Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Biomedicines ; 8(11)2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33217950

ABSTRACT

Despite numerous studies, the effects of ezetimibe on glucose metabolism are poorly understood. Here, we aimed to investigate the effects of ezetimibe on glucose metabolism and the expression of inflammatory markers. Thirteen rats were randomly assigned to an ezetimibe (n = 6) or control group (n = 7). The control group received a high fat diet (HFD; 60 Kcal%), whereas the ezetimibe group received an HFD (60 Kcal%) containing 160 mg/kg of ezetimibe. After 14 weeks, adipose and liver tissues, along with plasma, were collected and comparatively analyzed. The effects of combination therapy with ezetimibe and statins on glucose metabolism were investigated over a 1-year period using data from patients with hyperlipidemia. Several indices of glucose metabolism partially improved in the ezetimibe group. The sizes of adipocytes and the accumulation of pro-inflammatory cytokines were reduced in the ezetimibe group. Ezetimibe treatment induced anti-inflammatory cytokines and fatty acid oxidation in adipocytes and reduced serum levels of free fatty acids. Clinical data analysis revealed that statin monotherapy significantly increased insulin resistance. However, combination therapy with ezetimibe and statins did not increase insulin resistance. In conclusion, ezetimibe was found to reduce the sizes of adipocytes in visceral fat and serum levels of free fatty acids, to induce fatty acid oxidation, to improve adipocytic inflammation, and to partially improve glycemic index values.

2.
J Atheroscler Thromb ; 27(5): 429-440, 2020 May 01.
Article in English | MEDLINE | ID: mdl-31527323

ABSTRACT

AIM: Most statins increase the risk of new-onset diabetes. Unlike other statins, pitavastatin is reported to exert neutral effects on serum glucose level, but the precise mechanism is unknown. METHODS: Eight-week-old male C57BL/6J mice (n=26) were fed high-fat diet (HFD, 45% fat) with 0.01% placebo, rosuvastatin, or pitavastatin for 12 weeks. Cultured HepG2, C2C12, and 3T3-L1 cells and visceral adipocytes from HFD-fed mice were treated with vehicle or 10 µM statins for 24 h. The effects of pitavastatin and rosuvastatin on intracellular insulin signaling and glucose transporter 4 (GLUT4) translocation were evaluated. RESULTS: After 12 weeks, the fasting blood glucose level was significantly lower in pitavastatin-treated group than in rosuvastatin-treated group (115.2±7.0 versus 137.4±22.3 mg/dL, p=0.024). Insulin tolerance significantly improved in pitavastatin-treated group as compared with rosuvastatin-treated group, and no significant difference was observed in glucose tolerance. Although plasma adiponectin and insulin levels were not different between the two statin treatment groups, the insulin-induced protein kinase B phosphorylation was weakly attenuated in pitavastatin-treated adipocytes than in rosuvastatin-treated adipocytes. Furthermore, minor attenuation in insulin-induced GLUT4 translocation to the plasma membrane of adipocytes was observed in pitavastatin-treated group. CONCLUSION: Pitavastatin showed lower diabetogenic effects than rosuvastatin in mice that may be mediated by minor attenuations in insulin signaling in adipocytes.


Subject(s)
Adipocytes , Blood Glucose , Diabetes Mellitus , Quinolines/pharmacology , Rosuvastatin Calcium/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/blood , Adiponectin/metabolism , Animals , Anticholesteremic Agents/pharmacology , Blood Glucose/analysis , Blood Glucose/metabolism , Cells, Cultured , Correlation of Data , Diabetes Mellitus/blood , Diabetes Mellitus/chemically induced , Diabetes Mellitus/diagnosis , Glucose Transporter Type 4/metabolism , Humans , Insulin/blood , Insulin/metabolism , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
3.
Diabetes Obes Metab ; 22(3): 373-382, 2020 03.
Article in English | MEDLINE | ID: mdl-31692240

ABSTRACT

AIMS: To investigate the effect of dapagliflozin, a sodium-glucose co-transporter-2 (SGLT2) inhibitor, on renal gluconeogenesis in vitro, ex vivo and in vivo. MATERIALS AND METHODS: We treated HK-2 cells (human renal proximal tubule cells) and mouse primary renal proximal tubule cells with dapagliflozin, and evaluated the process of renal gluconeogenesis. We also examined the effect of dapagliflozin on renal gluconeogenesis in normoglycaemic and hyperglycaemic mice. RESULTS: Dapagliflozin enhanced renal gluconeogenesis in vitro, ex vivo and in vivo. It increased phosphoenolpyruvate carboxykinase (PEPCK), glucose-6-phosphatase (G6Pase), peroxisome proliferative activated receptor-gamma co-activator 1α (PGC-1α) and phosphorylated cyclic-AMP response element binding protein (CREB) expression and decreased phosphorylated Forkhead Box O1 (FOXO1) expression in HK-2 cells, mouse primary renal proximal tubule cells, and the mouse renal cortex. Glutamine enhanced the gluconeogenic effect of dapagliflozin in HK-2 cells. Also, dapagliflozin increased 14 C-glutamine utilization in HK-2 cells. Glucagon did not affect dapagliflozin-induced enhancement in renal gluconeogenesis in HK-2 cells. SGLT2 gene knockdown with siRNA resulted in an increase of gluconeogenic gene expression and associated transcription factors in HK-2 cells. Dapagliflozin reduced fasting plasma glucose levels and improved oral glucose tolerance and insulin tolerance in high-fat diet-fed hyperglycaemic mice, although renal gluconeogenesis was enhanced. CONCLUSIONS: Dapagliflozin increased levels of gluconeogenic enzyme in the renal cortex and consequently increased renal gluconeogenesis, which is mediated by SGLT2 inhibition.


Subject(s)
Gluconeogenesis , Sodium-Glucose Transporter 2 Inhibitors , Benzhydryl Compounds , Glucose/metabolism , Glucosides , Kidney Tubules, Proximal/metabolism , Liver/metabolism , Sodium/metabolism , Sodium-Glucose Transporter 2/metabolism
4.
Sci Rep ; 9(1): 5318, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926836

ABSTRACT

Recent studies have reported that statins are associated with increased incidence of diabetes. Although several mechanisms have been proposed, the role of the kidney's glucose metabolism upon statin treatment is still unclear. Thus, we investigated the role of pravastatin in gluconeogenesis and glycolysis. HK-2 and HepG2 cells were treated with pravastatin and cultured under either high- or normal-cholesterol conditions. In HK-2 cells treated with pravastatin under both high- and normal-cholesterol conditions, the protein expression of only pyruvate kinase isozymes L/R (PKLR) decreased in a dose-dependent manner, while the protein expression of other glucose metabolism related enzymes remained unchanged. Within the in vivo experiment, male C57BL/6 mice were fed either pravastatin-treated normal-fat diets for 2 or 4 weeks or pravastatin-treated high-fat diets for 16 weeks. Protein expression of PKLR in the kidneys from mice that consumed pravastatin-treated high-fat diets decreased significantly compared to the controls. Upon the treatments of pravastatin, only the PKLR expression decreased in lean mice. Furthermore, PKLR activity decreased significantly in the kidney after pravastatin treatments. However, there was no change in enzyme activity in the liver, suggesting that pravastatin decreased PKLR activity only in the kidney. This change may be associated with the hyperglycemic effect of statins.


Subject(s)
Carbohydrate Metabolism/drug effects , Gene Expression Regulation, Enzymologic , Glucose/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Pravastatin/pharmacology , Pyruvate Kinase/genetics , Blood Glucose , Cell Line , Diet, High-Fat , Humans , Pyruvate Kinase/metabolism
5.
Autophagy ; 13(10): 1767-1781, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28933629

ABSTRACT

Impairment in macroautophagy/autophagy flux and inflammasome activation are common characteristics of nonalcoholic steatohepatitis (NASH). Considering the lack of approved agents for treating NASH, drugs that can enhance autophagy and modulate inflammasome pathways may be beneficial. Here, we investigated the novel mechanism of ezetimibe, a widely prescribed drug for hypercholesterolemia, as a therapeutic option for ameliorating NASH. Human liver samples with steatosis and NASH were analyzed. For in vitro studies of autophagy and inflammasomes, primary mouse hepatocytes, human hepatoma cells, mouse embryonic fibroblasts with Ampk or Tsc2 knockout, and human or primary mouse macrophages were treated with ezetimibe and palmitate. Steatohepatitis and fibrosis were induced by feeding Atg7 wild-type, haploinsufficient, and knockout mice a methionine- and choline-deficient diet with ezetimibe (10 mg/kg) for 4 wk. Human livers with steatosis or NASH presented impaired autophagy with decreased nuclear TFEB and increased SQSTM1, MAP1LC3-II, and NLRP3 expression. Ezetimibe increased autophagy flux and concomitantly ameliorated lipid accumulation and apoptosis in palmitate-exposed hepatocytes. Ezetimibe induced AMPK phosphorylation and subsequent TFEB nuclear translocation, related to MAPK/ERK. In macrophages, ezetimibe blocked the NLRP3 inflammasome-IL1B pathway in an autophagy-dependent manner and modulated hepatocyte-macrophage interaction via extracellular vesicles. Ezetimibe attenuated lipid accumulation, inflammation, and fibrosis in liver-specific Atg7 wild-type and haploinsufficient mice, but not in knockout mice. Ezetimibe ameliorates steatohepatitis by autophagy induction through AMPK activation and TFEB nuclear translocation, related to an independent MTOR ameliorative effect and the MAPK/ERK pathway. Ezetimibe dampens NLRP3 inflammasome activation in macrophages by modulating autophagy and a hepatocyte-driven exosome pathway.


Subject(s)
Autophagy/drug effects , Ezetimibe/pharmacology , Fatty Liver/drug therapy , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cells, Cultured , Down-Regulation/drug effects , Ezetimibe/therapeutic use , Fatty Liver/metabolism , Female , Hep G2 Cells , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Medicine (Baltimore) ; 95(44): e5155, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27858848

ABSTRACT

Incretin hormone-based therapy in type 2 diabetes has been widely used, and dipepdityl peptidase-4 (DPP-4) inhibitors, which prevent incretin degradation, have become popular oral hypoglycemic agents. The efficacy of DPP-4 inhibitors varies from individuals, and factors determining responses to DPP-4 inhibitors have not been fully established. We aimed to investigate whether genetic variations in glucagon-like peptide (GLP-1) receptor are associated with responses to DPP-4 inhibitors in patients with type 2 diabetes.Genetic variations of rs3765467 in GLP-1 receptor were explored in 246 patients with type 2 diabetes who received DPP-4 inhibitors treatment for 24 weeks in addition to previous medication. Patients with glycated hemoglobin (HbA1c) > 7% and who were naive to any DPP-4 inhibitors were enrolled. Responders were defined as those who showed a > 10% reduction in HbA1c after DPP-4 inhibitor treatment.DPP-4 inhibitors improved glycemic parameters and lipid profiles. Compared to the major genotype (GG), a larger proportion of patients with the minor allele genotype (GA/AA) were responders (P = 0.018), and also showing greater HbA1c reductions (1.3 ±â€Š1.1 vs 0.9 ±â€Š1.2%; P = 0.022). This genetic effect remained significant even after adjustment for other confounding factors (OR = 2.00, 95% CI = 1.03-3.89).Polymorphism in the GLP-1 receptor may influence DPP-4 inhibitor response. Further studies in larger population will help determine the association between genetic variation and interindividual differences in DPP-4 inhibitor therapy.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucagon-Like Peptide-1 Receptor/genetics , Female , Genetic Variation , Humans , Incretins , Male , Middle Aged
8.
PLoS One ; 11(5): e0154683, 2016.
Article in English | MEDLINE | ID: mdl-27135947

ABSTRACT

Beneficial effects of omega-3 fatty acid (O3FA) supplementation in a wide range of disease condition have been well studied. However, there is limited information regarding the effects of O3FAs on chronic kidney disease (CKD), especially in diabetic nephropathy (DN) with hypertriglyceridemia. We investigate whether O3FA supplementation could help maintain renal function in patients with diabetes and hypertriglyceridemia. Total 344 type 2 diabetic patients with a history of O3FA supplementation for managing hypertriglyceridemia were included. Reduction in urine albumin to creatinine ratio (ACR) and glomerular filtrate rate (GFR) were examined. Subgroup analyses were stratified according to the daily O3FA doses. Serum total cholesterol, triglyceride, and urine ACR significantly reduced after O3FA supplementation. Overall, 172 (50.0%) patients did not experience renal function loss, and 125 (36.3%) patients had a GFR with a positive slope. The patients treated with O3FAs at 4g/day showed greater maintenance in renal function than those treated with lower dosages (p < 0.001). This dose dependent effect remains significant after adjustment for multiple variables. O3FA supplementation in diabetic patients with hypertriglyceridemia shows benefits of reducing albuminuria and maintaining renal function. The effects are dependent on the dose of daily O3FA supplementation.


Subject(s)
Diabetes Mellitus/drug therapy , Diabetic Nephropathies/drug therapy , Fatty Acids, Omega-3/therapeutic use , Hypertriglyceridemia/drug therapy , Aged , Albuminuria/drug therapy , Disease Progression , Female , Glomerular Filtration Rate/physiology , Humans , Logistic Models , Male , Middle Aged , Retrospective Studies
9.
Endocrinol Metab (Seoul) ; 31(1): 161-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26754588

ABSTRACT

BACKGROUND: The use of dipeptidyl peptidase-4 (DPP-4) inhibitors is increasing among renal transplant patients with diabetes. However, the glucose-lowering efficacies of various DPP-4 inhibitors and their effects on blood cyclosporine levels have not been fully investigated. We compared the glucose-lowering efficacies of DPP 4 inhibitors and evaluate their effects on the blood levels of cyclosporine in renal transplant recipients with diabetes. METHODS: Sixty-five renal allograft recipients who received treatment with DPP-4 inhibitors (vildagliptin, sitagliptin, or linagliptin) following kidney transplant were enrolled. The glucose-lowering efficacies of the DPP-4 inhibitors were compared according to the changes in the hemoglobin A1c (HbA1c) levels after 3 months of treatment. Changes in the trough levels of the cyclosporine were also assessed 2 months after treatment with each DPP-4 inhibitor. RESULTS: HbA1c significantly decreased in the linagliptin group in comparison with other DPP-4 inhibitors (vildagliptin -0.38%±1.03%, sitagliptin -0.53%±0.95%, and linagliptin -1.40±1.34; P=0.016). Cyclosporine trough levels were significantly increased in the sitagliptin group compared with vildagliptin group (30.62±81.70 ng/mL vs. -24.22±53.54 ng/mL, P=0.036). Cyclosporine trough levels were minimally changed in patients with linagliptin. CONCLUSION: Linagliptin demonstrates superior glucose-lowering efficacy and minimal effect on cyclosporine trough levels in comparison with other DPP-4 inhibitors in kidney transplant patients with diabetes.

10.
Cardiovasc Diabetol ; 14: 148, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26538247

ABSTRACT

BACKGROUND: The heart requires constant sources of energy mostly from free fatty acids (FFA) and glucose. The alteration in myocardial substrate metabolism occurs in the heart of diabetic patients, but its specific association with other metabolic variables remains unclear. We aimed to evaluate glucose uptake in hearts of subjects with normal glucose tolerance (NGT), prediabetes, and type 2 diabetes mellitus (T2DM) using [(18)F]-fluorodeoxyglucose-positron emission tomography ((18)FDG-PET) in association with visceral and subcutaneous adiposity, and metabolic laboratory parameters. METHODS: A total of 346 individuals (NGT, n = 76; prediabetes, n = 208; T2DM, n = 62) in a health promotion center of a tertiary hospital were enrolled. The fasting myocardial glucose uptake, and visceral and subcutaneous fat areas were evaluated using (18)FDG-PET and abdominal computed tomography, respectively. RESULTS: Myocardial glucose uptake was significantly decreased in subjects with T2DM compared to the NGT or prediabetes groups (p for trend = 0.001). Multivariate linear regression analyses revealed that visceral fat area (ß = -0.22, p = 0.018), fasting FFA (ß = -0.39, p < 0.001), and uric acid levels (ß = -0.21, p = 0.007) were independent determinants of myocardial glucose uptake. Multiple logistic analyses demonstrated that decreased myocardial glucose uptake (OR 2.32; 95% CI 1.02-5.29, p = 0.045) and visceral fat area (OR 1.02, 95% CI 1.01-1.03, p = 0.018) were associated with T2DM. CONCLUSIONS: Our findings indicate visceral adiposity is strongly associated with the alteration of myocardial glucose uptake evaluated by (18)FDG-PET, and its association further relates to T2DM.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Heart/diagnostic imaging , Intra-Abdominal Fat/diagnostic imaging , Myocardium/metabolism , Obesity, Abdominal/metabolism , Prediabetic State/metabolism , Subcutaneous Fat/diagnostic imaging , Adult , Aged , Diabetes Mellitus, Type 2/diagnostic imaging , Female , Fluorodeoxyglucose F18 , Humans , Male , Middle Aged , Obesity, Abdominal/diagnostic imaging , Positron-Emission Tomography , Prediabetic State/diagnostic imaging , Radiopharmaceuticals , Tomography, X-Ray Computed
11.
Autophagy ; 11(11): 2089-2101, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26389569

ABSTRACT

Statins (HMGCR/HMG-CoA reductase [3-hydroxy-3-methylglutaryl-CoA reductase] inhibitors) are widely used to lower blood cholesterol levels but have been shown to increase the risk of type 2 diabetes mellitus. However, the molecular mechanism underlying diabetogenic effects remains to be elucidated. Here we show that statins significantly increase the expression of key gluconeogenic enzymes (such as G6PC [glucose-6-phosphatase] and PCK1 (phosphoenolpyruvate carboxykinase 1 [soluble]) in vitro and in vivo and promote hepatic glucose output. Statin treatment activates autophagic flux in HepG2 cells. Acute suppression of autophagy with lysosome inhibitors in statin treated HepG2 cells reduced gluconeogenic enzymes expression and glucose output. Importantly, the ability of statins to increase gluconeogenesis was impaired when ATG7 was deficient and BECN1 was absent, suggesting that autophagy plays a critical role in the diabetogenic effects of statins. Moreover autophagic vacuoles and gluconeogenic genes expression in the liver of diet-induced obese mice were increased by statins, ultimately leading to elevated hepatic glucose production, hyperglycemia, and insulin resistance. Together, these data demonstrate that chronic statin therapy results in insulin resistance through the activation of hepatic gluconeogenesis, which is tightly coupled to hepatic autophagy. These data further contribute to a better understanding of the diabetogenic effects of stains in the context of insulin resistance.

12.
Metabolism ; 64(4): 482-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25312577

ABSTRACT

OBJECTIVE: 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins) are used to control blood cholesterol levels and reduce cardiovascular disease. It has been repeatedly reported that statins may cause new-onset diabetes mellitus (DM). However, limited evidence exists from direct head to head comparisons of statins on whether the risk of DM differs among statins. We investigated the risk of development of new-onset diabetes in subjects treated with different statins. METHODS: We retrospectively enrolled consecutive 3680 patients without DM or impaired fasting glucose who started receiving statin treatment for cholesterol control. We evaluated the incidence of new-onset diabetes according to the type of statin. RESULTS: The mean duration of follow-up was 62.6±15.3 months. The incidence of DM was significantly higher in the pitavastatin group (49 of 628; 7.8%) compared to that in the other statin groups [atorvastatin (68 of 1327; 5.1%), rosuvastatin (77 of 1191; 6.5%), simvastatin (11 of 326; 3.4%), and pravastatin (12 of 298; 5.8%); p=0.041]. The risk of diabetes was the highest in the pitavastatin group compared with that in the simvastatin group [hazard ratio (HR)=2.68, p=0.011]. Other statins showed no significant risk differences compared to that for simvastatin. Fasting blood glucose (FBG) level at baseline and body-mass index (BMI) were associated with the development of diabetes [FBG, HR=1.11, p<0.001; BMI, HR=1.02, p=0.005]. CONCLUSIONS: Among the five statins, pitavastatin showed the strongest effect on the development of new-onset diabetes.


Subject(s)
Diabetes Mellitus/epidemiology , Diabetes Mellitus/etiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Hypercholesterolemia/epidemiology , Aged , Atorvastatin , Female , Fluorobenzenes/therapeutic use , Heptanoic Acids/therapeutic use , Humans , Hypercholesterolemia/complications , Male , Middle Aged , Pravastatin/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Quinolines/therapeutic use , Retrospective Studies , Risk Factors , Rosuvastatin Calcium , Simvastatin/therapeutic use , Sulfonamides/therapeutic use
13.
Diabetes Metab J ; 38(3): 211-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25003075

ABSTRACT

BACKGROUND: We evaluated the effects of two dipeptidyl peptidase-4 (DPP-4) inhibitors, sitagliptin and vildagliptin, on metabolic parameters in patients with type 2 diabetes mellitus. METHODS: A total of 170 type 2 diabetes patients treated with sitagliptin or vildagliptin for more than 24 weeks were selected. The patients were separated into two groups, sitagliptin (100 mg once daily, n=93) and vildagliptin (50 mg twice daily, n=77). We compared the effect of each DPP-4 inhibitor on metabolic parameters, including the fasting plasma glucose (FPG), postprandial glucose (PPG), glycated hemoglobin (HbA1c), and glycated albumin (GA) levels, and lipid parameters at baseline and after 24 weeks of treatment. RESULTS: The HbA1c, FPG, and GA levels were similar between the two groups at baseline, but the sitagliptin group displayed a higher PPG level (P=0.03). After 24 weeks of treatment, all of the glucose-related parameters were significantly decreased in both groups (P=0.001). The levels of total cholesterol and triglycerides were only reduced in the vildagliptin group (P=0.001), although the sitagliptin group received a larger quantity of statins than the vildagliptin group (P=0.002).The mean change in the glucose- and lipid-related parameters after 24 weeks of treatment were not significantly different between the two groups (P=not significant). Neither sitagliptin nor vildagliptin treatment was associated with a reduction in the high sensitive C-reactive protein level (P=0.714). CONCLUSION: Vildagliptin and sitagliptin exert a similar effect on metabolic parameters, but vildagliptin exerts a more potent beneficial effect on lipid parameters.

14.
Transplantation ; 97(4): 419-25, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24285338

ABSTRACT

BACKGROUND: Dysglycemia and dyslipidemia are important metabolic complications of organ transplantation. Statins are widely used to control dyslipidemia; however, long-term use of statins is related to diabetes mellitus (DM) and impaired fasting glucose (IFG). The aim of this study was to evaluate the influence of statins on the development of dysglycemia (IFG and/or DM) in renal allograft recipients. METHODS: A total of 394 patients without previously known DM or IFG who underwent kidney transplantation were enrolled. Patients were grouped into the two groups according to the use of statin (control, n=149; statin, n=245). The major statins used were fluvastatin (80 mg/d, n=134) and atorvastatin (20 mg/d, n=111). We compared the incidence of IFG or DM during the follow-up period. RESULTS: The incidence of IFG was higher in the statin group than that in the control group (28.6% vs. 8.7%, P<0.001). The incidence of dysglycemia was significantly higher in the statin group (40.0% vs. 15.4%, P=0.001). Time to development of dysglycemia after transplantation was shorter in the statin group than in the control group (38.8±29.7 vs. 47.2±23.3 months, P=0.002). Statin use was associated with an increased risk for dysglycemia after adjustment for age, sex, body mass index, hypertension, cholesterol levels, hepatitis C infection, and type of immunosuppressant (hazard ratio=3.08, 95% confidence interval=1.91-4.98). The dysglycemic effect was more profound in the patients who used atorvastatin than in those who used fluvastatin (hazard ratio=2.21, 95% confidence interval=1.02-4.76). CONCLUSION: Statin treatment is associated with an elevation in fasting plasma glucose and in the development of dysglycemia in renal allograft recipients.


Subject(s)
Blood Glucose/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hyperglycemia/etiology , Kidney Transplantation/methods , Renal Insufficiency/complications , Adult , Anticholesteremic Agents/adverse effects , Atorvastatin , Diabetes Complications/diagnosis , Diabetes Mellitus/diagnosis , Dyslipidemias/chemically induced , Fasting , Fatty Acids, Monounsaturated/adverse effects , Female , Fluvastatin , Follow-Up Studies , Glomerular Filtration Rate , Heptanoic Acids/adverse effects , Humans , Hyperglycemia/complications , Immunosuppressive Agents/therapeutic use , Incidence , Indoles/adverse effects , Male , Middle Aged , Pyrroles/adverse effects , Renal Insufficiency/therapy
15.
Metabolism ; 62(5): 677-85, 2013 May.
Article in English | MEDLINE | ID: mdl-23260797

ABSTRACT

OBJECTIVE: The aim of this study was to examine the association between common polymorphisms of the adiponectin gene (ADIPOQ) and microvascular complications in patients with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS: Rs2241766 and rs1501299 of ADIPOQ were genotyped in 708 patients with T2DM. Fundus photography, nerve conducting velocity, and urine analysis were performed to check for the presence of microvascular complications including diabetic nephropathy, retinopathy and neuropathy. RESULTS: The prevalence of diabetic nephropathy tended to be different according to rs2241766 genotype (p=0.057) and the GG genotype of rs2241766 was associated with diabetic nephropathy [urine albumin/creatinine ratio (UACR) greater than 30 mg/g] after adjusting for age, sex, body mass index, duration of diabetes, HDL-cholesterol, smoking status, and blood pressure (odds ratio=1.96; 95% confidence interval=1.01-3.82, p=0.049). Also, the G allele of rs2241766 demonstrated a trend to be associated with an increase in UACR (p=0.087). Rs2241766 genotype was not associated with diabetic retinopathy (p=0.955) and neuropathy (p=0.104) or any diabetic microvascular complications (p=0.104). There was no significant association between the rs1501299 genotype of ADIPOQ and the prevalence of diabetic retinopathy and neuropathy or any diabetic microvascular complications even after adjustment. CONCLUSION: These data suggest that the GG genotype at rs2241766 is implicated in the pathogenesis of risk for diabetic nephropathy defined as UACR greater than 30 mg/day in patients with T2DM.


Subject(s)
Adiponectin/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Aged , Diabetes Mellitus, Type 2/epidemiology , Diabetic Angiopathies/epidemiology , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Microvessels/pathology , Middle Aged , Polymorphism, Single Nucleotide/physiology , Prevalence , Republic of Korea/epidemiology
16.
Metabolism ; 61(4): 496-503, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22154326

ABSTRACT

Reactive oxygen species (ROS), driven by excessive levels of glucose and free fatty acids, appears to induce cell apoptosis. However, the underlying molecular mechanism of this process remains unclear in cardiac myocytes. We investigated the glucolipotoxicity effects of high glucose and palmitic acid (C16:0) on the rat cardiomyoblast cell line (H9c2) focusing on tumor suppressor p53. Cultured H9c2 rat cardiomyoblasts were exposed to palmitate and /or to an elevated glucose concentration for 18 hours. Only the glucolipotoxic condition of 30 mM glucose in combination with 250 µM palmitate resulted in significant generation of ROS and upregulation of p53 which caused to an increased cleavage of caspase-3. On the other hand, the expression of NF-E2-related factor 2 (Nrf2) showed increased tendency while the expression of NAD(P)H: quinone oxidoreductase-1 (NQO1) was decreased. N-acetyl L cysteins and pifithrin-α, an inhibitor of p53 abrogated glucolipotoxicity-induced ROS generation and p53 expression. Chromatin immunoprecipitation analysis revealed that p53 interacted antioxidant responsive elements (ARE)-containing promoter of NQO1. Upregulated p53 counteracted the Nrf2-induced transcription of ARE-containing promoter of NQO1 gene and leaded to decrease in NQO1 expression. We demonstrated that the elevated p53 mediated glucolipotoxicity-induced apoptosis of rat cardiomyoblast cell through dual pathways: stimulating pro-apoptosis signaling as well as suppressing anti-apoptosis pathway of Nrf2-NQO1 signaling.


Subject(s)
Apoptosis/physiology , Glucose/metabolism , Myocytes, Cardiac/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Palmitic Acid/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylcysteine/pharmacology , Animals , Benzothiazoles/pharmacology , Cell Line , Free Radical Scavengers/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species/antagonists & inhibitors , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...