ABSTRACT
Insulin resistance impairs postprandial glucose uptake through glucose transporter type 4 (GLUT4) and is the primary defect preceding type 2 diabetes. We previously generated an insulin-resistant mouse model with human GLUT4 promoter-driven insulin receptor knockout (GIRKO) in the muscle, adipose, and neuronal subpopulations. However, the rate of diabetes in GIRKO mice remained low prior to 6 months of age on normal chow diet (NCD), suggesting that additional factors/mechanisms are responsible for adverse metabolic effects driving the ultimate progression of overt diabetes. In this study, we characterized the metabolic phenotypes of the adult GIRKO mice acutely switched to high-fat diet (HFD) feeding in order to identify additional metabolic challenges required for disease progression. Distinct from other diet-induced obesity (DIO) and genetic models (e.g., db/db mice), GIRKO mice remained leaner on HFD feeding, but developed other cardinal features of insulin resistance syndrome. GIRKO mice rapidly developed hyperglycemia despite compensatory increases in ß-cell mass and hyperinsulinemia. Furthermore, GIRKO mice also had impaired oral glucose tolerance and a limited glucose-lowering benefit from exendin-4, suggesting that the blunted incretin effect contributed to hyperglycemia. Secondly, GIRKO mice manifested severe dyslipidemia while on HFD due to elevated hepatic lipid secretion, serum triglyceride concentration, and lipid droplet accumulation in hepatocytes. Thirdly, GIRKO mice on HFD had increased inflammatory cues in the gut, which were associated with the HFD-induced microbiome alterations and increased serum lipopolysaccharide (LPS). In conclusion, our studies identified important gene/diet interactions contributing to diabetes progression, which might be leveraged to develop more efficacious therapies.
Subject(s)
Diabetes Mellitus, Type 2 , Diet, High-Fat , Glucose Intolerance , Glucose Transporter Type 4 , Hyperglycemia , Insulin Resistance , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/metabolism , Hyperglycemia/blood , Hyperglycemia/etiology , Hyperglycemia/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Mice, KnockoutABSTRACT
Cadmium (Cd) is an environmental contaminant that causes renal toxicity. We have previously demonstrated that Cd induces renal toxicity by altering transcriptional activities. In this study, we show that Cd markedly inhibited the activity of transcription factor MEF2A in HK-2 human proximal tubule cells, which generated significant cytotoxicity in the cells. This reduction in the nuclear levels of MEF2A protein may be involved in the Cd-induced inhibition of MEF2A activity. We also demonstrate that one of the glucose transporters, GLUT4, was downregulated not only by Cd treatment but also by MEF2A knockdown. Knockdown of SLC2A4, encoding GLUT4, eliminated both cell viability and Cd toxicity. Cd treatment or SLC2A4 deficiency reduced the cellular concentration of glucose. Therefore, the suppression of SLC2A4 expression, which mediates the reduction in cellular glucose, is involved in Cd toxicity. The Cd toxicity induced by the reduction in GLUT4 may be associated with a reduction of cellular ATP levels in HK-2 cells. The levels of Slc2a4 mRNA in the kidney of mice exposed to Cd for 6 or 12 months were significantly lower than those in the control group. These results demonstrate that Cd exerts its cytotoxicity through the suppression in SLC2A4 expression and the subsequent inhibition of MEF2A transcriptional activity. Cd-induced suppression of SLC2A4 expression also reduces cellular ATP levels, partly by reducing glucose levels. This study suggests that the glucose transporter plays an important role in the renal toxicity of Cd, and provides a crucial breakthrough in our understanding of the mechanism of Cd toxicity.
Subject(s)
Cadmium/toxicity , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/biosynthesis , Kidney Tubules, Proximal/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Glucose Transporter Type 4/genetics , Humans , Kidney Tubules, Proximal/pathology , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , MiceABSTRACT
Polycystic ovary syndrome (PCOS) is an endocrine-metabolic disorder characterized by hyperandrogenism and ovulatory dysfunction but also obesity and hyperinsulinemia. These characteristics induce an insulin-resistant state in tissues such as the endometrium, affecting its reproductive functions. Myo-inositol (MYO) is an insulin-sensitizing compound used in PCOS patients; however, its insulin-sensitizing mechanism is unclear. To understand the relationship of MYO with insulin action in endometrial cells, sodium/myo-inositol transporter 1 (SMIT-1) (MYO-transporter), and MYO effects on protein levels related to the insulin pathway were evaluated. SMIT-1 was assessed in endometrial tissue from women with normal weight, obesity, insulin resistance, and PCOS; additionally, using an in vitro model of human endometrial cells exposed to an environment resembling hyperinsulinemic-obese-PCOS, MYO effect was evaluated on p-AMPK and GLUT-4 levels and glucose uptake by Western blot, immunocytochemistry, and confocal microscopy, respectively. SMIT-1 was detected in endometrial tissue from all groups and decreased in PCOS and obesity (P < 0.05 vs. normal weight). In the in vitro model, PCOS conditions decreased p-AMPK levels, while they were restored with MYO (P < 0.05). The diminished GLUT-4 protein levels promoted by PCOS environment were restored by MYO through SMIT-1 and p-AMPK-dependent mechanism (P < 0.05). Also, MYO restored glucose uptake in cells under PCOS condition through a p-AMPK-dependent mechanism. Finally, these results were similar to those obtained with metformin treatment in the same in vitro conditions. Consequently, MYO could be a potential insulin sensitizer through its positive effects on insulin-resistant tissues as PCOS-endometrium, acting through SMIT-1, provoking AMPK activation and elevated GLUT-4 levels and, consequently, increase glucose uptake by human endometrial cells. Therefore, MYO may be used as an effective treatment option in insulin-resistant PCOS women.
Subject(s)
AMP-Activated Protein Kinases/metabolism , Endometrium/metabolism , Glucose Transporter Type 4/biosynthesis , Inositol/metabolism , Insulin Resistance , Polycystic Ovary Syndrome/metabolism , Adult , Cells, Cultured , Endometrium/cytology , Enzyme Activation , Female , Glucose/metabolism , Glucose Transporter Type 4/genetics , Heat-Shock Proteins/metabolism , Humans , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Obesity/metabolism , Polycystic Ovary Syndrome/genetics , Symporters/metabolismABSTRACT
BACKGROUND: The aim of the study was to investigate the relationship between post-myocardial infarction (MI) inflammation and left ventricular (LV) remodeling in a swine model by 18F-fluorodeoxyglucose (FDG) imaging. METHODS: MI was induced in swine by balloon occlusion of the left anterior descending coronary artery. A series of FDG positron emission tomography (PET) images were taken within 2 weeks post-MI, employing a comprehensive strategy to suppress the physiological uptake of cardiomyocytes. Echocardiography was applied to evaluate LV volume, global and regional function. CD68+ macrophage and glucose transporters (GLUT-1, -3 and -4) were investigated by immunostaining. RESULTS: The physiological uptake of myocardium was adequately suppressed in 92.3% of PET scans verified by visual analysis, which was further confirmed by the minimal expression of myocardial GLUT-4. Higher FDG uptake was observed in the infarct than in the remote area and persisted within the observational period of 2 weeks. The FDG uptake of infarcted myocardium on day 1 post-MI was correlated with LV global remodeling, and the FDG uptake of infarcted myocardium on days 1 and 8 post-MI had a trend of correlating with regional remodeling of the infarct area. CONCLUSIONS: We here report a feasible swine model for investigating post-MI inflammation. FDG signal in the infarct area of swine persisted for a longer duration than has been reported in small animals. FDG activity in the infarct area could predict LV remodeling.
Subject(s)
Fluorodeoxyglucose F18 , Heart Ventricles/diagnostic imaging , Inflammation/diagnostic imaging , Myocardial Infarction/diagnostic imaging , Myocardial Perfusion Imaging/methods , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Coronary Vessels/pathology , Echocardiography , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 3/biosynthesis , Glucose Transporter Type 4/biosynthesis , Heart/diagnostic imaging , Image Processing, Computer-Assisted , Muscle Cells/pathology , Myocytes, Cardiac/metabolism , Necrosis , Positron-Emission Tomography , Radiopharmaceuticals , SwineABSTRACT
In situ detection of the expression level of cell-surface receptors has become a hotspot study in recent years. We propose in this manuscript a novel strategy for sensitive electrochemiluminescence (ECL) detection of glucose transporter 4 (GLUT4) on human skeletal muscle cells (HSMCs). Graphene hydrogel (GH) was selected to fabricate a permeable electrode with the purpose of overcoming the steric hindrance of cells on electrode, which leads to errors in the detection of cell-surface receptors. GLUT4 was labeled with carbon dots (CDs), which generate ECL emission at the interface between GH and cells, so about half the amount of GLUT4 expressed at the cell surface could be determined, which provided an accurate GLUT4 expression quantification. The prepared cytosensor exhibited good analytical performance for HSMC cells, ranging from 500 to 1.0 × 106 cells·mL-1, with a detection limit of 200 cells·mL-1. The average amount of GLUT4 per HSMC cell was calculated to be 1.88 × 105. Furthermore, GLUT4 on HSMC surface had a 2.3-fold increase under the action of insulin. This strategy is capable of evaluating the receptors on the cell surface, which may push the application of ECL for disease diagnosis.
Subject(s)
Biosensing Techniques , Electrochemical Techniques , Glucose Transporter Type 4/biosynthesis , Luminescent Measurements , Mesenchymal Stem Cells/chemistry , Muscle, Skeletal/chemistry , Electrodes , Glucose Transporter Type 4/chemistry , Graphite/chemistry , Humans , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Muscle, Skeletal/cytology , Optical Imaging , Particle Size , Surface PropertiesABSTRACT
Skeletal muscle is the major site of postprandial peripheral glucose uptake, but in obesity-induced insulin-resistant states insulin-stimulated glucose disposal is markedly impaired. Despite the importance of skeletal muscle in regulating glucose homeostasis, the specific transcriptional changes associated with insulin-sensitive vs. -resistant states in muscle remain to be fully elucidated. Herein, using an RNA-seq approach we identified 20 genes differentially expressed in an insulin-resistant state in skeletal muscle, including cysteine- and glycine-rich protein 3 ( Csrp3), which was highly expressed in insulin-sensitive conditions but significantly reduced in the insulin-resistant state. CSRP3 has diverse functional roles including transcriptional regulation, signal transduction, and cytoskeletal organization, but its role in glucose homeostasis has yet to be explored. Thus, we investigated the role of CSRP3 in the development of obesity-induced insulin resistance in vivo. High-fat diet-fed CSRP3 knockout (KO) mice developed impaired glucose tolerance and insulin resistance as well as increased inflammation in skeletal muscle compared with wild-type (WT) mice. CSRP3-KO mice had significantly impaired insulin signaling, decreased GLUT4 translocation to the plasma membrane, and enhanced levels of phospho-PKCα in muscle, which all contributed to reduced insulin-stimulated glucose disposal in muscle in HFD-fed KO mice compared with WT mice. CSRP3 is a highly inducible protein and its expression is acutely increased after fasting. After 24h fasting, glucose tolerance was significantly improved in WT mice, but this effect was blunted in CSRP3-KO mice. In summary, we identify a novel role for Csrp3 expression in skeletal muscle in the development of obesity-induced insulin resistance.
Subject(s)
Glucose/metabolism , Homeostasis/physiology , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Animals , Diet, High-Fat , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/genetics , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Protein Kinase C/metabolismABSTRACT
Diabetes has been identified as major risk factor for atrial fibrillation (AF). Although glucose and insulin disturbances during diabetes may affect atrial function, little is known about the potential pathogenic role of glucose metabolism during AF. Glucose transport into the cell via glucose transporters (GLUTs) is the rate-limiting step of glucose utilization. Although GLUT4 is the major isoform, GLUT8 has emerged as a novel insulin-sensitive cardiac isoform. We hypothesized that atrial glucose homeostasis will be impaired during insulin resistance-induced AF. AF was induced by transesophageal atrial pacing in healthy mice and following a long-term high-fat-diet-induced insulin resistance. Active cell surface GLUT content was measured using the biotinylated photolabeling assay in the intact perfused heart. Atrial fibrosis, advanced glycation end products (AGEs) and glycogen were measured in the atria using histological analyses. Animals fed a high-fat-diet were obese and mildly hyperglycemic, and developed insulin resistance compared to controls. Insulin-resistant (IR) animals demonstrated an increased vulnerability to induced AF, as well as spontaneous AF. Insulin-stimulated translocation of GLUT4 and GLUT8 was down-regulated in the atria of IR animals, as well as their total protein expression. We also reported the absence of fibrosis, glycogen and AGE accumulation in the atria of IR animals. In the absence of structural remodeling and atrial fibrosis, these data suggest that insulin signaling dysregulation, resulting in impaired glucose transport in the atria, could provide a metabolic arrhythmogenic substrate and be a novel early pathogenic factor of AF.
Subject(s)
Atrial Fibrillation/metabolism , Gene Expression Regulation , Glucose Transporter Type 4/biosynthesis , Insulin Resistance , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Glucose Transport Proteins, Facilitative/genetics , Glucose Transporter Type 4/genetics , Heart Atria/metabolism , Heart Atria/pathology , Male , MiceABSTRACT
Insulin resistance is a condition in which there is a defect in insulin actions to induce glucose uptake into the cells. Overstimulation of ß2-adrenergic receptors (ß2ARs) is associated with the pathogenesis of insulin resistance in the heart. However, the mechanisms by which ß2-agonists affect insulin resistance in the heart are incompletely understood. The ß2-agonists are used for treatment of asthma due to bronchodilating effects. We also investigated the effects of ß2-agonists in human bronchial smooth muscle (HBSM) cells. In this study, we demonstrate that chronic treatment with salbutamol, salmeterol, and formoterol inhibited insulin-induced glucose uptake and GLUT4 synthesis in H9c2 myoblast cells. Sustained ß2AR stimulation also attenuated GLUT4 translocation to the plasma membrane, whereas short-term stimulation had no effect. In HBSM cells, prolonged treatment with ß2-agonists had no effect on insulin-induced glucose uptake and did not alter insulin-induced expressions of GLUT1, GLUT4, and GLUT10. In addition, genetic polymorphisms at amino acid positions 16 and 27 of ß2AR are linked to insulin resistance by significant suppression of GLUT4 translocation compared to wild-type. Thus, prolonged ß2AR stimulation by ß2-agonists impairs insulin actions through suppression of GLUT synthesis and translocation only in H9c2 cells.
Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Insulin Antagonists/pharmacology , Insulin/pharmacology , Receptors, Adrenergic, beta-2/metabolism , Albuterol/pharmacology , Cells, Cultured , Formoterol Fumarate/pharmacology , Glucose/metabolism , Glucose Transport Proteins, Facilitative/biosynthesis , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 4/biosynthesis , Humans , Insulin Resistance , Polymorphism, Genetic , Salmeterol Xinafoate/pharmacologyABSTRACT
The prevalence of obesity and type 2 diabetes is increasing worldwide and threatens to shorten lifespan. Impaired insulin action in peripheral tissues is a major pathogenic factor. Insulin stimulates glucose uptake in adipose tissue through the GLUT4 (also known as SLC2A4) glucose transporter, and alterations in adipose tissue GLUT4 expression or function regulate systemic insulin sensitivity. Downregulation of human and mouse adipose tissue GLUT4 occurs early in diabetes development. Here we report that adipose tissue GLUT4 regulates the expression of carbohydrate-responsive-element-binding protein (ChREBP; also known as MLXIPL), a transcriptional regulator of lipogenic and glycolytic genes. Furthermore, adipose ChREBP is a major determinant of adipose tissue fatty acid synthesis and systemic insulin sensitivity. We find a new mechanism for glucose regulation of ChREBP: glucose-mediated activation of the canonical ChREBP isoform (ChREBP-α) induces expression of a novel, potent isoform (ChREBP-ß) that is transcribed from an alternative promoter. ChREBP-ß expression in human adipose tissue predicts insulin sensitivity, indicating that it may be an effective target for treating diabetes.
Subject(s)
Adipose Tissue/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glucose/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/pathology , Adiposity , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Blood Glucose/metabolism , Body Mass Index , Body Weight , Cells, Cultured , Cohort Studies , Cross-Sectional Studies , Diabetes Mellitus/blood , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Female , Gene Expression Regulation/genetics , Genotype , Glucose/pharmacology , Glucose Intolerance/genetics , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Homeostasis/genetics , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance/genetics , Lipogenesis , Male , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Obesity/genetics , Obesity/metabolism , Promoter Regions, Genetic/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/geneticsABSTRACT
Abnormal glucose metabolism induces various metabolic disorders such as insulin resistance and type 2 diabetes. Regular exercise improved glucose uptake and enhanced glucose oxidation by increasing GLUT4 transcription in skeletal muscle. However, the regulatory mechanisms of GLUT4 transcription in response to exercise are poorly understood. AMPK is a sensor of exercise and upstream kinase of class II HDACs that act as transcriptional repressors. We used 6-week treadmill exercise or one single-bout exercise wild type or AMPKα2-/- C57BL/6J mice to explore how HDACs regulate GLUT4 transcription and the underlying molecular mechanisms mediated by AMPK in the physiologic process of exercise. We demonstrate that regular physical exercise significantly enhanced GLUT4 transcription by inactivating HDAC4/5 in skeletal muscle by ChIP experiment. HDAC4 coordinately regulated with HDAC5 represses transcriptional activity of GLUT4 promoter in C2C12 myotubes by Luciferase assay. If either HDAC4 or HDAC5 is silenced via RNAi technology, the functional compensation by the other will occur. In addition, a single-bout of exercise decreased HDAC4/5 activity in skeletal muscle of wild type but not in AMPKα2-/- mice, suggesting an AMPKα2-dependent manner. Those findings provide new insight into the mechanisms responsible for AMPKα2-dependent regulation of GLUT4 transcription after exercise.
Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose Transporter Type 4/biosynthesis , Histone Deacetylases/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Transcription, Genetic , AMP-Activated Protein Kinases/genetics , Animals , Cell Line , Glucose Transporter Type 4/genetics , Histone Deacetylases/genetics , Male , Mice , Mice, KnockoutABSTRACT
Stuart, CA, Lee, ML, South, MA, Howell, MEA, Cartwright, BM, Ramsey, MW, and Stone, MH. Pre-training muscle characteristics of subjects who are obese determine how well exercise training will improve their insulin responsiveness. J Strength Cond Res 31(3): 798-808, 2017-Only half of prediabetic subjects who are obese who underwent exercise training without weight loss increased their insulin responsiveness. We hypothesized that those who improved their insulin responsiveness might have pretraining characteristics favoring a positive response to exercise training. Thirty nondiabetic subjects who were obese volunteered for 8 weeks of either strength training or endurance training. During training, subjects increased their caloric intake to prevent weight loss. Insulin responsiveness by euglycemic clamps and muscle fiber composition, and expression of muscle key biochemical pathways were quantified. Positive responders initially had 52% higher intermediate muscle fibers (fiber type IIa) with 27% lower slow-twitch fibers (type I) and 23% lower expression of muscle insulin receptors. Whether after weight training or stationary bike training, positive responders' fiber type shifted away from type I and type IIa fibers to an increased proportion of type IIx fibers (fast twitch). Muscle insulin receptor expression and glucose transporter type 4 (GLUT4) expression increased in all trained subjects, but these moderate changes did not consistently translate to improvement in whole-body insulin responsiveness. Exercise training of previously sedentary subjects who are obese can result in muscle remodeling and increased expression of key elements of the insulin pathway, but in the absence of weight loss, insulin sensitivity improvement was modest and limited to about half of the participants. Our data suggest rather than responders being more fit, they may have been less fit, only catching up to the other half of subjects who are obese whose insulin responsiveness did not increase beyond their pretraining baseline.
Subject(s)
Exercise Therapy/methods , Insulin/metabolism , Muscle, Skeletal/metabolism , Obesity/physiopathology , Obesity/therapy , Adult , Energy Intake , Female , Glucose Transporter Type 4/biosynthesis , Humans , Insulin Resistance/physiology , Male , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Receptor, Insulin/biosynthesis , Resistance Training/methods , Retrospective StudiesABSTRACT
The glucose transporter GLUT4 facilitates insulin-stimulated glucose uptake in peripheral tissues including adipose, muscle, and heart. GLUT4 function is impaired in obesity and type 2 diabetes leading to hyperglycemia and an increased risk of cardiovascular disease and neuropathy. To better understand the regulation of GLUT4 function, a targeted siRNA screen was performed and led to the discovery that ZFP407 regulates insulin-stimulated glucose uptake in adipocytes. The decrease in insulin-stimulated glucose uptake due to ZFP407 deficiency was attributed to a reduction in GLUT4 mRNA and protein levels. The decrease in GLUT4 was due to both decreased transcription of Glut4 mRNA and decreased efficiency of Glut4 pre-mRNA splicing. Interestingly, ZFP407 coordinately regulated this decrease in transcription with an increase in the stability of Glut4 mRNA, resulting in opposing effects on steady-state Glut4 mRNA levels. More broadly, transcriptome analysis revealed that ZFP407 regulates many peroxisome proliferator-activated receptor (PPAR) γ target genes beyond Glut4. ZFP407 was required for the PPARγ agonist rosiglitazone to increase Glut4 expression, but was not sufficient to increase expression of a PPARγ target gene reporter construct. However, ZFP407 and PPARγ co-overexpression synergistically activated a PPARγ reporter construct beyond the level of PPARγ alone. Thus, ZFP407 may represent a new modulator of the PPARγ signaling pathway.
Subject(s)
DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose Transporter Type 4/biosynthesis , Glucose/metabolism , Insulin/metabolism , Transcription Factors/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Animals , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation/genetics , Glucose Transporter Type 4/genetics , Humans , Mice , PPAR gamma/biosynthesis , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Transcription Factors/geneticsABSTRACT
Activation of hepatic stellate cells (HSCs) is characterized by expression of extracellular matrix and loss of adipogenic phenotype during liver fibrogenesis. Emerging evidence suggests that HSCs adopt aerobic glycolysis during activation. The present work aimed at investigating whether the anti-fibrogenic effects of curcumin was associated with interfering with glycolysis in HSCs. Primary rat HSCs were cultured in vitro. We demonstrated that inhibition of glycolysis by 2-deoxyglucose or galloflavin reduced the expression of α-smooth muscle actin (α-SMA) and α1(I)procollagen at both mRNA and protein levels, and increased the intracellular lipid contents and upregulated the gene and protein expression of adipogenic transcription factors C/EBPα and PPAR-γ in HSCs. Curcumin at 20 µM produced similar effects. Moreover, curcumin decreased the expression of hexokinase (HK), phosphofructokinase-2 (PFK2), and glucose transporter 4 (glut4), three key glycolytic parameters, at both mRNA and protein levels. Curcumin also reduced lactate production concentration-dependently in HSCs. Furthermore, curcumin increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK), but AMPK inhibitor BML-275 significantly abolished the curcumin downregulation of HK, PFK2, and glut4. In addition, curcumin inhibition of α-SMA and α1(I)procollagen was rescued by BML-275, and curcumin upregulation of C/EBPα and PPAR-γ was abrogated by BML-275. These results collectively indicated that curcumin inhibited glycolysis in an AMPK activation-dependent manner in HSCs. We revealed a novel mechanism for curcumin suppression of HSC activation implicated in antifibrotic therapy. © 2016 IUBMB Life, 68(7):589-596, 2016.
Subject(s)
AMP-Activated Protein Kinases/genetics , Curcumin/administration & dosage , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/drug therapy , Liver/metabolism , Actins/antagonists & inhibitors , Animals , Collagen Type I/antagonists & inhibitors , Collagen Type I, alpha 1 Chain , Deoxyglucose/biosynthesis , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/biosynthesis , Glycolysis/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Hexokinase/biosynthesis , Humans , Isocoumarins/administration & dosage , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Phosphofructokinase-2/biosynthesis , Rats , Rats, Sprague-DawleyABSTRACT
BACKGROUND The aim of this study was to investigate the effects of different statins on glucose uptake and to confirm its mechanism in primary cultured rat cardiomyocytes after administration of atorvastatin, pravastatin, and rosuvastatin. MATERIAL AND METHODS Primary cultured rat cardiomyocytes were randomly assigned to 5 groups: normal control group (OB), insulin group (S1), statin 1-µM (S2), 5-µM (S3), and 10-µM (S4) groups for 3 different statins. The 2-[3H]-DG uptake of each group was determined and the mRNA and protein expression levels of glucose transporter type 4 (GLUT4), insulin receptor substrate (IRs), and RhoA were assessed. RESULTS After treatment with different concentrations of statins and insulin, the 2-[3H]-DG uptake showed a significant negative correlation with the concentration of atorvastatin (P<0.05), and no significant correlation with pravastatin and rosuvastatin. The mRNA and protein expression levels of GLUT4 and IRs-1 in primary cultured cardiomyocytes were both significantly reduced by atorvastatin treatment (P<0.05). Pravastatin and rosuvastatin showed no significant effects on GLUT4 and IRs-1 expression. The mRNA and protein expression levels of RhoA both showed no significant difference when treated with the 3 statins. CONCLUSIONS These results confirm that atorvastatin can inhibit insulin-induced glucose uptake in primary cultured rat cardiomyocytes by regulating the PI3K/Akt insulin signal transduction pathway.
Subject(s)
Glucose/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Adipocytes/drug effects , Animals , Deoxyglucose/pharmacokinetics , Female , Glucose/metabolism , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins/biosynthesis , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , TritiumABSTRACT
Dehydrozingerone (DHZ) exerts beneficial effects on human health; however, its mechanism of action remains unclear. Here, we found that DHZ suppressed high-fat diet-induced weight gain, lipid accumulation and hyperglycaemia in C57BL/6 mice and increased AMP-activated protein kinase (AMPK) phosphorylation and stimulated glucose uptake in C2C12 skeletal muscle cells. DHZ activated p38 mitogen-activated protein kinase (MAPK) signalling in an AMPK-dependent manner. Inhibiting AMPK or p38 MAPK blocked DHZ-induced glucose uptake. DHZ increased GLUT4 (major transporter for glucose uptake) expression in skeletal muscle. Glucose clearance and insulin-induced glucose uptake increased in DHZ-fed animals, suggesting that DHZ increases systemic insulin sensitivity in vivo. Thus, the beneficial health effects of DHZ could possibly be explained by its ability to activate the AMPK pathway in skeletal muscle.
Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Muscle Fibers, Skeletal/metabolism , Styrenes/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Animals , Biological Transport/drug effects , Biological Transport/genetics , Blood Glucose/metabolism , Cells, Cultured , Curcumin/analogs & derivatives , Deoxyglucose/metabolism , Diet, High-Fat , Enzyme Activation , Glucose Transporter Type 4/biosynthesis , Hyperglycemia/drug therapy , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering , Rats , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitorsABSTRACT
Right ventricular (RV) function is a key determinant of survival in patients with both RV and left ventricular (LV) failure, yet the mechanisms of RV failure are poorly understood. Recent studies suggest cardiac metabolism is altered in RV failure in pulmonary hypertension (PH). Accordingly, we assessed mitochondrial content, dynamics, and function in hearts from neonatal calves exposed to hypobaric hypoxia (HH). This model develops severe PH with concomitant RV hypertrophy, dilation, and dysfunction. After 2 wk of HH, pieces of RV and LV were obtained along with samples from age-matched controls. Comparison with control assesses the effect of hypoxia, whereas comparison between the LV and RV in HH assesses the additional impact of RV overload. Mitochondrial DNA was unchanged in HH, as was mitochondrial content as assessed by electron microscopy. Immunoblotting for electron transport chain subunits revealed a small increase in mitochondrial content in HH in both ventricles. Mitochondrial dynamics were largely unchanged. Activity of individual respiratory chain complexes was reduced (complex I) or unchanged (complex V) in HH. Key enzymes in the glycolysis pathway were upregulated in both HH ventricles, alongside upregulation of hypoxia-inducible factor-1α protein. Importantly, none of the changes in expression or activity were different between ventricles, suggesting the changes are in response to HH and not RV overload. Upregulation of glycolytic modulators without chamber-specific mitochondrial dysfunction suggests that mitochondrial capacity and activity are maintained at the onset of PH, and the early RV dysfunction in this model results from mechanisms independent of the mitochondria.
Subject(s)
Cattle , Disease Models, Animal , Heart Ventricles/physiopathology , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/physiopathology , Mitochondria/metabolism , Ventricular Dysfunction, Right/pathology , Animals , DNA Copy Number Variations , Electron Transport Complex I/metabolism , Glucose Transporter Type 4/biosynthesis , Heart Failure/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Male , Mitochondria/genetics , Phosphofructokinase-1/biosynthesis , Protein Kinase C/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Ventricular Function, RightABSTRACT
Maternal undernutrition (UN) is associated with the development of obesity and metabolic complications in adult offspring. This study investigated the impact of preweaning growth hormone (GH) treatment on adipocyte functionality in adult male offspring. Sprague-Dawley rats were assigned either standard (C) or undernourished (UN) diet (50% ad libitum) throughout gestation. Postnatal day 3-21, male C/UN pups received either saline (CS, UNS) or GH (2.5 µg/g/d; CGH, UNGH) by subcutaneous injection. Primary adipocytes were isolated following the collagenase digestion of adipose tissue. Primary adipocytes from UN offspring had significantly increased the secretion of pro-inflammatory cytokines accompanied by increased cytokine/cytokine receptor expression. This correlated with increased TLR4/NF-κB signaling. While increased inflammatory potential was not observed in adipocytes derived from UNGH offspring, there was a clear alteration in the expression of genes relating to carbohydrate and lipid metabolism along with nutrient transporters. Overall, preweaning GH treatment alters detrimental patterns of development, which predispose UN offspring to obesity and insulin resistance.
Subject(s)
Adipocytes/metabolism , Cytokines/metabolism , Growth Hormone/pharmacology , Malnutrition/metabolism , Receptors, Cytokine/biosynthesis , Adipose Tissue , Animals , Biological Transport , Blood Glucose , Carbohydrate Metabolism/genetics , Cytokines/biosynthesis , Female , Glucose Transporter Type 4/biosynthesis , Inflammation/genetics , Inflammation/immunology , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Lipid Metabolism/genetics , Male , Maternal Nutritional Physiological Phenomena , NF-kappa B/metabolism , Obesity/metabolism , PPAR gamma/biosynthesis , PPAR gamma/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolismABSTRACT
Gene expression studies of subcutaneous adipose tissue may help to better understand the mechanisms behind body fat changes in HIV-infected patients who initiate antiretroviral therapy (ART). Here, we evaluated early changes in adipose tissue gene expression and their relationship to fat changes in ART-naive HIV-infected patients randomly assigned to initiate therapy with emtricitabine/tenofovir plus efavirenz (EFV) or ritonavir-boosted lopinavir (LPV/r). Patients had abdominal subcutaneous adipose tissue biopsies at baseline and week 16 and dual-energy-X-ray absorptiometry at baseline and weeks 16 and 48. mRNA changes of 11 genes involved in adipogenesis, lipid and glucose metabolism, mitochondrial energy, and inflammation were assessed through reverse transcription-quantitative PCR (RT-qPCR). Additionally, correlations between gene expression changes and fat changes were evaluated. Fat increased preferentially in the trunk with EFV and in the limbs with LPV/r (P < 0.05). After 16 weeks of exposure to the drug regimen, transcripts of CEBP/A, ADIPOQ, GLUT4, LPL, and COXIV were significantly down-regulated in the EFV arm compared to the LPV/r arm (P < 0.05). Significant correlations were observed between LPL expression change and trunk fat change at week 16 in both arms and between CEBP/A or COXIV change and trunk fat change at the same time point only in the EFV arm and not in the LPV/r arm. When combined with emtricitabine/tenofovir as standard backbone therapy, EFV and LPV/r induced differential early expression of genes involved in adipogenesis and energy metabolism. Moreover, these mRNA expression changes correlated with trunk fat change in the EFV arm. (This was a substudy of a randomized clinical trial [LIPOTAR study] registered at ClinicalTrials.gov under identifier NCT00759070.).
Subject(s)
Adipogenesis/genetics , Benzoxazines/therapeutic use , Body Composition/drug effects , Lopinavir/therapeutic use , Ritonavir/therapeutic use , Subcutaneous Fat/cytology , Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/virology , Adenine/analogs & derivatives , Adenine/therapeutic use , Adiponectin/biosynthesis , Adult , Alkynes , Anti-HIV Agents/therapeutic use , CCAAT-Enhancer-Binding Proteins/biosynthesis , Cyclopropanes , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Combinations , Emtricitabine , Energy Metabolism/genetics , Female , Gene Expression , Glucose/metabolism , Glucose Transporter Type 4/biosynthesis , HIV-1/drug effects , Humans , Inflammation/genetics , Lipid Metabolism/genetics , Lipoprotein Lipase/genetics , Male , Organophosphonates/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , TenofovirABSTRACT
Melatonin is an old and ubiquitous molecule in nature showing multiple mechanisms of action and functions in practically every living organism. In mammals, pineal melatonin functions as a hormone and a chronobiotic, playing a major role in the regulation of the circadian temporal internal order. The anti-obesogen and the weight-reducing effects of melatonin depend on several mechanisms and actions. Experimental evidence demonstrates that melatonin is necessary for the proper synthesis, secretion, and action of insulin. Melatonin acts by regulating GLUT4 expression and/or triggering, via its G-protein-coupled membrane receptors, the phosphorylation of the insulin receptor and its intracellular substrates mobilizing the insulin-signaling pathway. Melatonin is a powerful chronobiotic being responsible, in part, by the daily distribution of metabolic processes so that the activity/feeding phase of the day is associated with high insulin sensitivity, and the rest/fasting is synchronized to the insulin-resistant metabolic phase of the day. Furthermore, melatonin is responsible for the establishment of an adequate energy balance mainly by regulating energy flow to and from the stores and directly regulating the energy expenditure through the activation of brown adipose tissue and participating in the browning process of white adipose tissue. The reduction in melatonin production, as during aging, shift-work or illuminated environments during the night, induces insulin resistance, glucose intolerance, sleep disturbance, and metabolic circadian disorganization characterizing a state of chronodisruption leading to obesity. The available evidence supports the suggestion that melatonin replacement therapy might contribute to restore a more healthy state of the organism.
Subject(s)
Adipose Tissue, Brown/metabolism , Energy Metabolism , Melatonin/metabolism , Obesity/metabolism , Adipose Tissue, Brown/pathology , Animals , Gene Expression Regulation , Glucose Transporter Type 4/biosynthesis , Humans , Melatonin/therapeutic use , Obesity/drug therapy , Obesity/pathologyABSTRACT
OBJECTIVES: Glucose transporter (GLUT) 4 is a major mediator of blood glucose levels and a key regulator of whole-body glucose homeostasis. This study aimed at evaluating the function of goat GLUT4 on glucose absorption and the effect of GLUT4 on lactose synthesis in goat mammary gland epithelial (GMGE) cells. METHODS: Currently, the cDNA of GLUT4, a putative facilitative GLUT, was cloned from goat. To investigate the function of goat GLUT4, we constructed the eukaryotic expression vector pcDNA3.1-GLUT4 and used it to transfect GMGE cells, and then GLUT4 transfected GMGE (G4T-GMGE) cells were generated. The deduced GLUT4 sequence comprised 509 amino acids, what meant that a putative protein with a molecular weight of approximately 55 kDa would be produced. Both glucose uptake and lactose synthesis increased in the G4T-GMGE cells compared with the GMGE cells. At the transcriptional level, GLUT4 expression increased by nearly 55-fold in the G4T-GMGE cells, and the expression of amino acid transporters (SLC1A5, SLC3A2 and SLC7A5) enhanced as well; in contrast, GLUT1 expression decreased by more than 50 % in the G4T-GMGE cells. CONCLUSION: These results suggest that goat GLUT4 functions in the transport of glucose and it may play a positive role in amino acid uptake in mammary glands.