11mM. After 5 hours, 11mM glucose induced significant insulin resistance in skeletal muscle. Interestingly, overexpression of either SIRT1 or SIRT1 (H363Y) for 1 week did not change markers of mitochondrial content or function. SIRT1 or SIRT1 (H363Y) overexpression had no effect on the reduction in glucose uptake and glycogen synthesis in muscle in response to hyperglycemia. Therefore we conclude that acute increases in SIRT1 protein have little impact on mitochondrial content and that overexpressing SIRT1 does not prevent the development of insulin resistance during hyperglycaemia.


Subject(s)
Glucose/pharmacology , Insulin Resistance , Muscle, Skeletal/metabolism , Sirtuin 1/physiology , Animals , Blood Glucose/metabolism , Cell Line , Electroporation , Hyperglycemia/metabolism , Male , Mice , Muscle, Skeletal/drug effects , Rats , Rats, Wistar , Signal Transduction
10.
PLoS One ; 10(2): e0115341, 2015.
Article in English | MEDLINE | ID: mdl-25647160

ABSTRACT

The aging process is perceived as resulting from a combination of intrinsic factors such as changes in intracellular signaling and extrinsic factors, most notably environmental stressors. In skin, the relationship between intrinsic changes and keratinocyte function is not clearly understood. Previously, we found that increasing the activity of AMP-activated protein kinase (AMPK) suppressed senescence in hydrogen peroxide (H2O2)-treated human primary keratinocytes, a model of oxidative stress-induced cellular aging. Using this model in the present study, we observed that resveratrol, an agent that increases the activities of both AMPK and sirtuins, ameliorated two age-associated phenotypes: cellular senescence and proliferative dysfunction. In addition, we found that treatment of keratinocytes with Ex527, a specific inhibitor of sirtuin 1 (SIRT1), attenuated the ability of resveratrol to suppress senescence. In keeping with the latter observation, we noted that compared to non-senescent keratinocytes, senescent cells lacked SIRT1. In addition to these effects on H2O2-induced senescence, resveratrol also prevented the H2O2-induced decrease in proliferation (as indicated by 3H-thymidine incorporation) in the presence of insulin. This effect was abrogated by inhibition of AMPK but not SIRT1. Compared to endothelium, we found that human keratinocytes expressed relatively high levels of Forkhead box O3 (FOXO3), a downstream target of both AMPK and SIRT1. Treatment of keratinocytes with resveratrol transactivated FOXO3 and increased the expression of its target genes including catalase. Resveratrol's effects on both senescence and proliferation disappeared when FOXO3 was knocked down. Finally, we performed an exploratory study which showed that skin from humans over 50 years old had lower AMPK activity than skin from individuals under age 20. Collectively, these findings suggest that the effects of resveratrol on keratinocyte senescence and proliferation are regulated by the AMPK-FOXO3 pathway and in some situations, but not all, by SIRT1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cellular Senescence/drug effects , Forkhead Transcription Factors/metabolism , Keratinocytes/cytology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Stilbenes/pharmacology , Adult , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Forkhead Box Protein O3 , Humans , Hydrogen Peroxide/pharmacology , Infant , Keratinocytes/drug effects , Keratinocytes/metabolism , Middle Aged , Resveratrol , Sirtuin 1/metabolism
11.
Diabetes ; 64(3): 673-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25713189

ABSTRACT

Stratifying the management of type 2 diabetes (T2D) has to take into account marked variability in patient phenotype due to heterogeneity in its pathophysiology, different stages of the disease process, and multiple other patient factors including comorbidities. The focus here is on the very challenging subgroup of patients with T2D who are overweight or obese with insulin resistance (IR) and the most refractory hyperglycemia due to an inability to change lifestyle to reverse positive energy balance. For this subgroup of patients with T2D, we question the dogma that IR is primarily harmful to the body and should be counteracted at any cost. Instead we propose that IR, particularly in this high-risk subgroup, is a defense mechanism that protects critical tissues of the cardiovascular system from nutrient-induced injury. Overriding IR in an effort to lower plasma glucose levels, particularly with intensive insulin therapy, could therefore be harmful. Treatments that nutrient off-load to lower glucose are more likely to be beneficial. The concepts of "IR as an adaptive defense mechanism" and "insulin-induced metabolic stress" may provide explanation for some of the unexpected outcomes of recent major clinical trials in T2D. Potential molecular mechanisms underlying these concepts; their clinical implications for stratification of T2D management, particularly in overweight and obese patients with difficult glycemic control; and future research requirements are discussed.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Stress, Physiological/drug effects , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Insulin/adverse effects , Insulin/therapeutic use
12.
Am J Physiol Cell Physiol ; 308(3): C249-63, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25354528

ABSTRACT

Dysregulated autophagy and decreased AMP-activated protein kinase (AMPK) activity are each associated with atherogenesis. Atherogenesis is preceded by high circulating concentrations of glucose and fatty acids, yet the mechanism by which these nutrients regulate autophagy in human aortic endothelial cells (HAECs) is not known. Furthermore, whereas AMPK is recognized as an activator of autophagy in cells with few nutrients, its effects on autophagy in nutrient-rich HAECs has not been investigated. We maintained and passaged primary HAECs in media containing 25 mM glucose and incubated them subsequently with 0.4 mM palmitate. These conditions impaired basal autophagy and rendered HAECs more susceptible to apoptosis and adhesion of monocytes, outcomes attenuated by the autophagy activator rapamycin. Glucose and palmitate diminished AMPK activity and phosphorylation of the uncoordinated-51-like kinase 1 (ULK1) at Ser555, an autophagy-activating site targeted by AMPK. 5-Aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR)-mediated activation of AMPK phosphorylated acetyl-CoA carboxylase, but treatment with AICAR or other AMPK activators (A769662, phenformin) did not restore ULK1 phosphorylation or autophagosome formation. To determine whether palmitate-induced ceramide accumulation contributed to this finding, we overexpressed a ceramide-metabolizing enzyme, acid ceramidase. The increase in acid ceramidase expression ameliorated the effects of excess nutrients on ULK1 phosphorylation, without altering the effects of the AMPK activators. Thus, unlike low nutrient conditions, AMPK becomes uncoupled from autophagy in HAECs in a nutrient-rich environment, such as that found in patients with increased cardiovascular risk. These findings suggest that combinations of AMPK-independent and AMPK-dependent therapies may be more effective alternatives than either therapy alone for treating nutrient-induced cellular dysfunction.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aorta/physiology , Autophagy/physiology , Endothelium, Vascular/physiology , Glucose/administration & dosage , Palmitic Acid/administration & dosage , Aorta/drug effects , Autophagy/drug effects , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Uncoupling Agents/administration & dosage
13.
J Clin Endocrinol Metab ; 99(12): E2518-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25210878

ABSTRACT

CONTEXT: A subpopulation of obese individuals remains insulin sensitive (ISO). They represent a unique human model to investigate factors underlying insulin resistance (IR) without the confounding effect of major differences in weight/adiposity. Altered fatty-acid (FA) metabolism in sc adipose tissue (SAT) contributes to obesity-associated IR. OBJECTIVE: To test the hypothesis that ISO and body mass index-matched insulin-resistant obese (IRO) patients demonstrate differential SAT expression profiles of genes involved in glycerolipid-FA metabolism and that weight loss-induced improvement of IR ameliorates these changes. DESIGN AND SETTING: A cross-sectional and longitudinal study. PATIENTS AND INTERVENTION: Thirty-eight nondiabetic obese women were stratified into ISO (n = 25) or IRO (n = 13) groups based on hyperinsulinemic-euglycemic clamp results. Subjects were studied before and after a 6-month hypocaloric diet intervention. MAIN OUTCOME MEASURES: mRNA (quantitative RT-PCR) and protein (mass spectrometry and immunoblots) levels were measured in SAT biopsies. RESULTS: Despite having age, body mass index, and fat mass similar to ISO individuals, IRO patients had lower insulin sensitivity and glucose tolerance (P < .05). Baseline SAT mRNA and protein levels of genes involved in both the synthesis and lipolysis of glycerolipid-FAs were higher in IRO individuals (P < .05), even when groups were matched for visceral adipose tissue content. The dietary intervention resulted in approximately 6% weight loss in both the IRO and ISO groups (P < .05) but only ameliorated insulin sensitivity in IRO individuals (P < .05). Likewise, the intervention reduced the expression of most glycerolipid-FA metabolism genes (P < .05), with expression levels in IRO individuals being restored to ISO levels. CONCLUSIONS: Increased SAT expression of genes involved in both the synthesis and hydrolysis of glycerolipid-FAs is closely associated with IR in obese women. The results suggest that enhanced glycerolipid-FA cycling in SAT contributes to obesity-associated IR.


Subject(s)
Fatty Acids/genetics , Fatty Acids/metabolism , Glycolipids/genetics , Glycolipids/metabolism , Insulin Resistance/genetics , Obesity/genetics , Obesity/metabolism , Subcutaneous Fat/metabolism , Aged , Cohort Studies , Cross-Sectional Studies , Diet, Reducing , Female , Gene Expression , Humans , Longitudinal Studies , Middle Aged , Obesity/diet therapy , Postmenopause/genetics
14.
Arch Biochem Biophys ; 562: 62-9, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25172224

ABSTRACT

Recent studies have highlighted the importance of an inhibitory phosphorylation site, Ser(485/491), on the α-subunit of AMP-activated protein kinase (AMPK); however, little is known about the regulation of this site in liver and skeletal muscle. We examined whether the inhibitory effects of insulin on AMPK activity may be mediated through the phosphorylation of this inhibitory Ser(485/491) site in hepatocytes, myotubes and incubated skeletal muscle. HepG2 and C2C12 cells were stimulated with or without insulin for 15-min. Similarly, rat extensor digitorum longus (EDL) muscles were treated +/- insulin for 10-min. Insulin significantly increased Ser(485/491) p-AMPK under all conditions, resulting in a subsequent reduction in AMPK activity, ranging from 40% to 70%, despite no change in p-AMPK Thr(172). Akt inhibition both attenuated the increase in Ser(485/491) p-AMPK caused by insulin, and prevented the decrease in AMPK activity. Similarly, the growth factor IGF-1 stimulated Ser(485/491) AMPK phosphorylation, and this too was blunted by inhibition of Akt. Inhibition of the mTOR pathway with rapamycin, however, had no effect on insulin-stimulated Ser(485/491) p-AMPK. These data suggest that insulin and IGF-1 diminish AMPK activity in hepatocytes and muscle, most likely through Akt activation and the inhibitory phosphorylation of Ser(485/491) on its α-subunit.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Serine/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line , Hep G2 Cells , Hepatocytes/drug effects , Humans , Insulin-Like Growth Factor I/metabolism , Male , Mice , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Phosphorylation , Rats , Rats, Sprague-Dawley , Sirolimus/pharmacology
15.
Diabetes Metab Syndr Obes ; 7: 241-53, 2014.
Article in English | MEDLINE | ID: mdl-25018645

ABSTRACT

Type 2 diabetes (T2D) is a metabolic disease characterized by insulin resistance, ß-cell dysfunction, and elevated hepatic glucose output. Over 350 million people worldwide have T2D, and the International Diabetes Federation projects that this number will increase to nearly 600 million by 2035. There is a great need for more effective treatments for maintaining glucose homeostasis and improving insulin sensitivity. AMP-activated protein kinase (AMPK) is an evolutionarily conserved serine/threonine kinase whose activation elicits insulin-sensitizing effects, making it an ideal therapeutic target for T2D. AMPK is an energy-sensing enzyme that is activated when cellular energy levels are low, and it signals to stimulate glucose uptake in skeletal muscles, fatty acid oxidation in adipose (and other) tissues, and reduces hepatic glucose production. There is substantial evidence suggesting that AMPK is dysregulated in animals and humans with metabolic syndrome or T2D, and that AMPK activation (physiological or pharmacological) can improve insulin sensitivity and metabolic health. Numerous pharmacological agents, natural compounds, and hormones are known to activate AMPK, either directly or indirectly - some of which (for example, metformin and thiazolidinediones) are currently used to treat T2D. This paper will review the regulation of the AMPK pathway and its role in T2D, some of the known AMPK activators and their mechanisms of action, and the potential for future improvements in targeting AMPK for the treatment of T2D.

16.
Curr Obes Rep ; 3(2): 248-55, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24891985

ABSTRACT

Although a correlation exists between obesity and insulin resistance, roughly 25 % of obese individuals are insulin sensitive. AMP-activated protein kinase (AMPK) is a cellular energy sensor that among its many actions, integrates diverse physiological signals to restore energy balance. In addition, in many situations it also increases insulin sensitivity. In this context, AMPK activity is decreased in very obese individuals undergoing bariatric surgery who are insulin resistant compared to equally obese patients who are insulin sensitive. In this review, we will both explore what distinguishes these individuals, and evaluate the evidence that diminished AMPK is associated with insulin resistance and metabolic syndrome-associated disorders in other circumstances.

17.
PLoS One ; 9(5): e97554, 2014.
Article in English | MEDLINE | ID: mdl-24835252

ABSTRACT

Liraglutide is a glucagon-like peptide-1 (GLP-1) mimetic used for the treatment of Type 2 diabetes. Similar to the actions of endogenous GLP-1, liraglutide potentiates the post-prandial release of insulin, inhibits glucagon release and increases satiety. Recent epidemiological studies and clinical trials have suggested that treatment with GLP-1 mimetics may also diminish the risk of cardiovascular disease in diabetic patients. The mechanism responsible for this effect has yet to be determined; however, one possibility is that they might do so by a direct effect on vascular endothelium. Since low grade inflammation of the endothelium is an early event in the pathogenesis of atherosclerotic cardiovascular disease (ASCVD), we determined the effects of liraglutide on inflammation in cultured human aortic endothelial cells (HAECs). Liraglutide reduced the inflammatory responses to TNFα and LPS stimulation, as evidenced by both reduced protein expression of the adhesion molecules VCAM-1 and E-Selectin, and THP-1 monocyte adhesion. This was found to result from increased cell Ca2+ and several molecules sensitive to Ca2+ with known anti inflammatory actions in endothelial cells, including CaMKKß, CaMKI, AMPK, eNOS and CREB. Treatment of the cells with STO-609, a CaMKK inhibitor, diminished both the activation of AMPK, CaMKI and the inhibition of TNFα and LPS-induced monocyte adhesion by liraglutide. Likewise, expression of an shRNA against AMPK nullified the anti-inflammatory effects of liraglutide. The results indicate that liraglutide exerts a strong anti-inflammatory effect on HAECs. They also demonstrate that this is due to its ability to increase intracellular Ca2+ and activate CAMKKß, which in turn activates AMPK.


Subject(s)
Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Incretins/pharmacology , Inflammation/drug therapy , AMP-Activated Protein Kinases/metabolism , Analysis of Variance , Benzimidazoles , Blotting, Western , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Humans , Immunoenzyme Techniques , Incretins/metabolism , Incretins/therapeutic use , Liraglutide , Naphthalimides , RNA Interference , RNA, Small Interfering/genetics
18.
J Clin Invest ; 123(7): 2764-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23863634

ABSTRACT

Insulin resistance (IR) and hyperinsulinemia are hallmarks of the metabolic syndrome, as are central adiposity, dyslipidemia, and a predisposition to type 2 diabetes, atherosclerotic cardiovascular disease, hypertension, and certain cancers. Regular exercise and calorie restriction have long been known to increase insulin sensitivity and decrease the prevalence of these disorders. The subsequent identification of AMP-activated protein kinase (AMPK) and its activation by exercise and fuel deprivation have led to studies of the effects of AMPK on both IR and metabolic syndrome-related diseases. In this review, we evaluate this body of literature, with special emphasis on the hypothesis that dysregulation of AMPK is both a pathogenic factor for these disorders in humans and a target for their prevention and therapy.


Subject(s)
Adenylate Kinase/physiology , Insulin Resistance , Metabolic Syndrome/enzymology , Adipose Tissue/enzymology , Adipose Tissue/immunology , Animals , Endoplasmic Reticulum Stress , Enzyme Activators/metabolism , Enzyme Activators/pharmacology , Humans , Macrophage Activation , Metabolic Syndrome/drug therapy , Metabolic Syndrome/immunology , Obesity/enzymology , Obesity/immunology , Oxidative Stress , Sirtuin 1/metabolism
19.
Mol Cell ; 50(5): 686-98, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746352

ABSTRACT

Lipid metabolism is tightly controlled by the nutritional state of the organism. Nutrient-rich conditions increase lipogenesis, whereas nutrient deprivation promotes fat oxidation. In this study, we identify the mitochondrial sirtuin, SIRT4, as a regulator of lipid homeostasis. SIRT4 is active in nutrient-replete conditions to repress fatty acid oxidation while promoting lipid anabolism. SIRT4 deacetylates and inhibits malonyl CoA decarboxylase (MCD), an enzyme that produces acetyl CoA from malonyl CoA. Malonyl CoA provides the carbon skeleton for lipogenesis and also inhibits fat oxidation. Mice lacking SIRT4 display elevated MCD activity and decreased malonyl CoA in skeletal muscle and white adipose tissue. Consequently, SIRT4 KO mice display deregulated lipid metabolism, leading to increased exercise tolerance and protection against diet-induced obesity. In sum, this work elucidates SIRT4 as an important regulator of lipid homeostasis, identifies MCD as a SIRT4 target, and deepens our understanding of the malonyl CoA regulatory axis.


Subject(s)
Carboxy-Lyases/metabolism , Lipid Metabolism , Mitochondrial Proteins/metabolism , Sirtuins/metabolism , Acetylation , Adipose Tissue, White/metabolism , Animals , Diet , Fatty Acids/metabolism , Lipid Metabolism/genetics , Lipids/biosynthesis , Male , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Obesity/etiology , Obesity/metabolism , Oxidation-Reduction , Sirtuins/genetics
20.
Diabetes ; 62(6): 2122-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23378607

ABSTRACT

Our objective was to determine if the insulin-sensitizing drug pioglitazone acutely reduces insulin secretion and causes metabolic deceleration in vivo independently of change in insulin sensitivity. We assessed glucose homeostasis by hyperinsulinemic-euglycemic and hyperglycemic clamp studies and energy expenditure by indirect calorimetry and biotelemetry in male Wistar and obese hyperinsulinemic Zucker diabetic fatty (ZDF) rats 45 min after a single oral dose of pioglitazone (30 mg/kg). In vivo insulin secretion during clamped hyperglycemia was reduced in both Wistar and ZDF rats after pioglitazone administration. Insulin clearance was slightly increased in Wistar but not in ZDF rats. Insulin sensitivity in Wistar rats assessed by the hyperinsulinemic-euglycemic clamp was minimally affected by pioglitazone at this early time point. Pioglitazone also reduced energy expenditure in Wistar rats without altering respiratory exchange ratio or core body temperature. Glucose-induced insulin secretion (GIIS) and oxygen consumption were reduced by pioglitazone in isolated islets and INS832/13 cells. In conclusion, pioglitazone acutely induces whole-body metabolic slowing down and reduces GIIS, the latter being largely independent of the insulin-sensitizing action of the drug. The results suggest that pioglitazone has direct metabolic deceleration effects on the ß-cell that may contribute to its capacity to lower insulinemia and antidiabetic action.


Subject(s)
Insulin/metabolism , Thiazolidinediones/therapeutic use , Animals , Calorimetry, Indirect , Energy Metabolism/drug effects , Glucose/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin Secretion , Male , Oxygen Consumption/drug effects , Pioglitazone , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...