Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Diabetol Int ; 14(3): 243-251, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37397908

ABSTRACT

Pioglitazone ameliorates liver dysfunction in type 2 diabetes (T2D) patients with non-alcoholic fatty liver disease (NAFLD); however, its efficacy in T2D patients with alcoholic fatty liver disease (AFLD) is unclear. Here, we conducted a retrospective single-center trial investigating whether pioglitazone ameliorates liver dysfunction in T2D patients with AFLD. T2D patients (n = 100) receiving 3 months of additional pioglitazone were divided into those with or without fatty liver (FL), and those with FL were further classified into AFLD (n = 21) and NAFLD (n = 57) groups. The effects of pioglitazone were compared across groups using medical record data on body weight changes; HbA1c, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and gamma-glutamyl transpeptidase (γ-GTP) levels; and fibrosis-4 (FIB-4) index. The pioglitazone dose (mean dose: 10.6 ± 4.6 mg/day) did not affect weight gain but significantly decreased the HbA1c level in patients with or without FL (P < 0.01 and P < 0.05, respectively). The decrease in HbA1c level was significantly more pronounced in patients with FL than in those without FL (P < 0.05). In patients with FL, the HbA1c, AST, ALT, and γ-GTP levels significantly decreased after pioglitazone treatment than before (P < 0.01). The AST and ALT levels, but not the γ-GTP level, and the FIB-4 index significantly decreased after pioglitazone addition in the AFLD group, similar to that in the NAFLD group (P < 0.05 and P < 0.01, respectively). Similar effects were observed following low-dose pioglitazone treatment (≤ 7.5 mg/day) (P < 0.05) in T2D patients with AFLD and NAFLD. These results suggest that pioglitazone may be also an effective treatment option for T2D patients with AFLD.

2.
Intern Med ; 62(20): 2981-2988, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-36858518

ABSTRACT

Mineralocorticoid deficiency (MD) with hyperkalemia is an important complication of adrenalectomy in patients with primary aldosteronism (PA). We herein report a 52-year-old man with refractory hypertension, hypokalemia, and severe renal dysfunction due to PA caused by a right adrenal adenoma. His estimated glomerular filtration rate (eGFR) transiently increased immediately after adrenalectomy but then gradually declined, and he developed hyperkalemia. A postoperative endocrine examination revealed MD. Considering the patient's hypertension and severe renal dysfunction, we administered hydrocortisone instead of fludrocortisone, which improved the hyperkalemia and stopped the decline in the eGFR. Alternative therapy with hydrocortisone may be useful in such patients with MD.


Subject(s)
Hyperaldosteronism , Hyperkalemia , Hypertension , Kidney Diseases , Male , Humans , Middle Aged , Hyperkalemia/etiology , Hydrocortisone/therapeutic use , Hyperaldosteronism/drug therapy , Hyperaldosteronism/surgery , Hyperaldosteronism/complications , Hypertension/etiology , Adrenalectomy , Kidney Diseases/complications , Aldosterone
4.
Endocr J ; 62(6): 485-92, 2015.
Article in English | MEDLINE | ID: mdl-25892189

ABSTRACT

We aimed to examine the association between impaired proinsulin processing in pancreatic beta cells and type 2 diabetes mellitus in non-obese Japanese patients. Participants were divided into groups for normal glucose tolerance, prediabetes, and type 2 diabetes based on the oral glucose tolerance test (OGTT). Activities of prohormone convertase (PC) 1/3 and PC2 in fasting states were estimated. Multiple regression analysis was undertaken to ascertain if alteration of the activities of these enzymes contributes to the development of impaired glucose tolerance by comparison with HOMA-ß and the oral disposition index (DI(O)). Overall, 452 subjects were included. PC1/3 activity tended to decrease in type 2 diabetes compared with normal glucose tolerance. PC2 activity showed no difference among the three groups. Decreased estimated PC1/3 activity was significantly associated with type 2 diabetes after adjustment for sex, age, creatinine, triglycerides, HOMA-ß and DI(O). Odds ratios (95% CI) of PC1/3, HOMA-ß, and DI(O) were 2.16 (1.12-4.19), 3.44 (1.82-6.52) and 14.60 (7.87-27.11), respectively. Furthermore, decreased PC1/3(≤1.7) combined with decreased HOMA-ß (≤30) had a sensitivity of 73% and specificity of 62%. Decreased PC1/3 activity may be a useful measurement of beta-cell function alongside decreased HOMA-ß or DI(O). A combined decrease in estimated fasting PC1/3 activity and HOMA-ß measurement led to suspicion of type 2 diabetes in the non-obese Japanese population studied.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Proinsulin/metabolism , Proprotein Convertases/metabolism , Protein Processing, Post-Translational , Adult , Aged , Algorithms , Biomarkers/blood , Cohort Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/ethnology , Female , Humans , Insulin/blood , Insulin Resistance/ethnology , Isoenzymes/metabolism , Japan , Male , Middle Aged , Proinsulin/blood , Proteolysis , Sensitivity and Specificity
5.
J Cell Physiol ; 230(1): 199-209, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24958127

ABSTRACT

Ghrelin is a physiological-active peptide with growth hormone-releasing activity, orexigenic activity, etc. In addition, the recent study has also suggested that ghrelin possesses the pathophysiological abilities related with type 2 diabetes. However, the ghrelin-direct-effects implicated in type 2 diabetes on peripheral tissues have been still unclear, whereas its actions on the central nervous system (CNS) appear to induce the development of diabetes. Thus, to assess its peripheral effects correlated with diabetes, we investigated the regulatory mechanisms about adipokines, which play a central role in inducing peripheral insulin resistance, secreted from mature 3T3-L1 adipocytes stimulated with ghrelin in vitro . The stimulation with 50 nmol/L ghrelin for 24 h resulted in the significant 1.9-fold increase on vascular endothelial growth factor-120 (VEGF(120)) releases (p < 0.01) and the 1.7-fold on monocyte chemoattractant protein-1 (MCP-1) (p < 0.01) from 3T3-L1 adipocytes, respectively, while ghrelin failed to enhance tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), IL-6, IL-10 and adiponectin secretions. In addition, Akt phosphorylation on Ser473 and c-Jun NH2 -terminal protein kinase (JNK) phosphorylation on Thr183/Tyr185 were markedly enhanced 1.4-fold (p < 0.01) and 1.6-fold (p < 0.01) in the ghrelin-stimulated adipocytes, respectively. Furthermore, the treatment with LY294002 (50 µmol/L) and Wortmannin (10nmol/L), inhibitors of phosphatidylinositol 3-kinase (PI3K), significantly decreased the amplified VEGF(120) secretion by 29% (p < 0.01) and 28% (p < 0.01) relative to the cells stimulated by ghrelin alone, respectively, whereas these inhibitors had no effects on increased MCP-1 release. On the other hand, JNK inhibitor SP600125 (10 µmol/L) clearly reduced the increased MCP-1, but not VEGF(120), release by 35% relative to the only ghrelin-stimulated cells (p < 0.01). In conclusion, ghrelin can enhance the secretions of proinflammatory adipokines, VEGF(120) and MCP-1, but fails to affect IL-10 and adiponectin which are considered to be anti-inflammatory adipokines. Moreover, this augmented VEGF(120) release is invited through the activation of PI3K pathways and the MCP-1 is through JNK pathways. Consequently, our results strongly suggest that ghrelin can induce the development of diabetes via its direct-action in peripheral tissues as well as via in CNS.


Subject(s)
Adipocytes/drug effects , Chemokine CCL2/metabolism , Diabetes Mellitus, Type 2/metabolism , Ghrelin/pharmacology , Vascular Endothelial Growth Factor A/metabolism , 3T3 Cells , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/biosynthesis , Adipocytes/metabolism , Adiponectin/biosynthesis , Androstadienes/pharmacology , Animals , Anthracenes/pharmacology , Cell Line , Chromones/pharmacology , Endoplasmic Reticulum Stress/drug effects , Interleukin-10/biosynthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Morpholines/pharmacology , Oxidative Stress/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Wortmannin
6.
Biochem Biophys Res Commun ; 451(2): 339-44, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25109805

ABSTRACT

Recent studies have suggested glucagon-like peptide-1 (GLP-1) signaling to exert anti-inflammatory effects on endothelial cells, although the precise underlying mechanism remains to be elucidated. In the present study, we investigated whether PPARγ activation is involved in the GLP-1-mediated anti-inflammatory action on endothelial cells. When we treated HUVEC cells with 0.2ng/ml exendin-4, a GLP-1 receptor agonist, endogenous PPARγ transcriptional activity was significantly elevated, by approximately 20%, as compared with control cells. The maximum PPARγ activity enhancing effect of exendin-4 was observed 12h after the initiation of incubation with exendin-4. As H89, a PKA inhibitor, abolished GLP-1-induced PPARγ enhancement, the signaling downstream from GLP-1 cross-talk must have been involved in PPARγ activation. In conclusion, our results suggest that GLP-1 has the potential to induce PPARγ activity, partially explaining the anti-inflammatory effects of GLP-1 on endothelial cells. Cross-talk between GLP-1 signaling and PPARγ activation would have major impacts on treatments for patients at high risk for cardiovascular disease.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , PPAR gamma/metabolism , Peptides/pharmacology , Receptors, Glucagon/agonists , Venoms/pharmacology , Anilides/pharmacology , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Exenatide , Gene Expression/drug effects , Glucagon-Like Peptide-1 Receptor , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Isoquinolines/pharmacology , NADPH Oxidase 1 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/metabolism , PPAR gamma/antagonists & inhibitors , Phosphorylation , Pioglitazone , Protein Kinase Inhibitors/pharmacology , Receptor Cross-Talk , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thiazolidinediones/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
7.
Endocr J ; 61(6): 607-14, 2014.
Article in English | MEDLINE | ID: mdl-24705588

ABSTRACT

Type 2 diabetic (T2D) patients exhibit fasting relative hyperproinsulinemia owing to pancreatic ß-cell dysfunction. To clarify the mechanism underlying this hyperproinsulinemic state, we evaluated the activities of the endopeptidases prohormone convertase (PC) 1/3 and PC2 in T2D patients. Fasting blood levels of intact proinsulin (IPI), total proinsulin (t-PI) and C-peptide were measured simultaneously, and intravenous glucagon loading was performed to investigate the dynamics of circulating proinsulin-related molecules released from pancreatic ß-cells in 12 healthy volunteers and 18 T2D patients. Taking advantage of the 95% cross-reactivity between proinsulin and des-31,32-proinsulin (des-31,32-PI) with the human proinsulin radioimmunoassay kit used in this study, we estimated PC1/3 and PC2 activities using the following formulas: des-31,32-PI = (t-PI-IPI)/0.95; PC1/3 activity = des-31,32-PI/IPI; and PC2 activity = C-peptide/des-31,32-PI. C-peptide responses to glucagon were slightly lower among T2D patients. IPI and the IPI/C-peptide ratio were significantly higher in T2D patients (p<0.05 and p<0.01, respectively). There was no difference in des-31,32-PI levels or PC2 activity between the two groups. However, PC1/3 activity was significantly lower in T2D patients than in the control group (p<0.01). We propose that decreased activity of PC1/3 rather than PC2 in pancreatic ß-cells is involved in the impaired proinsulin processing, resulting in elevated IPI levels in T2D patients.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Proinsulin/metabolism , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/metabolism , Protein Processing, Post-Translational , Administration, Intravenous , Adult , Aged , C-Peptide/metabolism , Glucagon/administration & dosage , Humans , Middle Aged , Protein Processing, Post-Translational/drug effects , Statistics as Topic
8.
Biochem Biophys Res Commun ; 445(2): 275-81, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24491567

ABSTRACT

It have been reported that abnormal bone metabolism often occurs in patients with type 2 diabetes, but the underlying mechanisms remain to be elucidated. In recent years dyslipidemia (hyperlipidemia) has been presumed to have an influence on bone metabolism. In addition, the involvements of VEGF and MCP-1 derived from osteoblasts in bone abnormal metabolism were also observed. Thus, we investigated the pathogenic mechanism of this abnormal bone metabolism, which is included in the regulation of VEGF and MCP-1 secretions from osteoblasts, by using UMR-106 osteosarcoma cells as an osteoblast cell model and treating them with palmitate in order to mimic a state of hyperlipidemia. Palmitate-preloaded cells showed the significant increase of VEGF120 release (1.8-fold vs. control cells, p<0.01). Moreover, the treatment with palmitate significantly increased VEGF-A mRNA with the maximal 2.5-fold upregulation at 12h after the treatment (p<0.01). However, MCP-1 release was not affected by palmitate. Moreover, the amplified VEGF120 secretion with palmitate was significantly decreased by the treatment with TLR4 antagonist or PI3K pathway inhibitors, LY294002 and wortmannin (p<0.01, respectively). On the other hand, the stimulation with TNF-α, which osteoclasts were able to release, significantly enhanced MCP-1 secretion (p<0.01), but had no effect on VEGF120. On the contrary IL-1ß amplified VEGF120 release (p<0.01), but not MCP-1. These results suggest that palmitate can increase VEGF120 release from UMR-106 osteosarcoma cells, which is accelerated at the transcriptional level, and this increase of VEGF120 release may be mediated though, at least partly, TLR4 and the PI3K pathways. In addition, we also verified that TNF-α and IL-1ß, which are considered to be derived from osteoclasts, amplified the secretions of MCP-1 and VEGF120 from UMR-106 cells, respectively.


Subject(s)
Hyperlipidemias/metabolism , Osteoblasts/metabolism , Palmitates/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Chemokine CCL2/metabolism , Hyperlipidemias/genetics , Osteoblasts/cytology , RNA, Messenger/genetics , Rats , Signal Transduction , Toll-Like Receptor 4/metabolism , Vascular Endothelial Growth Factor A/genetics
9.
Obesity (Silver Spring) ; 21(8): 1625-34, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23670941

ABSTRACT

OBJECTIVE: Expressions of vascular endothelial growth factor (VEGF) are increased in obese adipocytes and is secreted from obese adipose tissue through hypoxia-independent pathways. Therefore, we investigated the hypoxia-independent mechanism underlying increased expression and release of VEGF in obese adipocytes. DESIGN AND METHODS: We compared signal transduction pathways regulating VEGF with those regulating monocyte chemoattractant protein-1 (MCP-1), which is increased in obese adipocytes, in an in vitro model of artificially hypertrophied 3T3-L1 adipocytes preloaded with palmitate, without the influence of hypoxia. RESULTS: Palmitate-preloaded cells exhibited significantly enhanced oxidative stress (P < 0.01) and showed increased VEGF120 and MCP-1 release (P < 0.01, respectively), while endoplasmic reticulum (ER) stress was not induced. Increased VEGF120 release was significantly decreased with PI3K inhibitor LY294002 (P < 0.01). In addition, antioxidant N-acetyl-cysteine (NAC) markedly diminished not only VEGF120 secretion (P < 0.01) but also augmented Akt phosphorylation on Ser473 (P < 0.01). In contrast, increased MCP-1 release was suppressed with JNK inhibitor SP600125 and p38 MAPK inhibitor SB203580 (P < 0.01). CONCLUSIONS: VEGF120 release from hypertrophied adipocytes can be enhanced through PI3K pathways activated by oxidative stress but not by ER stress, suggesting that VEGF120 secretion is regulated through oxidative stress-dependent pathways distinct from those involved in MCP-1 release through either JNK or p38 MAPK activation.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , 3T3-L1 Cells , Acetylcysteine/pharmacology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Hypoxia , Chemokine CCL2/metabolism , Chromones/pharmacology , Imidazoles/pharmacology , Mice , Morpholines/pharmacology , Palmitates/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Biochem Biophys Res Commun ; 419(2): 200-5, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22330806

ABSTRACT

Although white adipocytes contain a larger number of mitochondria per cytoplasmic volume, adipocyte mitochondrial uncoupling to reduce the efficiency of ATP production on cellular function including secretory regulation of bioactive molecules such as VEGF and MCP-1 remains to be elucidated. Here we induce mitochondrial uncoupling under hypoxia-independent conditions in mature 3T3-L1 adipocytes using a metabolic uncoupler, dinitrophenol (DNP). MCP-1 release was significantly decreased by 26% (p<0.01) in 24h DNP (30 µmol/L)-treated adipocytes compared to control cells. In contrast, secreted VEGF(120) lacking a heparin-binding domain was markedly increased 2.0-fold (p<0.01). CHOP content in these cells also were augmented (p<0.01), but no significant increase of endogenous oxidative stress was observed. Treatment with thapsigargin, which can induce exogenous endoplasmic reticulum (ER) stress, clearly attenuated MCP-1 release (p<0.01), but exhibited no effects on VEGF(120) secretion. On the other hand, exogenous H(2)O(2) amplified both MCP-1 and VEGF(120) secretion (p<0.05). In addition, under chronic activation of AMPK by AICAR, MCP-1 release was significantly diminished (p<0.05) but VEGF(120) secretion was increased (p<0.01). JNK phosphorylation in mature adipocytes was decreased by treatment with either DNP or AICAR (p<0.01). Enhanced VEGF(120) secretion with either DNP or AICAR was markedly suppressed by PI3K inhibitor LY294002 (p<0.01). Thus, induced mitochondrial uncoupling in adipocytes can reduce MCP-1 release through induction of endogenous ER stress and by reduced JNK activities via chronic activation of AMPK. Under this condition, VEGF(120) secretion was increased through PI3K-dependent pathways, which were chronically activated by AMPK, and not through ER stress. Because the decrease of MCP-1 secretion under mitochondrial uncoupling might attenuate chronic low-grade inflammation by suppressing macrophages recruitment to adipose tissue, clarification of the mechanism might reveal novel therapeutic targets for ameliorating obesity-associated insulin resistance in metabolic syndrome and type 2 diabetes.


Subject(s)
Adipocytes/physiology , Adipogenesis , Chemokine CCL2/antagonists & inhibitors , Endoplasmic Reticulum Stress , Mitochondria/physiology , Protein Kinases/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , 3T3-L1 Cells , AMP-Activated Protein Kinase Kinases , Adipocytes/metabolism , Animals , Chemokine CCL2/metabolism , Diabetes Mellitus, Type 2/metabolism , Metabolic Networks and Pathways , Metabolic Syndrome/metabolism , Mice , Mitochondria/metabolism , Obesity/metabolism
11.
Diabetes Res Clin Pract ; 82(1): 119-26, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18774197

ABSTRACT

In order to investigate the underlying mechanism of alterations in bone mineral metabolism in patients with type 2 diabetes, we determined circulating levels of bone functional markers along with urinary excretion of sorbitol (SOR) and bone mineral density (BMD), and also examined their mutual interrelationship. A total of 151 male type 2 diabetic patients were examined in this study. Forty-eight age-matched male healthy subjects were also studied as the controls. A significant reduction of serum intact osteocalcin (i-OC) was found in the diabetic groups (p<0.01). On the other hand, circulating levels of tartrate resistant acid phosphatase (TRAP) in the diabetic patients were significantly higher than those in the controls (p<0.01). Interestingly, a significantly negative relationship was observed between BMD and serum TRAP (p<0.01), although no significant relationship was noted between BMD and serum i-OC in diabetic patients. Urinary excretion of SOR was significantly elevated in the diabetic patients when compared with the controls (p<0.01). In addition, a significantly positive correlation was observed between serum TRAP and urinary SOR (p<0.01), but not between serum i-OC and urinary SOR. Elevated serum TRAP in diabetes was reduced after the administration of aldose reductase inhibitor (p<0.05). It seems most likely that the increase in osteoclastic function probably due to accelerated polyol pathway plays a crucial role in the pathogenesis of decreased bone mineral content in male patients with type 2 diabetes.


Subject(s)
Bone Diseases, Metabolic/blood , Bone Resorption/blood , Diabetes Mellitus, Type 2/blood , Osteocalcin/blood , Acid Phosphatase/blood , Bone Density , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Humans , Isoenzymes/blood , L-Iditol 2-Dehydrogenase/metabolism , Male , Middle Aged , Polymers/metabolism , Signal Transduction , Sorbitol/blood , Sorbitol/metabolism , Sorbitol/urine , Tartrate-Resistant Acid Phosphatase
12.
Am J Physiol Endocrinol Metab ; 294(5): E898-909, 2008 May.
Article in English | MEDLINE | ID: mdl-18303122

ABSTRACT

Obese conditions increase the expression of adipocytokine monocyte chemoattractant protein-1 (MCP-1) in adipose tissue as well as MCP-1 plasma levels. To investigate the mechanism behind increased MCP-1, we used a model in which 3T3-L1 adipocytes were artificially hypertrophied by preloading with palmitate in vitro. As observed in obesity, under our model conditions, palmitate-preloaded cells showed significantly increased oxidative stress and increased MCP-1 expression relative to control cells. This increased MCP-1 expression was enhanced by adding exogenous tumor necrosis factor-alpha (TNF-alpha; 17.8-fold vs. control cells, P < 0.01) rather than interleukin-1beta (IL-1beta; 2.6-fold vs. control cells, P < 0.01). However, endogenous TNF-alpha and IL-1beta release was not affected in hypertrophied cells, suggesting that these endogenous cytokines do not mediate hypertrophy-induced increase in MCP-1. MCP-1 secretion from hypertrophied cells was significantly decreased by treatment with antioxidant N-acetyl-cysteine, JNK inhibitors SP600125 and JIP-1 peptide, and IkappaB phosphorylation inhibitors BAY 11-7085 and BMS-345541 (P < 0.01). MCP-1 secretion was not affected by peroxisome proliferator-activated receptor-gamma (PPARgamma) antagonists assayed. Adiponectin, another adipocytokine studied in parallel, also showed increased release in hypertrophy relative to control cells. But in contrast to MCP-1, adiponectin release was significantly suppressed by both exogenous TNF-alpha and IL-1beta as well as by PPARgamma antagonists bisphenol A diglycidyl ether and T0070907 (P < 0.01). JNK inhibitors and IkappaB phosphorylation inhibitors showed no significant effect on adiponectin. We conclude that adipocyte hypertrophy through palmitate loading causes oxidative stress, which in turn increases MCP-1 expression and secretion through JNK and IkappaB signaling. In contrast, the parallel increase in adiponectin expression appears to be related to the PPARgamma ligand properties of palmitate.


Subject(s)
Adipocytes/metabolism , Adiponectin/metabolism , Chemokine CCL2/metabolism , I-kappa B Kinase/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Palmitates/pharmacology , Signal Transduction/physiology , 3T3 Cells , Adipocytes/drug effects , Adipocytes/ultrastructure , Animals , Blotting, Western , Cell Size/drug effects , Hydrogen Peroxide/metabolism , Interleukin-1beta/pharmacology , Mice , PPAR gamma/metabolism , PPAR gamma/physiology , Triglycerides/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
13.
Metabolism ; 55(6): 722-30, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16713429

ABSTRACT

Increased oxidative stress under hyperglycemia may contribute to progressive deterioration of peripheral insulin sensitivity. In this study, we investigated whether gliclazide, a second-generation sulfonylurea, can protect 3T3L1 adipocytes from insulin resistance induced by oxidative stress, and whether gliclazide can restore insulin-stimulated glucose transporter 4 (GLUT4) translocation under oxidative stress. We incubated 3T3L1 adipocytes in hydrogen peroxide to produce oxidative stress, then administered various concentrations of gliclazide, N-acetylcystein (NAC), or glibenclamide. Cells treated with these drugs were next exposed to insulin, subsequent glucose uptake was measured, and the insulin-stimulated GLUT4 translocation was monitored in living cells. We found that hydrogen peroxide treatment alone suppressed glucose uptake by insulin stimulation to 65.9%+/-7.8% of the corresponding controls (P<.01). However, addition of 0.1 to 10 micromol/L gliclazide to hydrogen peroxide-treated cells dose-dependently restored glucose uptake, with 5 micromol/L gliclazide significantly restoring glucose uptake to 93.3+/-6.6% (P<.01) even under hydrogen peroxide. Treatment with the known anti-oxidant NAC also dose-dependently (0.1-10 mmol/L) restored insulin-induced glucose uptake in the presence of hydrogen peroxide. However, glibenclamide (0.1-10 micromol/L), another second-generation sulfonylurea, failed to improve glucose uptake. Similarly, treatment with 5 micromol/L gliclazide or 10 mmol/L NAC significantly overcome the reduction in insulin-stimulated GLUT4 translocation by hydrogen peroxide (P<.01), whereas 5 micromol/L glibenclamide did not. Therefore our data regarding gliclazide further characterize its mechanism of hypoglycemic effect: the observed improvements in insulin sensitivity and in GLUT4 translocation indicate that gliclazide counters the hydrogen peroxide-induced insulin resistance in 3T3L1 adipocytes and also would further augment the hypoglycemic effect of this drug as insulinotropic sulfonylurea.


Subject(s)
Adipocytes/drug effects , Gliclazide/pharmacology , Glucose Transporter Type 4/metabolism , Insulin Resistance , 3T3-L1 Cells , Animals , Glucose/metabolism , Glucose Transporter Type 4/genetics , Hydrogen Peroxide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Mice , Oxidative Stress , Protein Transport
15.
Biochem Biophys Res Commun ; 324(2): 810-4, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15474499

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) regulates several cellular functions, but its physiological role in pancreatic islet cells remains to be investigated. In this study, we confirmed the presence of PPAR-gamma in rat isolated islets and examined its role on insulin and glucagon secretion by using PPAR-gamma-overexpressed islets. PPAR-gamma overexpression significantly suppressed insulin secretion induced by stimulatory concentration of glucose (p<0.05). In addition, insulin secretion evoked by high potassium depolarization also was significantly decreased from PPAR-gamma-overexpressed islets (p<0.05). On the other hand, no significant change in glucagon release was observed after high potassium depolarization between PPAR-gamma-overexpressed and control islets. Insulin and glucagon content in islets was not statistically different between the two groups. In addition, the expression of uncoupling protein-2 (UCP-2) was found to be induced in PPAR-gamma-overexpressed islets. This result clearly indicates that the deteriorative effect of PPAR-gamma overexpression on the secretory machinery is selective for pancreatic beta-cells. And it is possible that its site of action can be located in the energy-consuming exocytotic process of insulin secretory granules, and that the reduction of ATP production through increased UCP-2 reduces insulin exocytosis.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , PPAR gamma/metabolism , Adenosine Triphosphate/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Cells, Cultured , DNA, Complementary/metabolism , Exocytosis , Glucagon/metabolism , Glucose/metabolism , Immunoblotting , Insulin Secretion , Ion Channels , Male , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Uncoupling Protein 2
16.
Metabolism ; 53(4): 488-94, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15045697

ABSTRACT

In order to assess the beneficial effect of the peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonist pioglitazone on reduction of mass and alteration of function of pancreatic beta cells under diabetic conditions, diabetic C57BL/KsJ db/db mice were treated with pioglitazone for 6 weeks, and insulin secretory capacity and insulin content of isolated pancreatic islets were evaluated. In addition, the expression of oxidative stress markers, 4-hydroxy-2-nonenal (HNE)-modified proteins and heme oxygenase-1, in endocrine pancreas was examined to measure reduction of oxidative stress in pancreatic beta cells. The capacity for glucose-induced insulin secretion from isolated islets and their insulin content were improved by pioglitazone treatment (P <.01). When beta cells were stained with anti-insulin antibodies, those of db/db mice treated with pioglitazone exhibited strong staining, as also observed in their lean littermates. The density of immunostaining for oxidative stress markers was significantly reduced in pancreatic islets of pioglitazone-treated db/db mice (P <.05). This study clearly demonstrates the benefit of long-term treatment with pioglitazone in decreasing hyperglycemia and improving glucose-induced insulin secretory capacity in diabetic db/db mice. The results of immunocytochemical examination suggest that this treatment reduces oxidative stress and thereby preserves beta-cell mass. Treatment with pioglitazone thus protects against beta-cell damage and would be useful for restoration of insulin secretory capacity in obese diabetes individuals.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Oxidative Stress/physiology , Thiazolidinediones/pharmacology , Aldehydes/metabolism , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Glucose/metabolism , Glucose/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Hemostatics , Immunohistochemistry , Insulin/blood , Insulin Resistance , Insulin Secretion , Islets of Langerhans/pathology , Islets of Langerhans/ultrastructure , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Obesity/blood , Obesity/metabolism , Obesity/pathology , Oxidative Stress/drug effects , Pioglitazone , Triglycerides/blood
17.
Biochem Biophys Res Commun ; 311(3): 660-4, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-14623322

ABSTRACT

To investigate the cellular mechanism of insulinotropic effect of glutamate in pancreatic beta cells, we utilized patch-clamp technique to monitor directly the activities of ATP-sensitive potassium channels (K(ATP) channels). Dimethylglutamate (5mM), a membrane-permeable analog of glutamate, augmented the insulin release induced by the stimulatory concentrations of glucose (p<0.05-0.01). In the cell-attached configurations, dimethylglutamate reversibly and significantly suppressed the K(ATP) channel activities (p<0.01). On the other hand, no significant effect was observed when glutamate itself was applied to the inside-out patches, whereas the prompt and reversible suppression was recorded in the case of ATP (p<0.01). These results indicate that the insulinotropic action of glutamate in beta cells could be derived from the inhibition of K(ATP) channel activities, probably due to generation of messengers via intracellular metabolism such as ATP.


Subject(s)
Adenosine Triphosphate/metabolism , Glutamic Acid/chemistry , Insulin/metabolism , Islets of Langerhans/metabolism , Animals , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Glucose/chemistry , Glutamic Acid/pharmacology , Insulin Secretion , Male , Mice , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels/chemistry , Rats , Rats, Wistar
18.
Biochem Biophys Res Commun ; 303(1): 112-9, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12646174

ABSTRACT

Oxidative stress is induced under diabetic conditions and possibly causes various forms of tissue damage in patients with diabetes. Recently, it has become aware that susceptibility of pancreatic beta-cells to oxidative stress contributes to the progressive deterioration of beta-cell function in type 2 diabetes. A hypoglycemic sulfonylurea, gliclazide, is known to be a general free radical scavenger and its beneficial effects on diabetic complications have been documented. In the present study, we investigated whether gliclazide could protect pancreatic beta-cells from oxidative damage. One hundred and fifty microM hydrogen peroxide reduced viability of mouse MIN6 beta-cells to 29.3%. Addition of 2 microM gliclazide protected MIN6 cells from the cell death induced by H(2)O(2) to 55.9%. Glibenclamide, another widely used sulfonylurea, had no significant effects even at 10 microM. Nuclear chromatin staining analysis revealed that the preserved viability by gliclazide was due to inhibition of apoptosis. Hydrogen peroxide-induced expression of an anti-oxidative gene heme oxygenase-1 and stress genes A20 and p21(CIP1/WAF1), whose induction was suppressed by gliclazide. These results suggest that gliclazide reduces oxidative stress of beta-cells by H(2)O(2) probably due to its radical scavenging activity. Gliclazide may be effective in preventing beta-cells from the toxic action of reactive oxygen species in diabetes.


Subject(s)
Gliclazide/pharmacology , Hydrogen Peroxide/pharmacology , Hypoglycemic Agents/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Animals , Antioxidants/pharmacology , Apoptosis , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Hydrogen Peroxide/metabolism , L-Lactate Dehydrogenase/metabolism , Mice , Microscopy, Fluorescence , Oxidative Stress , Polymerase Chain Reaction , Protein Binding , RNA/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species
19.
Biochem J ; 363(Pt 1): 73-80, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11903049

ABSTRACT

The dynamics of exocytosis/endocytosis of insulin secretory granules in pancreatic beta-cells remains to be clarified. In the present study, we visualized and analysed the motion of insulin secretory granules in MIN6 cells using pH-sensitive green fluorescent protein (pHluorin) fused to either insulin or the vesicle membrane protein, phogrin. In order to monitor insulin exocytosis, pHluorin, which is brightly fluorescent at approximately pH 7.4, but not at approximately pH 5.0, was attached to the C-terminus of insulin. To monitor the motion of insulin secretory granules throughout exocytosis/endocytosis, pHluorin was inserted between the third and fourth amino acids after the identified signal-peptide cleavage site of rat phogrin cDNA. Using this method of cDNA construction, pHluorin was located in the vesicle lumen, which may enable discrimination of the unfused acidic secretory granules from the fused neutralized ones. In MIN6 cells expressing insulin-pHluorin, time-lapse confocal laser scanning microscopy (5 or 10 s intervals) revealed the appearance of fluorescent spots by depolarization after stimulation with 50 mM KCl and 22 mM glucose. The number of these spots in the image at the indicated times was counted and found to be consistent with the results of insulin release measured by RIA during the time course. In MIN6 cells expressing phogrin-pHluorin, data showed that fluorescent spots appeared following high KCl stimulation and remained stationary for a while, moved on the plasma membrane and then disappeared. Thus we demonstrate the visualized motion of insulin granule exocytosis/endocytosis using the pH-sensitive marker, pHluorin.


Subject(s)
Endocytosis , Exocytosis , Islets of Langerhans/metabolism , Luminescent Proteins/metabolism , Animals , Cell Line , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Glucose/metabolism , Green Fluorescent Proteins , Humans , Hydrogen-Ion Concentration , Insulin/metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Models, Genetic , Plasmids/metabolism , Rats , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...