Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biol Trace Elem Res ; 180(2): 223-232, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28378114

ABSTRACT

The objective of this study was to investigate the effects of peroral administration of chromium-enriched yeast on glucose tolerance in Holstein calves, assessed by insulin signaling pathway molecule determination and intravenous glucose tolerance test (IVGTT). Twenty-four Holstein calves, aged 1 month, were chosen for the study and divided into two groups: the PoCr group (n = 12) that perorally received 0.04 mg of Cr/kg of body mass daily, for 70 days, and the NCr group (n = 12) that received no chromium supplementation. Skeletal tissue samples from each calf were obtained on day 0 and day 70 of the experiment. Chromium supplementation increased protein content of the insulin ß-subunit receptor, phosphorylation of insulin receptor substrate 1 at Tyrosine 632, phosphorylation of Akt at Serine 473, glucose transporter-4, and AMP-activated protein kinase in skeletal muscle tissue, while phosphorylation of insulin receptor substrate 1 at Serine 307 was not affected by chromium treatment. Results obtained during IVGTT, which was conducted on days 0, 30, 50, and 70, suggested an increased insulin sensitivity and, consequently, a better utilization of glucose in the PoCr group. Lower basal concentrations of glucose and insulin in the PoCr group on days 30 and 70 were also obtained. Our results indicate that chromium supplementation improves glucose utilization in calves by enhancing insulin intracellular signaling in the skeletal muscle tissue.


Subject(s)
Animal Nutritional Physiological Phenomena , Chromium/therapeutic use , Glucose Intolerance/veterinary , Insulin Resistance , Muscle, Skeletal/metabolism , Signal Transduction , Yeast, Dried/therapeutic use , Animals , Animals, Inbred Strains , Biopsy/veterinary , Cattle , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Dairying , Female , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Intolerance/prevention & control , Glucose Transporter Type 4/agonists , Glucose Transporter Type 4/metabolism , Hamstring Muscles , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/metabolism , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/agonists , Receptor, Insulin/metabolism , Weaning
2.
J. physiol. biochem ; 72(2): 345-352, jun. 2016. tab, graf, ilus
Article in English | IBECS | ID: ibc-168278

ABSTRACT

The aim of this present study was to investigate the effect of ursolic acid (UA) and rosiglitazone (RSG) on insulin sensitivity and proximal insulin signaling pathways in high-fat diet (HFD)-fed C57/BL/6J mice. Male C57BL/6J mice were fed either normal diet or HFD for 10 weeks, after which animals in each dietary group were divided into the following six groups (normal diet, normal diet plus UA and RSG, HFD alone, HFD plus UA, HFD plus RSG, and HFD plus UA and RSG) for the next 5 weeks. UA (5 mg/kg BW) and RSG (4 mg/kg BW) were administered as suspensions directly into the stomach using a gastric tube. The HFD diet elevated fasting plasma glucose, insulin, and homeostasis model assessment index. The expression of insulin receptor substrate (IRS)-1, phosphoinositide 3-kinase (PI3-kinase), Akt, and glucose transporter (GLUT) 4 were determined by Western blot analyses. The results demonstrated that combination treatment (UA/RSG) ameliorated HFD-induced glucose intolerance and insulin resistance by improving the homeostatic model assessment (HOMA) index. Further, combination treatment (UA/RSG) stimulated the IRS-1, PI3-kinase, Akt, and GLUT 4 translocation. These results strongly suggest that combination treatment (UA/RSG) activates IRS-PI3-kinase-Akt-dependent signaling pathways to induce GLUT 4 translocation and increases the expression of insulin receptor to improve glucose intolerance (AU)


No disponible


Subject(s)
Animals , Male , Diabetes Mellitus, Type 2/drug therapy , Insulin Resistance , Muscle, Skeletal , Hypoglycemic Agents/therapeutic use , Triterpenes/therapeutic use , Thiazolidinediones/therapeutic use , Insulin Receptor Substrate Proteins/agonists , Anti-Obesity Agents , Antioxidants/therapeutic use , Diet, High-Fat/adverse effects , Drug Therapy, Combination/adverse effects , Weight Gain , Obesity/metabolism , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism
3.
J Physiol Biochem ; 72(2): 345-52, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27090933

ABSTRACT

The aim of this present study was to investigate the effect of ursolic acid (UA) and rosiglitazone (RSG) on insulin sensitivity and proximal insulin signaling pathways in high-fat diet (HFD)-fed C57/BL/6J mice. Male C57BL/6J mice were fed either normal diet or HFD for 10 weeks, after which animals in each dietary group were divided into the following six groups (normal diet, normal diet plus UA and RSG, HFD alone, HFD plus UA, HFD plus RSG, and HFD plus UA and RSG) for the next 5 weeks. UA (5 mg/kg BW) and RSG (4 mg/kg BW) were administered as suspensions directly into the stomach using a gastric tube. The HFD diet elevated fasting plasma glucose, insulin, and homeostasis model assessment index. The expression of insulin receptor substrate (IRS)-1, phosphoinositide 3-kinase (PI3-kinase), Akt, and glucose transporter (GLUT) 4 were determined by Western blot analyses. The results demonstrated that combination treatment (UA/RSG) ameliorated HFD-induced glucose intolerance and insulin resistance by improving the homeostatic model assessment (HOMA) index. Further, combination treatment (UA/RSG) stimulated the IRS-1, PI3-kinase, Akt, and GLUT 4 translocation. These results strongly suggest that combination treatment (UA/RSG) activates IRS-PI3-kinase-Akt-dependent signaling pathways to induce GLUT 4 translocation and increases the expression of insulin receptor to improve glucose intolerance.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin Receptor Substrate Proteins/agonists , Insulin Resistance , Muscle, Skeletal/drug effects , Thiazolidinediones/therapeutic use , Triterpenes/therapeutic use , Animals , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/therapeutic use , Antioxidants/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Drug Therapy, Combination/adverse effects , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/adverse effects , Insulin Receptor Substrate Proteins/metabolism , Male , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/chemically induced , Obesity/complications , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Rosiglitazone , Second Messenger Systems/drug effects , Thiazolidinediones/adverse effects , Triterpenes/adverse effects , Weight Gain/drug effects , Ursolic Acid
4.
J Biol Chem ; 291(9): 4547-60, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26702053

ABSTRACT

The ubiquitous phosphatidylinositol 3-kinase (PI3K) signaling pathway regulates many cellular functions. However, the mechanism by which G protein-coupled receptors (GPCRs) signal to activate PI3K is poorly understood. We have used ovarian granulosa cells as a model to investigate this pathway, based on evidence that the GPCR agonist follicle-stimulating hormone (FSH) promotes the protein kinase A (PKA)-dependent phosphorylation of insulin receptor substrate 1 (IRS1) on tyrosine residues that activate PI3K. We report that in the absence of FSH, granulosa cells secrete a subthreshold concentration of insulin-like growth factor-1 (IGF-1) that primes the IGF-1 receptor (IGF-1R) but fails to promote tyrosine phosphorylation of IRS1. FSH via PKA acts to sensitize IRS1 to the tyrosine kinase activity of the IGF-1R by activating protein phosphatase 1 (PP1) to promote dephosphorylation of inhibitory Ser/Thr residues on IRS1, including Ser(789). Knockdown of PP1ß blocks the ability of FSH to activate PI3K in the presence of endogenous IGF-1. Activation of PI3K thus requires both PKA-mediated relief of IRS1 inhibition and IGF-1R-dependent tyrosine phosphorylation of IRS1. Treatment with FSH and increasing concentrations of exogenous IGF-1 triggers synergistic IRS1 tyrosine phosphorylation at PI3K-activating residues that persists downstream through protein kinase B (AKT) and FOXO1 (forkhead box protein O1) to drive synergistic expression of genes that underlies follicle maturation. Based on the ability of GPCR agonists to synergize with IGFs to enhance gene expression in other cell types, PP1 activation to relieve IRS1 inhibition may be a more general mechanism by which GPCRs act with the IGF-1R to activate PI3K/AKT.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Follicle Stimulating Hormone/metabolism , Granulosa Cells/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Protein Phosphatase 1/metabolism , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation , Female , Granulosa Cells/cytology , Humans , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Insulin-Like Growth Factor I/genetics , Mutation , Phosphatidylinositol 3-Kinase/chemistry , Phosphorylation , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/genetics , Protein Processing, Post-Translational , RNA Interference , Rats, Sprague-Dawley , Receptor, IGF Type 1/agonists , Receptor, IGF Type 1/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Tyrosine/metabolism
5.
Mol Med Rep ; 12(5): 6555-60, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26352439

ABSTRACT

Previous studies have indicated that 4­hydroxyisoleucine (4­HIL) improves insulin resistance, however, the underlying mechanisms remain to be elucidated. In the present study, the molecular mechanisms underlying how 4­HIL improves insulin resistance in hepatocytes were examined. HepG2 cells were co­cultured with insulin and a high glucose concentration to obtain insulin­resistant (IR) HepG2 cells. Insulin sensitivity was determined by measuring the glucose uptake rate. The IR HepG2 cells were treated with different concentrations of 4­HIL to determine its effect on IR Hep2 cells. The levels of tumor necrosis factor­α (TNF­α) were measured by an enzyme­linked immunosorbent assay and protein levels of TNF­α converting enzyme (TACE)/tissue inhibitor of metalloproteinase 3 (TIMP3), insulin receptor substrate (IRS)­1, IRS­2, phosphorylated (p)­IRS­1 (Ser307) and glucose transporter type 4 (GLUT4) were measured by western blot analysis. The results of the present study demonstrated that insulin­induced glucose uptake was reduced in IR HepG2 cells; however, this reduction was reversed by 4­HIL in a dose­dependent manner. 4­HIL achieved this effect by downregulating the expression of TNF­α and TACE, and upregulating the expression of TIMP3 in IR HepG2 cells. In addition, 4­HIL increased the expression of the insulin transduction regulators IRS­1 and GLUT4, and decreased the expression of p­IRS­1 (Ser307), without affecting the expression of IRS­2. The present study suggests that 4­HIL improved insulin resistance in HepG2 cells by the following mechanisms: 4­HIL reduced TNF­α levels by affecting the protein expression of the TACE/TIMP3 system and 4­HIL stimulated the expression of IRS­1 and GLUT4, but inhibited the expression of p­IRS­1 (Ser307).


Subject(s)
Glucose/pharmacology , Insulin Resistance , Insulin/pharmacology , Isoleucine/analogs & derivatives , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Dose-Response Relationship, Drug , Gene Expression Regulation , Glucose/metabolism , Glucose Transporter Type 4/agonists , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Hep G2 Cells , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Isoleucine/isolation & purification , Isoleucine/pharmacology , Phosphorylation/drug effects , Signal Transduction/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Trigonella/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Nutr Food Res ; 59(8): 1472-81, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25944785

ABSTRACT

SCOPE: Insulin resistance represents an independent risk factor for metabolic and cardiovascular diseases. Researchers have been interested in identifying active harmless compounds, as many insulin-sensitizing drugs have shown unwanted side-effects. It has been demonstrated that anthocyanins and one of their representative metabolites, protocatechuic acid (PCA), ameliorate hyperglycemia, and insulin sensitivity. This study investigated the mechanism of action of PCA responsible for the glucose uptake upregulation. METHODS AND RESULTS: In human visceral adipocytes, PCA stimulated insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation (+40% with respect to untreated cells) and the downstream events, i.e. phosphoinositide 3-kinase binding to IRS-1 and Akt phosphorylation (+100%, +180%, respectively, with respect to untreated cells). The insulin-like activity of PCA seemed to be mediated by insulin receptor since by inhibiting its autophosphorylation, the PCA effects were completely abolished. Furthermore, PCA was able to activate adenosine monophosphate-activated protein kinase, a serine/threonine kinase whose activation elicits insulin-sensitizing effects. CONCLUSION: This study showed that PCA stimulates the insulin signaling pathway in human adipocytes increasing GLUT4 translocation and glucose uptake. Decreasing insulin resistance is a most desirable aim to be reached for an effective therapeutic/preventive action against metabolic syndrome and type 2 diabetes. Identifying specific food/food components able to improve glucose metabolism can offer an attractive, novel, and economical strategy.


Subject(s)
Hydroxybenzoates/metabolism , Hypoglycemic Agents/metabolism , Insulin Receptor Substrate Proteins/agonists , Insulin Resistance , Intra-Abdominal Fat/metabolism , Protein Processing, Post-Translational , Signal Transduction , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/metabolism , Absorption, Physiological/drug effects , Cells, Cultured , Dietary Supplements , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Glucose Transporter Type 4/agonists , Glucose Transporter Type 4/metabolism , Humans , Hydroxybenzoates/antagonists & inhibitors , Hypoglycemic Agents/antagonists & inhibitors , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Insulin Receptor Substrate Proteins/metabolism , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/drug effects , Lipoproteins, LDL/adverse effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
7.
Diabetes Obes Metab ; 16 Suppl 1: 33-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25200294

ABSTRACT

Dysregulation of hepatic glucose uptake (HGU) and inability of insulin to suppress hepatic glucose production (HGP) contribute to hyperglycaemia in patients with type 2 diabetes (T2D). Growing evidence suggests that insulin can inhibit HGP not only through a direct effect on the liver but also through a mechanism involving the brain. Yet, the notion that insulin action in the brain plays a physiological role in the control of HGP continues to be controversial. Although studies in dogs suggest that the direct hepatic effect of insulin is sufficient to explain day-to-day control of HGP, a surprising outcome has been revealed by recent studies in mice, investigating whether the direct hepatic action of insulin is necessary for normal HGP: when the hepatic insulin signalling pathway was genetically disrupted, HGP was maintained normally even in the absence of direct input from insulin. Here, we present evidence that points to a potentially important role of the brain in the physiological control of both HGU and HGP in response to input from insulin as well as other hormones and nutrients.


Subject(s)
Brain/metabolism , Feedback, Physiological , Gluconeogenesis , Glycolysis , Islets of Langerhans/metabolism , Liver/metabolism , Models, Biological , Animals , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/metabolism , Insulin Secretion , Islets of Langerhans/innervation , Liver/innervation , Neurons/metabolism , Signal Transduction
8.
Mol Cell Endocrinol ; 384(1-2): 1-11, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24394757

ABSTRACT

Early in life, leptin plays a crucial role in hypothalamic neural organization. Leptin, most likely, controls neural gene expression conferring then specific phenotype regarding energy homeostasis. MicroRNAs are new regulators for several physiological functions, including the regulation of metabolism. However, the impact of leptin on hypothalamic microRNA patterns remains unknown. Here, we demonstrate that miR-200a, miR-200b and miR-429 are up-regulated in the hypothalamus of genetically obese and leptin deficient ob/ob mice. Leptin treatment down-regulates these miRNAs in ob/ob hypothalamus. The hypothalamic silencing of miR-200a increased the expression level of leptin receptor and insulin receptor substrate 2, reduced body weight gain, and restored liver insulin responsiveness. In addition, the overexpression of pre-miR-200a in a human neuroblastoma cell line impaired insulin and leptin signaling. These findings link the alteration of leptin and insulin signaling to the up-regulation of hypothalamic miR-200a which could be a new target for treatment of obesity.


Subject(s)
Hypothalamus/metabolism , Insulin/metabolism , Leptin/genetics , MicroRNAs/genetics , Obesity/genetics , Signal Transduction , Animals , Cell Line, Tumor , Feeding Behavior , Gene Expression Regulation , Humans , Hypothalamus/physiopathology , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Leptin/deficiency , Liver/metabolism , Liver/physiopathology , Male , Mice , Mice, Obese , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neurons/cytology , Neurons/metabolism , Obesity/metabolism , Obesity/physiopathology , Receptors, Leptin/agonists , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Weight Gain
9.
Food Chem Toxicol ; 59: 373-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23810794

ABSTRACT

Endocrine disrupting chemicals (EDCs) and estrogens appear to promote development of estrogen-dependent cancers, including breast and ovarian carcinomas. In this study, we evaluated the cell viability effect of BPA on BG-1 human ovarian cancer cells, along with the growth inhibitory effect of resveratrol (trans-3,4,5-trihydroxystilbene; RES), a naturally occurring phytoestrogen. In addition, we investigated the underlying mechanism(s) of BPA and RES in regulating the interaction between estrogen receptor alpha (ERα) and insulin-like growth factor-1 receptor (IGF-1R) signals, a non- genomic pathway induced by 17ß-estradiol (E2). BPA induced a significant increase in BG-1 cell growth and up-regulated mRNA levels of ERα and IGF-1R. In parallel with its mRNA level, the protein expression of ERα was induced, and phosphorylated insulin receptor substrate-1 (p-IRS-1), phosphorylated Akt1/2/3, and cyclin D1 were increased by BPA or E2. However, RES effectively reversed the BG-1 cell proliferation induced by E2 or BPA by inversely down-regulating the expressions of ERα, IGF-1R, p-IRS-1, and p-Akt1/2/3, and cyclin D1 at both transcriptional and translational levels. Taken together, these results suggest that RES is a novel candidate for prevention of tumor progression caused by EDCs, including BPA via effective inhibition of the cross-talk of ERα and IGF-1R signaling pathways.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Phytoestrogens/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Signal Transduction/drug effects , Stilbenes/pharmacology , Benzhydryl Compounds/antagonists & inhibitors , Benzhydryl Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/agonists , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Down-Regulation/drug effects , Estradiol/chemistry , Estradiol/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens, Non-Steroidal/antagonists & inhibitors , Estrogens, Non-Steroidal/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Phenols/antagonists & inhibitors , Phenols/pharmacology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Receptor, IGF Type 1/agonists , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Resveratrol
10.
J Nutr Sci Vitaminol (Tokyo) ; 59(5): 393-401, 2013.
Article in English | MEDLINE | ID: mdl-24418873

ABSTRACT

Our group recently demonstrated that simultaneous administration of trehalose with a high-fat diet (HFD) suppresses adipocyte hypertrophy and mitigates insulin resistance in mice. For the present study, we hypothesized that similar effects of trehalose would be observed in mice with previously-established obesity. Obese mice were fed a HFD and drinking water containing 0.3 or 2.5% (weight/volume) trehalose or distilled water (DW) ad libitum for 8 wk. After 7 wk intake of a HFD and trehalose, fasting serum insulin levels and homeostasis model assessment-insulin resistance (HOMA-IR) in the 0.3% Tre/HFD group were significantly lower than those in the DW/HFD group (p<0.05). After 8 wk of treatment, mesenteric adipocytes in the 0.3% Tre/HFD group showed significantly less hypertrophy than those in the DW/HFD group. Mechanistic analysis indicated that levels of high molecular weight (HMW) adiponectin in the serum of the 0.3% Tre/HFD group were significantly higher than those in the DW/HFD group. The expression levels of insulin receptor substrate-1 (IRS-1) and insulin receptor substrate-2 (IRS-2) messenger RNA (mRNA) in muscle were also significantly increased by trehalose intake. Our data therefore suggest that administration of trehalose to obese mice mitigates insulin resistance by suppressing adipocyte hypertrophy and increasing serum HMW adiponectin, resulting in upregulation of IRS-1, and IRS-2 expression in muscle. These results further suggest that trehalose is a functional saccharide that may be used to prevent the progression of insulin resistance.


Subject(s)
Dietary Supplements , Hyperinsulinism/diet therapy , Insulin Resistance , Intra-Abdominal Fat/pathology , Metabolic Syndrome/prevention & control , Obesity/physiopathology , Trehalose/therapeutic use , Adiponectin/agonists , Adiponectin/blood , Adiponectin/metabolism , Adiposity , Animals , Diet, High-Fat/adverse effects , Female , Gene Expression Regulation , Hyperinsulinism/etiology , Hyperinsulinism/pathology , Hyperinsulinism/physiopathology , Hypertrophy , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Intra-Abdominal Fat/metabolism , Metabolic Syndrome/etiology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/etiology , Random Allocation , Trehalose/administration & dosage
11.
Can J Physiol Pharmacol ; 90(11): 1544-52, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23181282

ABSTRACT

This study investigates the effects of astaxanthin (ASX) on insulin signaling and glucose metabolism in the liver of mice fed a high fat and high fructose diet (HFFD). Adult male Mus musculus mice of body mass 25-30 g were fed either normal chow or the HFFD. After 15 days, mice in each group were subdivided among 2 smaller groups and treated with ASX (2 mg·(kg body mass)⁻¹) in olive oil for 45 days. At the end of 60 days, HFFD-fed mice displayed insulin resistance while ASX-treated HFFD animals showed marked improvement in insulin sensitivity parameters. ASX treatment normalized the activities of hexokinase, pyruvate kinase, glucose-6-phosphatase, fructose-1,6-bisphosphatase, glycogen phosphorylase, and increased glycogen reserves in the liver. Liver tissue from ASX-treated HFFD-fed animals showed increased tyrosine phosphorylation and decreased serine phosphorylation of insulin receptor substrates (IRS)-1 and -2. ASX increased IRS 1/2 and phosphatidylinositol 3-kinase (PI3K) association and serine phosphorylation of Akt. In addition, ASX decreased HFFD-induced serine kinases (c-jun N-terminal kinase-1 and extracellular signal-regulated kinase-1). The results suggest that ASX treatment promotes the IRS-PI3K-Akt pathway of insulin signaling by decreasing serine phosphorylation of IRS proteins, and improves glucose metabolism by modulating metabolic enzymes.


Subject(s)
Hyperglycemia/diet therapy , Hypoglycemic Agents/therapeutic use , Insulin Receptor Substrate Proteins/agonists , Insulin Resistance , Liver/drug effects , Signal Transduction/drug effects , Animals , Antioxidants/therapeutic use , Dietary Supplements , Hyperglycemia/etiology , Insulin Receptor Substrate Proteins/metabolism , Liver/enzymology , Liver/metabolism , Liver Glycogen/metabolism , Male , Mice , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Serine/metabolism , Tyrosine/metabolism , Xanthophylls/therapeutic use
12.
Clin Exp Pharmacol Physiol ; 39(9): 759-64, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22670687

ABSTRACT

1. Insulin resistance (IR) is crucially involved in the pathophysiology of metabolic syndrome (MS). The aim of the present study was to investigate the effects of simvastatin on IR in rats with MS. 2. A rat model of MS was established and myocardial damage was examined by transmission electron microscopy. Twenty-two MS rats were divided into two groups of 11 rats each: (i) an MS group; and (ii) a simvastatin-treated MS. Ten Wistar rats were used as controls. The phosphorylation of myosin phosphatase target subunit 1 (MYPT-1), insulin receptor substrate 1 (IRS-1) and Akt were analysed by immunohistochemistry and western blotting. 3. Insulin resistance-induced MS was associated with a significant increase in Rho kinase (ROCK) activity and inhibition of the phosphatidylinositol 3-kinase (PI3-K)/Akt pathway. Decreased levels of phosphorylated (p-) MYPT-1 and p-IRS-1 (Ser³°7) and increased levels of p-Akt were found in hearts from the MS + simvastatin compared with the MS group. These results suggest that simvastatin reduces ROCK activity and increases Akt activity. 4. Simvastatin exerts cardioprotective effects and improves IR, which can be attributed, at least in part, to the inhibition of ROCK and activation of PI3-K/Akt.


Subject(s)
Cardiotonic Agents/therapeutic use , Heart Diseases/prevention & control , Insulin Resistance , Metabolic Syndrome/drug therapy , Myocardium/enzymology , Simvastatin/therapeutic use , rho-Associated Kinases/antagonists & inhibitors , Animals , Heart/drug effects , Heart/physiopathology , Heart Diseases/etiology , Heart Diseases/metabolism , Heart Diseases/physiopathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/metabolism , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Mitochondrial Swelling/drug effects , Myocardium/metabolism , Myocardium/ultrastructure , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Protein Phosphatase 1/metabolism , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats , Rats, Wistar , Vacuoles/drug effects , Vacuoles/ultrastructure , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...