Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Br J Nutr ; 121(12): 1334-1344, 2019 06.
Article in English | MEDLINE | ID: mdl-30924427

ABSTRACT

Reduced plasma vitamin D (VD) levels may contribute to excessive white adipose tissue, insulin resistance (IR) and dyslipidaemia. We evaluated the effect of chronic oral VD supplementation on adiposity and insulin secretion in monosodium glutamate (MSG)-treated rats. During their first 5 d of life, male neonate rats received subcutaneous injections of MSG (4 g/kg), while the control (CON) group received saline solution. After weaning, groups were randomly distributed into VD supplemented (12 µg/kg; three times/week) and non-supplemented (NS) rats, forming four experimental groups (n 15 rats/group): CON-NS, CON-VD, MSG-NS and MSG-VD. At 76 d of life, rats were submitted to an oral glucose tolerance test (OGTT; 2 g/kg), and at 86 d, obesity, IR and plasma metabolic parameters were evaluated. Pancreatic islets were isolated for glucose-induced insulin secretion (GIIS), cholinergic insulinotropic response and muscarinic 3 receptor (M3R), protein kinase C (PKC) and protein kinase A (PKA) expressions. Pancreas was submitted to histological analyses. VD supplementation decreased hyperinsulinaemia (86 %), hypertriacylglycerolaemia (50 %) and restored insulin sensibility (89 %) in MSG-VD rats, without modifying adiposity, OGTT or GIIS, compared with the MSG-NS group. The cholinergic action was reduced (57 %) in islets from MSG-VD rats, without any change in M3R, PKA or PKC expression. In conclusion, chronic oral VD supplementation of MSG-obese rats was able to prevent hyperinsulinaemia and IR, improving triacylglycerolaemia without modifying adiposity. A reduced cholinergic pancreatic effect, in response to VD, could be involved in the normalisation of plasma insulin levels, an event that appears to be independent of M3R and its downstream pathways.


Subject(s)
Adiposity/drug effects , Dietary Supplements , Insulin Secretion/drug effects , Vitamin D/pharmacology , Vitamins/pharmacology , Animals , Hypothalamus/metabolism , Rats
2.
J Biol Chem ; 292(8): 3466-3480, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28077579

ABSTRACT

Pancreatic beta cell failure is the central event leading to diabetes. Beta cells share many phenotypic traits with neurons, and proper beta cell function relies on the activation of several neuron-like transcription programs. Regulation of gene expression by alternative splicing plays a pivotal role in brain, where it affects neuronal development, function, and disease. The role of alternative splicing in beta cells remains unclear, but recent data indicate that splicing alterations modulated by both inflammation and susceptibility genes for diabetes contribute to beta cell dysfunction and death. Here we used RNA sequencing to compare the expression of splicing-regulatory RNA-binding proteins in human islets, brain, and other human tissues, and we identified a cluster of splicing regulators that are expressed in both beta cells and brain. Four of them, namely Elavl4, Nova2, Rbox1, and Rbfox2, were selected for subsequent functional studies in insulin-producing rat INS-1E, human EndoC-ßH1 cells, and in primary rat beta cells. Silencing of Elavl4 and Nova2 increased beta cell apoptosis, whereas silencing of Rbfox1 and Rbfox2 increased insulin content and secretion. Interestingly, Rbfox1 silencing modulates the splicing of the actin-remodeling protein gelsolin, increasing gelsolin expression and leading to faster glucose-induced actin depolymerization and increased insulin release. Taken together, these findings indicate that beta cells share common splicing regulators and programs with neurons. These splicing regulators play key roles in insulin release and beta cell survival, and their dysfunction may contribute to the loss of functional beta cell mass in diabetes.


Subject(s)
Insulin-Secreting Cells/cytology , RNA-Binding Proteins/metabolism , Alternative Splicing , Animals , Apoptosis , Cell Line , Cell Survival , Cells, Cultured , ELAV-Like Protein 4/genetics , ELAV-Like Protein 4/metabolism , Gene Expression Regulation , Glucose/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , Rats
3.
Eur J Nutr ; 57(4): 1471-1483, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28314963

ABSTRACT

PURPOSE: To evaluate the role of miR-124a in the regulation of genes involved in insulin exocytosis and its effects on the kinetics of insulin secretion in pancreatic islets from pregnant rats submitted to a low-protein diet. METHODS: Adult control non-pregnant (CNP) and control pregnant (CP) rats were fed a normal protein diet (17%), whereas low-protein non-pregnant (LPNP) and low-protein pregnant (LPP) rats were fed a low-protein diet (6%) from days 1 to 15 of pregnancy. Kinetics of the glucose-induced insulin release and measurement of [Ca2+]i in pancreatic islets were assessed by standard protocols. The miR-124a expression and gene transcriptions from pancreatic islets were determined by real-time polymerase chain reaction. RESULTS: In islets from LPP rats, the first phase of insulin release was abrogated. The AUC [Ca2+]i from the LPP group was lower compared with the other groups. miR-124a expression was reduced by a low-protein diet. SNAP-25 mRNA, protein expression, and Rab3A protein content were lower in the LPP rats than in CP rats. Syntaxin 1A and Kir6.2 mRNA levels were decreased in islets from low-protein rats compared with control rats, whereas their protein content was reduced in islets from pregnant rats. CONCLUSIONS: Loss of biphasic insulin secretion in islets from LPP rats appears to have resulted from reduced [Ca2+]i due, at least in part, to Kir6.2 underexpression and from the changes in exocytotic elements that are influenced either directly or indirectly by miR-124a.


Subject(s)
Diet, Protein-Restricted , Insulin/metabolism , Islets of Langerhans/metabolism , MicroRNAs/metabolism , Animals , Female , Glucose , Male , Pregnancy , Rats , Rats, Wistar
4.
Eur J Nutr ; 56(2): 705-713, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26621632

ABSTRACT

PURPOSE: Obesity is usually associated with low-grade inflammation, which impairs insulin action. The amino acid, taurine (TAU), regulates glucose homeostasis and lipid metabolism and presents anti-inflammatory actions. Here, we evaluated whether inflammatory markers are altered in the serum and retroperitoneal adipose tissue of monosodium glutamate (MSG) obese rats, supplemented or not with TAU. METHODS: Male Wistar rats received subcutaneous injections of MSG (4 mg/kg body weight/day, MSG group) or hypertonic saline (CTL) during the first 5 days of life. From 21 to 120 days of age, half of each of the MSG and CTL groups received 2.5 % TAU in their drinking water (CTAU and MTAU). RESULTS: At 120 days of age, MSG rats were obese and hyperinsulinemic. TAU supplementation reduced fat deposition without affecting insulinemia in MTAU rats. MSG rats presented increased pIκ-Bα/Iκ-Bα protein expression in the retroperitoneal adipose tissue. TAU supplementation decreased the ratio of pIκ-Bα/Iκ-Bα protein, possibly contributing to the increased Iκ-Bα content in MTAU adipose tissue. Furthermore, MSG obesity or supplementation did not alter TNF-α, IL-1ß or IL-6 content in adipose tissue. In contrast, MSG rats presented lower serum TNF-α, IL-4 and IL-10 concentrations, and these alterations were prevented by TAU treatment. CONCLUSION: MSG obesity in rats was not associated with alterations in pro-inflammatory markers in retroperitoneal fat stores; however, reductions in the serum concentrations of anti-inflammatory cytokines and of TNF-α were observed. TAU treatment decreased adiposity, and this effect was associated with the normalization of circulating TNF-α and IL-4 concentrations in MTAU rats.


Subject(s)
Anti-Obesity Agents/therapeutic use , Dietary Supplements , Gene Expression Regulation , Intra-Abdominal Fat/metabolism , NF-KappaB Inhibitor alpha/metabolism , Obesity/diet therapy , Taurine/therapeutic use , Adiposity , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Hyperinsulinism/diet therapy , Hyperinsulinism/etiology , Hyperinsulinism/immunology , Hyperinsulinism/metabolism , I-kappa B Proteins/agonists , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Injections, Subcutaneous , Interleukin-4/antagonists & inhibitors , Interleukin-4/blood , Interleukin-4/metabolism , Intra-Abdominal Fat/immunology , Male , NF-KappaB Inhibitor alpha/agonists , NF-KappaB Inhibitor alpha/genetics , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Phosphorylation , Protein Processing, Post-Translational , Rats, Wistar , Sodium Glutamate/administration & dosage , Sodium Glutamate/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
5.
Nucleic Acids Res ; 42(18): 11818-30, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25249621

ABSTRACT

Alternative splicing (AS) is a fundamental mechanism for the regulation of gene expression. It affects more than 90% of human genes but its role in the regulation of pancreatic beta cells, the producers of insulin, remains unknown. Our recently published data indicated that the 'neuron-specific' Nova1 splicing factor is expressed in pancreatic beta cells. We have presently coupled specific knockdown (KD) of Nova1 with RNA-sequencing to determine all splice variants and downstream pathways regulated by this protein in beta cells. Nova1 KD altered the splicing of nearly 5000 transcripts. Pathway analysis indicated that these genes are involved in exocytosis, apoptosis, insulin receptor signaling, splicing and transcription. In line with these findings, Nova1 silencing inhibited insulin secretion and induced apoptosis basally and after cytokine treatment in rodent and human beta cells. These observations identify a novel layer of regulation of beta cell function, namely AS controlled by key splicing regulators such as Nova1.


Subject(s)
Alternative Splicing , Insulin-Secreting Cells/metabolism , RNA-Binding Proteins/physiology , Animals , Apoptosis , Calcium/metabolism , Cytokines/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Insulin/metabolism , Nerve Tissue Proteins/metabolism , Neuro-Oncological Ventral Antigen , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Rats, Wistar , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
6.
Diabetologia ; 58(10): 2307-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26099855

ABSTRACT

AIMS/HYPOTHESIS: Proinflammatory cytokines contribute to beta cell damage in type 1 diabetes in part through activation of endoplasmic reticulum (ER) stress. In rat beta cells, cytokine-induced ER stress involves NO production and consequent inhibition of the ER Ca(2+) transporting ATPase sarco/endoplasmic reticulum Ca(2+) pump 2 (SERCA2B). However, the mechanisms by which cytokines induce ER stress and apoptosis in mouse and human pancreatic beta cells remain unclear. The purpose of this study is to elucidate the role of ER stress on cytokine-induced beta cell apoptosis in these three species and thus solve ongoing controversies in the field. METHODS: Rat and mouse insulin-producing cells, human pancreatic islets and human EndoC-ßH1 cells were exposed to the cytokines IL-1ß, TNF-α and IFN-γ, with or without NO inhibition. A global comparison of cytokine-modulated gene expression in human, mouse and rat beta cells was also performed. The chemical chaperone tauroursodeoxycholic acid (TUDCA) and suppression of C/EBP homologous protein (CHOP) were used to assess the role of ER stress in cytokine-induced apoptosis of human beta cells. RESULTS: NO plays a key role in cytokine-induced ER stress in rat islets, but not in mouse or human islets. Bioinformatics analysis indicated greater similarity between human and mouse than between human and rat global gene expression after cytokine exposure. The chemical chaperone TUDCA and suppression of CHOP or c-Jun N-terminal kinase (JNK) protected human beta cells against cytokine-induced apoptosis. CONCLUSIONS/INTERPRETATION: These observations clarify previous results that were discrepant owing to the use of islets from different species, and confirm that cytokine-induced ER stress contributes to human beta cell death, at least in part via JNK activation.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Insulin-Secreting Cells/drug effects , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Cytokines/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Nitric Oxide Synthase Type II/antagonists & inhibitors , Rats , Rats, Wistar , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor CHOP/pharmacology , omega-N-Methylarginine/pharmacology
7.
J Biol Chem ; 289(30): 20677-93, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24936061

ABSTRACT

Type 1 diabetes is an autoimmune disease with a strong inflammatory component. The cytokines interleukin-1ß and interferon-γ contribute to beta cell apoptosis in type 1 diabetes. These cytokines induce endoplasmic reticulum stress and the unfolded protein response (UPR), contributing to the loss of beta cells. IRE1α, one of the UPR mediators, triggers insulin degradation and inflammation in beta cells and is critical for the transition from "physiological" to "pathological" UPR. The mechanisms regulating inositol-requiring protein 1α (IRE1α) activation and its signaling for beta cell "adaptation," "stress response," or "apoptosis" remain to be clarified. To address these questions, we combined mammalian protein-protein interaction trap-based IRE1α interactome and functional genomic analysis of human and rodent beta cells exposed to pro-inflammatory cytokines to identify novel cytokine-induced regulators of IRE1α. Based on this approach, we identified N-Myc interactor (NMI) as an IRE1α-interacting/modulator protein in rodent and human pancreatic beta cells. An increased expression of NMI was detected in islets from nonobese diabetic mice with insulitis and in rodent or human beta cells exposed in vitro to the pro-inflammatory cytokines interleukin-1ß and interferon-γ. Detailed mechanistic studies demonstrated that NMI negatively modulates IRE1α-dependent activation of JNK and apoptosis in rodent and human pancreatic beta cells. In conclusion, by using a combined omics approach, we identified NMI induction as a novel negative feedback mechanism that decreases IRE1α-dependent activation of JNK and apoptosis in cytokine-exposed beta cells


Subject(s)
Endoribonucleases/metabolism , Insulin-Secreting Cells/metabolism , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology , Aged , Animals , Apoptosis/physiology , Endoribonucleases/genetics , HEK293 Cells , Humans , Insulin-Secreting Cells/cytology , Interferon-gamma/genetics , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Middle Aged , Multienzyme Complexes/genetics , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Wistar
8.
Amino Acids ; 46(9): 2123-36, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24866813

ABSTRACT

Pancreatic ß-cells are highly sensitive to suboptimal or excess nutrients, as occurs in protein-malnutrition and obesity. Taurine (Tau) improves insulin secretion in response to nutrients and depolarizing agents. Here, we assessed the expression and function of Cav and KATP channels in islets from malnourished mice fed on a high-fat diet (HFD) and supplemented with Tau. Weaned mice received a normal (C) or a low-protein diet (R) for 6 weeks. Half of each group were fed a HFD for 8 weeks without (CH, RH) or with 5% Tau since weaning (CHT, RHT). Isolated islets from R mice showed lower insulin release with glucose and depolarizing stimuli. In CH islets, insulin secretion was increased and this was associated with enhanced KATP inhibition and Cav activity. RH islets secreted less insulin at high K(+) concentration and showed enhanced KATP activity. Tau supplementation normalized K(+)-induced secretion and enhanced glucose-induced Ca(2+) influx in RHT islets. R islets presented lower Ca(2+) influx in response to tolbutamide, and higher protein content and activity of the Kir6.2 subunit of the KATP. Tau increased the protein content of the α1.2 subunit of the Cav channels and the SNARE proteins SNAP-25 and Synt-1 in CHT islets, whereas in RHT, Kir6.2 and Synt-1 proteins were increased. In conclusion, impaired islet function in R islets is related to higher content and activity of the KATP channels. Tau treatment enhanced RHT islet secretory capacity by improving the protein expression and inhibition of the KATP channels and enhancing Synt-1 islet content.


Subject(s)
Calcium/metabolism , Dietary Fats/pharmacology , Dietary Supplements , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Malnutrition/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Taurine/pharmacology , Animals , Humans , Insulin Secretion , Male , Mice , Synaptosomal-Associated Protein 25/metabolism
9.
Amino Acids ; 41(4): 901-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21042817

ABSTRACT

The aim of the present study was to evaluate the preventive effects of taurine (TAU) supplementation upon monosodium glutamate (MSG)-induced obesity. Rats treated during the first 5 days of life with MSG or saline were distributed into the following groups: control (CTL), CTL-treated with TAU (CTAU), MSG and MSG-supplemented with TAU (MTAU). CTAU and MTAU received 2.5% of TAU in their drinking water from 21 to 90 days of life. At the end of treatment, MSG and MTAU rats were hyperinsulinemic, glucose intolerant and insulin resistant, as judged by the HOMA index. MSG and MTAU rat islets secreted more insulin at 16.7 mM glucose compared to CTL. MSG rats also showed higher triglycerides (TG) and non-esterified fatty acids (NEFA) plasma levels, Lee Index, retroperitoneal and periepidydimal fat pads, compared with CTL, whereas plasma lipid concentrations and fat depots were lower in MTAU, compared with MSG rats. In addition, MSG rats had a higher liver TG content compared with CTL. TAU decreased liver TG content in both supplemented groups, but fat content only in MTAU rats. TAU supplementation did not change glucose homeostasis, insulin secretion and action, but reduced plasma and liver lipid levels in MSG rats.


Subject(s)
Adipose Tissue/drug effects , Lipids/blood , Obesity/metabolism , Taurine/pharmacology , Adipose Tissue/metabolism , Animals , Blood Glucose/analysis , Body Weight/drug effects , Cholesterol/blood , Disease Models, Animal , Fatty Acids, Nonesterified/metabolism , Glucose/metabolism , Hyperinsulinism/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Liver/drug effects , Liver/metabolism , Male , Obesity/chemically induced , Rats , Rats, Wistar , Sodium Glutamate , Taurine/blood , Triglycerides/blood
10.
J Mol Endocrinol ; 61(1): 25-36, 2018 07.
Article in English | MEDLINE | ID: mdl-29632026

ABSTRACT

Type 1 diabetes is caused by an autoimmune assault that induces progressive beta-cell dysfunction and dead. Pro-inflammatory cytokines, such as interleukin 1 beta (IL1B), tumor necrosis factor (TNF) and interferon gamma (IFNG) contribute for beta-cell death, which involves the activation of the nuclear factor kappa B (NFκB) and c- Jun N-terminal kinase (JNK). Prolactin (PRL), a physiological mediator for beta-cell proliferation, was shown to protect beta cells against cytokines pro-apoptotic effects. We presently investigated the mechanisms involved in the protective effects of prolactin against cytokine-induced beta-cell death. The findings obtained indicate that STAT3 activation is involved in the anti-apoptotic role of PRL in rat beta cells. PRL prevents the activation of JNK via AKT and promotes a shift from expression of pro- to anti-apoptotic proteins downstream of the JNK cascade. Furthermore, PRL partially prevents the activation of NFκB and the transcription of its target genes IkBa, Fas, Mcp1, A20 and Cxcl10 and also decreases NO production. On the other hand, the pro-survival effects of PRL do not involve modulation of cytokine-induced endoplasmic reticulum stress. These results suggest that the beneficial effects of PRL in beta cells involve augmentation of anti-apoptotic mechanisms and, at the same time, reduction of pro-apoptotic effectors, rendering beta cells better prepared to deal with inflammatory insults. The better understanding of the pro-survival mechanisms modulated by PRL in beta cells can provide tools to prevent cell demise during an autoimmune attack or following islet transplantation.


Subject(s)
Apoptosis/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Prolactin/pharmacology , Animals , Blotting, Western , Cells, Cultured , Female , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
11.
Oxid Med Cell Longev ; 2017: 1838679, 2017.
Article in English | MEDLINE | ID: mdl-28163820

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the principal manifestation of liver disease in obesity and metabolic syndrome. By comparing hypertriglyceridemic transgenic mice expressing apolipoprotein (apo) CIII with control nontransgenic (NTg) littermates, we demonstrated that overexpression of apoCIII, independent of a high-fat diet (HFD), produces NAFLD-like features, including increased liver lipid content; decreased antioxidant power; increased expression of TNFα, TNFα receptor, cleaved caspase-1, and interleukin-1ß; decreased expression of adiponectin receptor-2; and increased cell death. This phenotype is aggravated and additional NAFLD features are differentially induced in apoCIII mice fed a HFD. HFD induced glucose intolerance together with increased gluconeogenesis, indicating hepatic insulin resistance. Additionally, the HFD led to marked increases in plasma TNFα (8-fold) and IL-6 (60%) in apoCIII mice. Cell death signaling (Bax/Bcl2), effector (caspase-3), and apoptosis were augmented in apoCIII mice regardless of whether a HFD or a low-fat diet was provided. Fenofibrate treatment reversed several of the effects associated with diet and apoCIII expression but did not normalize inflammatory traits even when liver lipid content was fully corrected. These results indicate that apoCIII and/or hypertriglyceridemia plays a major role in liver inflammation and cell death, which in turn increases susceptibility to and the severity of diet-induced NAFLD.


Subject(s)
Apolipoprotein C-III/biosynthesis , Hypertriglyceridemia/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Apolipoprotein C-III/metabolism , Cell Death/physiology , Diet, High-Fat , Inflammation/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Transgenic , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
12.
J Endocrinol ; 229(3): 221-32, 2016 06.
Article in English | MEDLINE | ID: mdl-27000684

ABSTRACT

The aim of this study was to investigate the insulin clearance in diet-induced obese (DIO) mice submitted to acute endurance exercise (3h of treadmill exercise at 60-70% VO2max). Glucose-stimulated insulin secretion in isolated islets; ipGTT; ipITT; ipPTT; in vivo insulin clearance; protein expression in liver, skeletal muscle, and adipose tissue (insulin degrading enzyme (IDE), insulin receptor subunitß(IRß), phospho-Akt (p-Akt) and phospho-AMPK (p-AMPK)), and the activity of IDE in the liver and skeletal muscle were accessed. In DIO mice, acute exercise reduced fasting glycemia and insulinemia, improved glucose and insulin tolerance, reduced hepatic glucose production, and increased p-Akt protein levels in liver and skeletal muscle and p-AMPK protein levels in skeletal muscle. In addition, insulin secretion was reduced, whereas insulin clearance and the expression of IDE and IRß were increased in liver and skeletal muscle. Finally, IDE activity was increased only in skeletal muscle. In conclusion, we propose that the increased insulin clearance and IDE expression and activity, primarily, in skeletal muscle, constitute an additional mechanism, whereby physical exercise reduces insulinemia in DIO mice.


Subject(s)
Insulin/metabolism , Obesity/metabolism , Obesity/therapy , Physical Conditioning, Animal/physiology , AMP-Activated Protein Kinases/metabolism , Animals , C-Peptide/blood , Diet, High-Fat/adverse effects , Glucose/metabolism , Insulin/blood , Insulysin/metabolism , Liver/metabolism , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/metabolism , Obesity/etiology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism
13.
PLoS One ; 11(7): e0160239, 2016.
Article in English | MEDLINE | ID: mdl-27467214

ABSTRACT

The effects of exercise on insulin clearance and IDE expression are not yet fully elucidated. Here, we have explored the effect of acute exercise on insulin clearance and IDE expression in lean mice. Male Swiss mice were subjected to a single bout of exercise on a speed/angle controlled treadmill for 3-h at approximately 60-70% of maximum oxygen consumption. As expected, acute exercise reduced glycemia and insulinemia, and increased insulin tolerance. The activity of AMPK-ACC, but not of IR-Akt, pathway was increased in the liver and skeletal muscle of trained mice. In an apparent contrast to the reduced insulinemia, glucose-stimulated insulin secretion was increased in isolated islets of these mice. However, insulin clearance was increased after acute exercise and was accompanied by increased expression of the insulin-degrading enzyme (IDE), in the liver and skeletal muscle. Finally, C2C12, but not HEPG2 cells, incubated at different concentrations of 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR) for 3-h, showed increased expression of IDE. In conclusion, acute exercise increases insulin clearance, probably due to an augmentation of IDE expression in the liver and skeletal muscle. The elevated IDE expression, in the skeletal muscle, seems to be mediated by activation of AMPK-ACC pathway, in response to exercise. We believe that the increase in the IDE expression, comprise a safety measure to maintain glycemia at or close to physiological levels, turning physical exercise more effective and safe.


Subject(s)
Insulin/metabolism , Liver/enzymology , Muscle, Skeletal/enzymology , Physical Conditioning, Animal , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Enzyme Activation , Hep G2 Cells , Humans , Hydrolysis , Insulysin , Islets of Langerhans/metabolism , Male , Mice , Oxygen Consumption
SELECTION OF CITATIONS
SEARCH DETAIL