Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Nutr Biochem ; 26(3): 219-26, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25488546

ABSTRACT

To assess the molecular mechanisms by which dietary restriction preserves the ß-cell mass and function in diabetic db/db mice. Male db/db mice were divided into two groups with or without diet restriction. Daily food intake of db/db mice was adjusted to that of the control db/m mice, which was determined in advance. A dietary restriction was implemented for 6 weeks from 6 weeks of age. Islet morphology, ß-cell function and gene expression profiles specific for pancreatic islet cells were compared. Food intake in db/m mice was 50% of that in db/db mice. Impaired glucose tolerance and insulin sensitivity were significantly ameliorated in db/db mice with dietary restriction. The pancreatic ß-cell mass was greater in mice with dietary restriction than that in mice without intervention. The dietary restriction significantly increased cyclin D gene expression and down-regulated CAD gene expression at 12 weeks compared with untreated db/db mice. Antiapoptotic bcl-2 gene expression was significantly increased, whereas genes related to oxidative stress, ER stress and inflammatory processes, such as NADPH oxidase, CHOP10 and TNF, were markedly down-regulated in mice with dietary restriction. Dietary restriction preserved the pancreatic ß-cell function and ß-cell mass in diabetic db/db mice, suggesting that alimentary therapy prevented ß-cell loss by suppressing cellular apoptosis and antioxidative stress in the pancreatic ß cells.


Subject(s)
Apoptosis , Caloric Restriction , Diabetes Mellitus, Type 2/diet therapy , Endoplasmic Reticulum Stress , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Oxidative Stress , Animals , Cell Proliferation , Cell Size , Crosses, Genetic , Cyclin D/genetics , Cyclin D/metabolism , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gene Expression Profiling , Gene Expression Regulation , Insulin/blood , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Kinetics , Male , Matched-Pair Analysis , Mice, Mutant Strains , Poly-ADP-Ribose Binding Proteins
2.
Mol Cell Endocrinol ; 349(2): 281-8, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22108438

ABSTRACT

Based on the hypothesis that MIN6 cells could produce glucagon-like peptide-1 (GLP-1) to maintain cell survival, we analyzed the effects of GLP-1 receptor agonist, exendin-4 (Ex4), and antagonist, exendin-(9-39) (Ex9) on cell function and cell differentiation. MIN6 cells expressed proglucagon mRNAs and produced GLP-1, which was accelerated by Ex4 and suppressed by Ex9. Moreover, Ex4 further enhanced glucose-stimulated GLP-1 secretion, suggesting autocrine loop-contributed amplification of the GLP-1 signal. Ex4 up-regulated cell differentiation- and cell function-related CREBBP, Pdx-1, Pax6, proglucagon, and PC1/3 gene expressions. The confocal laser scanning images revealed that GLP-1 positive cells were dominant in the early stage of cells, but positive for insulin were more prominent in the mature stage of cells. Ex4 accelerated cell viability, while Ex9 and anti-GLP-1 receptor antibody enhanced cell apoptosis. MIN6 cells possess a mechanism of GLP-1 signal amplification in an autocrine fashion, by which the cells maintained insulin production and cell survival.


Subject(s)
Glucagon-Like Peptide 1/biosynthesis , Insulin/metabolism , Receptors, Glucagon/metabolism , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Autocrine Communication , CREB-Binding Protein/biosynthesis , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Exenatide , Eye Proteins/biosynthesis , Glucagon-Like Peptide-1 Receptor , Glucose/metabolism , Glucose/pharmacology , Homeodomain Proteins/biosynthesis , Humans , Insulin Secretion , Microscopy, Confocal , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Peptide Fragments/pharmacology , Peptides/pharmacology , Proglucagon/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Glucagon/antagonists & inhibitors , Repressor Proteins/biosynthesis , Signal Transduction , Trans-Activators/biosynthesis , Venoms/pharmacology
3.
Mol Endocrinol ; 26(1): 95-109, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22108800

ABSTRACT

The phosphatidylinositol 3-kinase signaling pathway in vascular endothelial cells is important for systemic angiogenesis and glucose metabolism. In this study, we addressed the precise role of the 3-phosphoinositide-dependent protein kinase 1 (PDK1)-regulated signaling network in endothelial cells in vivo, using vascular endothelial PDK1 knockout (VEPDK1KO) mice. Surprisingly, VEPDK1KO mice manifested enhanced glucose tolerance and whole-body insulin sensitivity due to suppression of their hepatic glucose production with no change in either peripheral glucose disposal or even impaired vascular endothelial function at 6 months of age. When mice were fed a standard diet at 6 months of age and a high-fat diet at 3 months of age, hypertrophy of epididymal adipose tissues was inhibited, adiponectin mRNA was significantly increased, and mRNA of MCP1, leptin, and TNFα was decreased in the white adipose tissue of VEPDK1KO mice in comparison with controls. Consequently, both the circulating adiponectin levels and the activity of hepatic AMP-activated protein kinase were significantly increased, subsequently enhancing whole-body insulin sensitivity and energy expenditure with increased hepatic fatty acid oxidation in VEPDK1KO mice. These results provide the first in vivo evidence that lowered angiogenesis through the deletion of PDK1 signaling not only interferes with the growth of adipose tissue but also induces increased energy expenditure due to amelioration of the adipocytokine profile. This demonstrates an unexpected role of PDK1 signaling in endothelial cells on the maintenance of proper glucose homeostasis through the regulation of adipocyte development.


Subject(s)
Endothelial Cells/metabolism , Insulin Resistance , Intra-Abdominal Fat/metabolism , Neovascularization, Physiologic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Adiponectin/biosynthesis , Adiponectin/blood , Adiponectin/genetics , Adipose Tissue, White/metabolism , Animals , Chemokine CCL2/biosynthesis , Glucose/metabolism , Leptin/biosynthesis , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Knockout , Obesity/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Protein Serine-Threonine Kinases/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis
4.
Am J Physiol Endocrinol Metab ; 298(2): E278-86, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19920213

ABSTRACT

Pioglitazone preserves pancreatic beta-cell morphology and function in diabetic animal models. In this study, we investigated the molecular mechanisms by which pioglitazone protects beta-cells in diabetic db/db mice. In addition to the morphological analysis of the islets, gene expression profiles of the pancreatic islet were analyzed using laser capture microdissection and were compared with real-time RT-PCR of db/db and nondiabetic m/m mice treated with or without pioglitazone for 2 wk or 2 days. Pioglitazone treatment (2 wk) ameliorated dysmetabolism, increased islet insulin content, restored glucose-stimulated insulin secretion, and preserved beta-cell mass in db/db mice but had no significant effects in m/m mice. Pioglitazone upregulated genes that promote cell differentiation/proliferation in diabetic and nondiabetic mice. In db/db mice, pioglitazone downregulated the apoptosis-promoting caspase-activated DNase gene and upregulated anti-apoptosis-related genes. The above-mentioned effects of pioglitazone treatment were also observed after 2 days of treatment. By contrast, the oxidative stress-promoting NADPH oxidase gene was downregulated, and antioxidative stress-related genes were upregulated, in db/db mice treated with pioglitazone for 2 wk, rather than 2 days. Morphometric results for proliferative cell number antigen and 4-hydroxy-2-noneal modified protein were consistent with the results of gene expression analysis. The present results strongly suggest that pioglitazone preserves beta-cell mass in diabetic mice mostly by two ways; directly, by acceleration of cell differentiation/proliferation and suppression of apoptosis (acute effect); and indirectly, by deceleration of oxidative stress because of amelioration of the underlying metabolic disorder (chronic effect).


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Thiazolidinediones/pharmacology , Animals , Apoptosis/physiology , Cell Differentiation/drug effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Gene Expression Profiling , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Obese , Obesity/complications , Obesity/metabolism , Organ Size/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , PPAR gamma/drug effects , PPAR gamma/metabolism , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , Pioglitazone
5.
Intern Med ; 48(17): 1545-9, 2009.
Article in English | MEDLINE | ID: mdl-19721301

ABSTRACT

A 29-year-old woman was diagnosed as having type 1 diabetes mellitus and received insulin aspart and NPH insulin (NovolinN). On day 22, she had leg edema and right abdominal pain. The serum hepatobiliary enzyme levels were markedly elevated. Computed tomography revealed gallbladder edema. After an injection of human regular insulin and NPH insulin (HumacartN), the elevated liver enzyme levels were no longer observed. Challenge testing demonstrated that protamine was the cause of her allergy. Furthermore, tests revealed increased VEGF levels. This is an extremely rare case with a delayed-type protamine allergy caused by NovolinN resulting in gallbladder edema.


Subject(s)
Diabetes Mellitus, Type 1 , Drug Hypersensitivity/diagnosis , Edema/diagnosis , Gallbladder Diseases/diagnosis , Hypersensitivity, Delayed/diagnosis , Insulin/adverse effects , Adult , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Drug Hypersensitivity/complications , Edema/etiology , Female , Gallbladder Diseases/etiology , Humans , Hypersensitivity, Delayed/complications
6.
Endocr J ; 56(8): 997-1008, 2009.
Article in English | MEDLINE | ID: mdl-19706988

ABSTRACT

The db gene homozygous, but not heterozygous, mice develop diabetes with severe beta-cell damage. We investigated the molecular mechanism underlying db gene-associated pancreatic beta-cell dysfunction. Islet morphology, beta-cell function, and gene expression profiles specific for pancreatic islet cells were compared among db gene homozygous(db/db), heterozygous (db/m) and unrelated m/m mice. The beta-cell ratio decreased in db/db mice with age, but not in non-diabetic db/m and m/m mice. The islet insulin content was lower, but the triglyceride content was higher in db/db than other mice. The islet cell specific gene expression profiles analyzed by laser capture microdissection method and morphological findings suggested an augmentation of beta-cell apoptosis, oxidative stress and ER stress in db/db mice. Interestingly, insulin I and II, anti-apoptotic bcl-2, and proliferation promoting ERK-1 gene expressions were significantly upregulated in db/m mice. An impaired glucose tolerance was shown in m/m mice fed a high fat diet, but not in db/m mice, in which a higher insulin response and increased beta-cell mass were observed. Expressions of insulin I and II, bcl-2, and ERK-1 gene were increased in db/m mice, but not in m/m fed a high fat diet. The present results strongly suggest that the db gene heterozygote, but not homozygote, acquires a compensatory mechanism suppressing beta-cell apoptosis and augmenting the capacity of beta-cell function.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/physiology , Receptors, Leptin/physiology , Animals , Apoptosis/genetics , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Genetic Predisposition to Disease , Glucose/pharmacology , Heterozygote , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreas/physiopathology , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
7.
Circ J ; 73(3): 562-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19194047

ABSTRACT

BACKGROUND: Direct measurement of plasma nitric oxide (NO) concentration is possible with a newly developed catheter-type sensor. METHODS AND RESULTS: Adult mongrel dogs (n=5) were fed a high-fat diet (120 kcal . kg(-1) . day(-1)) for 8 months, then endothelial function was assessed by the change in NO concentration induced by acetylcholine (ACh) (DeltaNO). Simultaneously, average peak velocity (APV) was obtained by Doppler guide wire. Although fasting plasma glucose levels did not change after high-fat diet, fasting plasma insulin levels increased significantly (103+/-36 vs 106+/-27 mg/dl, P=0.89 and 0.15+/-0.15 vs 0.26+/-0.07 ng/ml, P=0.04, respectively). ACh-induced peak APV after high-fat feeding was not significantly different from that at baseline (ACh 0.1 microg/kg; 43+/-17 vs 51+/-7 cm/s, P=NS, ACh 0.4 microg/kg; 45+/-20 vs 47+/-16 cm/s, P=NS, respectively). The DeltaNO was significantly smaller after high-fat diet than at baseline (ACh 0.1 microg/kg; 2.6+/-1.6 vs 1.0+/-0.5 nmol/L, P=0.03, ACh 0.4 microg/kg; 3.8+/-2.3 vs 1.8+/-1.1 nmol/L, P=0.04, respectively). CONCLUSIONS: In high-fat-diet-induced obese dogs NO production was impaired in the early stage when the coronary flow response to ACh may be preserved.


Subject(s)
Cardiac Catheterization/methods , Coronary Vessels/physiology , Endothelium, Vascular/metabolism , Nitric Oxide/blood , Obesity/metabolism , Acetylcholine/pharmacology , Animals , Blood Glucose/metabolism , Cardiac Catheterization/instrumentation , Coronary Circulation/drug effects , Coronary Circulation/physiology , Dietary Fats/pharmacology , Dogs , Female , Insulin/metabolism , Male , Obesity/physiopathology , Vasodilator Agents/pharmacology
8.
Endocr J ; 56(1): 45-53, 2009.
Article in English | MEDLINE | ID: mdl-18845907

ABSTRACT

MIN6 cells retains glucose-stimulated insulin secretion (GSIS) as isolated islets. We comprehensively evaluated the gene expression and production of other islet hormones in MIN6 cells. Islet hormones were demonstrated by immunohistochemical staining and measured by ELISA. The gene expression profiles of MIN6 cells were compared with those in the mouse islets obtained by the laser capture micro-dissection (LCM). MIN6 cells excreted insulin, glucagon, somatostatin and ghrelin. They expressed mRNAs of insulin I and II, proglucagon, somatostatin, pancreatic polypeptide (PP) and ghrelin which were shown in the mouse pancreatic islet core and periphery obtained by LCM. A variety of genes closely related to the islet hormone producing cells were expressed in MIN6. Confocal laser scanning microscopy revealed that MIN6 cells included not only insulin positive cells but also insulin and glucagon or somatostin double positive cells. Glucagon, somatostatin and ghrelin were detectable in the culture medium. The present study clearly demonstrated that MIN6 produce pancreatic endocrine cells. It would be possible to use this cell line as a model to research the development, cell differentiation and function of pancreatic islets.


Subject(s)
Cell Line/metabolism , Cell Line/physiology , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Pancreatic Hormones/metabolism , Animals , Cell Differentiation , Cell Line/cytology , Cell Proliferation , Cell Separation , Gene Expression Profiling , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Pancreatic Hormones/genetics
9.
Diabetes Res Clin Pract ; 81(1): 13-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18455831

ABSTRACT

Adiponectin is a fat-derived cytokine with anti-diabetic and anti-atherogenic properties. In this study, effects of sulfonylureas (SUs) on adiponectin production and the action mechanism were evaluated using 3T3-L1 adipocytes. The cells were incubated with glimepiride, glibenclamide, gliclazide, pioglitazone, metformin and the medium only as the control. In the control, the adiponectin level evaluated as the production rate per 24 h was not changed, while pioglitazone significantly increased the adiponectin level. SUs also increased the adiponectin level, but metformin failed to show any increase in adiponectin production. SUs induced adiponectin gene expression as well as pioglitazone. Pioglitazone significantly increased adipogenesis, but glimepiride did not. The aP2 gene expression was increased by pioglitazone, but not by glimepiride. Forskolin, a protein kinase A stimulator, reduced the adiponectin production stimulated by glimepiride but not by pioglitazone. These observations strongly suggest that SUs stimulate the adiponectin production through a different mechanism from pioglitazone, namely an interaction with protein kinase A activity. The significance of the extrapancreatic action of SUs observed in this study should be further evaluated in the clinical field.


Subject(s)
Adipocytes/physiology , Adiponectin/biosynthesis , Sulfonylurea Compounds/pharmacology , Thiazolidinediones/pharmacology , 3T3 Cells , Adipocytes/drug effects , Animals , Mice , Pioglitazone
11.
Diabetes Res Clin Pract ; 77 Suppl 1: S220-3, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17467108

ABSTRACT

A necessary condition of advocated criteria to determine the metabolic syndrome (MetS) in Japan is waist circumference (WC), which varies among races. In this study, we measured WC and visceral fat area (VFA) in subjects with type 2 diabetes (T2DM) and assessed the propriety of new criteria of MetS in Japan. Four hundred and nineteen patients (M/F: 258/161, age: 60.4+/-0.7 years, BMI: 24.4+/-0.2 kg/m(2)) who received abdominal CT examination were analyzed, and 178 (M/F: 111/67) subjects sufficed the criteria of MetS. Average VFA was significantly larger in subjects with MetS (162+/-3 cm(2) versus 82+/-3 cm(2), p<0.01). The WC and VFA were correlated significantly in both male (r=0.78, p<0.001) and female (r=0.82, p<0.001), and corresponding VFA at 85 cm of WC in male and at 90 cm in female were 125 cm(2) and 120 cm(2). Incidence of cardio- and cerebro-vascular diseases (CVD) was not different between subjects with and without MetS. The present cross-sectional study strongly suggests that the recommended WC is not suitable to define the current criteria of MetS (VFA, > or =100 cm(2)) and its criteria is not appropriate to segregate a risk of CVD in Japanese T2DM subjects. Further prospective analysis should be required to validate the criteria and clinical significance of MetS in T2DM.


Subject(s)
Body Size , Diabetes Mellitus, Type 2/physiopathology , Metabolic Syndrome/diagnosis , Abdomen , Adipose Tissue/anatomy & histology , Aged , Diabetes Mellitus, Type 2/diagnostic imaging , Diagnosis, Differential , Female , Humans , Japan , Male , Metabolic Syndrome/diagnostic imaging , Middle Aged , Tomography, X-Ray Computed , Viscera
12.
Am J Physiol Endocrinol Metab ; 288(3): E510-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15522998

ABSTRACT

To evaluate preventive effects of pioglitazone on pancreatic beta-cell damage in C57BL/KsJ db/db mice, an obese diabetic animal model, the pancreatic islets were compared morphologically between pioglitazone-treated (100 mg/kg daily po) and untreated db/db mice (n = 7 for each) after a 12-wk intervention (6-18 wk of age). The fasting blood glucose level was significantly improved by the treatment with pioglitazone (260 +/- 12 vs. 554 +/- 62 mg/dl, P < 0.05). The islet mass in the pancreas was significantly greater in pioglitazone-treated mice than in untreated mice (10.2 +/- 1.1 vs. 4.6 +/- 0.2 mg, P < 0.01). Subsequently, biochemical and physiological analyses of the beta-cell function were employed using pioglitazone-treated and untreated db/db mice (n = 6 for each) and pioglitazone-treated and untreated db/+ mice (n = 6 for each). After 2 wk of treatment (10-12 wk of age), the plasma levels of triglyceride and free fatty acid were significantly decreased, whereas the plasma adiponectin level increased significantly compared with the untreated group (65.2 +/- 18.0 vs. 18.3 +/- 1.3 microg/ml, P < 0.05). Pioglitazone significantly reduced the triglyceride content in the islets (43.3 +/- 3.6 vs. 65.6 +/- 7.6 ng/islet, P < 0.05) with improved glucose-stimulated insulin secretion. Pioglitazone showed no significant effects on the biochemical and physiological parameters in db/+ mice. The present study first demonstrated that pioglitazone prevents beta-cell damage in an early stage of the disease progression in db/db mice morphologically and physiologically. Our results suggest that pioglitazone improves glucolipotoxicity by increasing insulin sensitivity and reducing fat accumulation in the pancreatic islets.


Subject(s)
Diabetes Mellitus, Type 2/prevention & control , Islets of Langerhans/drug effects , Thiazolidinediones/pharmacology , Adiponectin , Animals , Blood Glucose/drug effects , Blood Glucose/genetics , Blood Glucose/metabolism , Body Weight/drug effects , Body Weight/genetics , Cell Count , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Glucose/pharmacology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/blood , Insulin/metabolism , Insulin/pharmacology , Insulin Secretion , Intercellular Signaling Peptides and Proteins/blood , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pioglitazone , Receptors, Cell Surface/genetics , Receptors, Leptin , Thiazolidinediones/therapeutic use , Triglycerides/blood , Triglycerides/metabolism
14.
Diabetes Care ; 27(6): 1276-80, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161775

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the effect of apolipoprotein (apo)E4 allele on plasma LDL cholesterol response to calorie-restricted diet therapy in type 2 diabetic patients. RESEARCH DESIGN AND METHODS: Twenty-four diabetic patients with the apoE3/3 genotype and 11 diabetic patients with the apoE4/3 genotype were recruited. Participants were hospitalized for calorie-restricted diet therapy (25.0 kcal. kg body wt(-1). day(-1)) for 14 days. Body weight, fasting plasma glucose (FPG) levels, and plasma lipid levels on hospital days 1 and 14 were compared between the two apoE genotype groups. RESULTS: There were no significant differences in baseline FPG levels, HbA(1c) levels, BMI, and plasma levels of total cholesterol, triglyceride, and HDL cholesterol between the two apoE genotype groups, but baseline plasma levels of LDL cholesterol were significantly higher in the apoE4/3 group than in the apoE3/3 group. Body weight decreased slightly and FPG levels decreased significantly after diet therapy in both apoE genotype groups. In the apoE3/3 group, only plasma levels of triglyceride decreased significantly after diet therapy, whereas in the apoE4/3 group, plasma levels of triglyceride, total cholesterol, and LDL cholesterol decreased significantly after diet therapy. The decrease (percentage of change) in total cholesterol (-16.3 vs. -6.6%) and LDL cholesterol (-15.6 vs. -0.7%) after diet therapy was significantly greater in the apoE4/3 group than in the apoE3/3 group. CONCLUSIONS: Calorie-restricted diet therapy is more effective in reducing plasma LDL cholesterol in type 2 diabetic patients with the apoE4 allele.


Subject(s)
Apolipoproteins E/genetics , Cholesterol, LDL/blood , Diabetes Mellitus, Type 2/diet therapy , Diabetes Mellitus, Type 2/genetics , Diet, Diabetic , Apolipoprotein E4 , Cholesterol, LDL/genetics , Diabetes Mellitus, Type 2/blood , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...