Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Diabetes Complications ; 38(6): 108746, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749296

ABSTRACT

AIMS: This study aimed to investigate the relationship between changes in glucose metabolism and body composition in patients with diabetes. METHODS: We included 380 patients with type 2 diabetes, who underwent bioelectrical impedance analysis, in this longitudinal study. Changes in HbA1c (ΔHbA1c) levels and body composition indices were compared between baseline and 6 months. A multivariate analysis was performed to examine the relationship between ΔHbA1c and changes in body composition. RESULTS: HbA1c levels were significantly decreased at 6 months (P < 0.01), but there was no significant change in BMI. A linear multiple regression analysis showed that ΔHbA1c was negatively correlated with changes in muscle mass (ß = -0.18; P = 0.047) and bone mineral content (ß = -0.28; P < 0.001), but there was no significant association between ΔHbA1c levels and a change in body fat percentage. CONCLUSIONS: This study shows a limited association between short-term changes in glucose metabolism and changes in body composition in patients with type 2 diabetes. Therefore, interventions aimed at reducing adiposity may not affect glucose metabolism in the short term, while interventions focused on maintaining or enhancing muscle mass and bone mineral content may play an important role in diabetes management.


Subject(s)
Body Composition , Bone Density , Diabetes Mellitus, Type 2 , Glycated Hemoglobin , Muscle, Skeletal , Humans , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Male , Glycated Hemoglobin/analysis , Glycated Hemoglobin/metabolism , Female , Middle Aged , Body Composition/physiology , Aged , Longitudinal Studies , Muscle, Skeletal/metabolism , Bone Density/physiology , Electric Impedance , Blood Glucose/metabolism , Blood Glucose/analysis , Adiposity/physiology , Body Mass Index
2.
Diabetol Int ; 15(1): 109-116, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38264231

ABSTRACT

Introduction: This study aimed to investigate the association between scan frequency and intermittently scanned continuous glucose monitoring (isCGM) metrics and to clarify the factors affecting scan frequency in adults with type 1 diabetes mellitus (T1D). Methods: We enrolled adults with T1D who used FreeStyle® Libre. Scan and self-monitoring of blood glucose (SMBG) frequency and CGM metrics from the past 90-day glucose data were collected. The receiver operating characteristic curve was plotted to obtain the optimal cutoff values of scan frequency for the target values of time in range (TIR), time above range (TAR), and time below range (TBR). Results: The study was conducted on 211 adults with T1D (mean age, 50.9 ± 15.2 years; male, 40.8%; diabetes duration, 16.4 ± 11.9 years; duration of CGM use, 2.1 ± 1.0 years; and mean HbA1c, 7.6 ± 0.9%). The average scan frequency was 10.5 ± 3.3 scan/day. Scan frequency was positively correlated with TIR and negatively correlated with TAR, although it was not significantly correlated with TBR. Scan frequency was positively correlated with the hypoglycemia fear survey-behavior score, while it was negatively correlated with some glycemic variability metrics. Adult patients with T1D and good exercise habits had a higher scan frequency than those without exercise habits. The AUC for > 70% of the TIR was 0.653, with an optimal cutoff of 11 scan/day. Conclusions: In real-world conditions, frequent scans were linked to improved CGM metrics, including increased TIR, reduced TAR, and some glycemic variability metrics. Exercise habits and hypoglycemia fear-related behavior might affect scan frequency. Our findings could help healthcare professionals use isCGM to support adults with T1D.Clinical Trial Registry No. UMIN000039376.

3.
Diabetol Int ; 14(4): 339-343, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37781466

ABSTRACT

Phase angle, obtained using bioelectrical impedance analysis, non-invasively reflects the whole-body cellular condition and nutritional status and may be helpful as a prognostic factor. Patients with diabetes had a smaller phase angle than healthy subjects. However, the clinical significance of phase angle has not yet been elucidated. Therefore, the purpose of this study was to clarify the relationship between phase angle and HbA1c in patients with diabetes and the clinical relevance of phase angle. A retrospective, multicenter, cross-sectional study was conducted with Japanese patients with diabetes. Body composition was determined with bioelectrical impedance analysis, and this was used to obtain phase angle. Phase angle was assessed in relation to clinical parameters, body composition parameters, and HbA1c levels. A total of 655 patients were enrolled (400 men and 255 women, aged 57.1 ± 14.8 years, body mass index 25.6 ± 5.2 kg/m2, HbA1c 8.1 ± 1.9%). Even in patients with diabetes, the phase angle was higher in men than in women and did not differ between the types of diabetes. Multiple regression analysis, performed with phase angle as the objective variable, and age, sex, diabetes type, HbA1c, albumin level, and body mass index as explanatory variables, revealed that phase angle was negatively affected by HbA1c (B = - 0.043, 95% Confidence interval: - 0.07 to - 0.02, p < 0.001). HbA1c, age, sex, albumin level, and body mass index were independent determinants of phase angle in participants with diabetes.

4.
J Diabetes Investig ; 14(4): 582-590, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36789495

ABSTRACT

AIMS/INTRODUCTION: The discrepancy between HbA1c and glucose exposure may have significant clinical implications; however, the association between the hemoglobin glycation index (HGI) and clinical parameters in type 1 diabetes remains controversial. This study aimed to find the factors associated with HGI (laboratory HbA1c - predicted HbA1c derived from the continuous glucose monitoring [CGM]). MATERIALS AND METHODS: We conducted a cross-sectional study of adults with type 1 diabetes (n = 211, age 50.9 ± 15.2 years old, female sex = 59.2%, duration of CGM use = 2.1 ± 1.0 years). All subjects wore the CGM for 90 days before HbA1c measurement. Data derived from the FreeStyle Libre sensor were used to calculate the glucose management indicator (GMI) and glycemic variability (GV) parameters. HGI was defined as the difference between the GMI and the laboratory HbA1c levels. The participants were divided into three groups according to the HGI tertile (low, moderate, and high). Multivariate regression analyses were performed. RESULTS: The female sex ratio, HbA1c, and % coefficient of variation (%CV) significantly increased over the HGI tertile, while eGFR and Hb decreased over the HGI tertile. In multivariate analysis, the factors associated with HGI were %CV and eGFR, after adjusting for HbA1c level and sex (R2  = 0.44). CONCLUSIONS: This study demonstrated that HGI is associated with female sex, eGFR, and some glycemic variability indices, independently of HbA1c. Minimizing glycemic fluctuations might reduce HGI. This information provides diabetic health professionals and patients with personalized diabetes management for adults with type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Humans , Adult , Female , Middle Aged , Aged , Diabetes Mellitus, Type 1/complications , Glycated Hemoglobin , Diabetes Mellitus, Type 2/complications , Blood Glucose/analysis , Maillard Reaction , Blood Glucose Self-Monitoring , Japan/epidemiology , Cross-Sectional Studies , Hemoglobins/analysis
5.
Intern Med ; 62(18): 2607-2615, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-36631091

ABSTRACT

Objective This study investigated self-monitoring of blood glucose (SMBG) adherence and flash glucose monitoring patterns using a cluster analysis in Japanese type 1 diabetes (T1D) patients with intermittently scanned continuous glucose monitoring (isCGM). Methods We measured SMBG adherence and performed a data-driven cluster analysis using a hierarchical clustering in T1D patients from Japan using the FreeStyle Libre system. Clusters were based on three variables (testing glucose frequency and referred Libre data for hyperglycemia or hypoglycemia). Patients We enrolled 209 participants. Inclusion criteria were patients with T1D, duration of isCGM use ≥3 months, age ≥20 years old, and regular attendance at the collaborating center. Results The rate of good adherence to SMBG recommended by a doctor was 85.0%. We identified three clusters: cluster 1 (low SMBG test frequency but high reference to Libre data, 17.7%), cluster 2 (high SMBG test frequency but low reference to Libre data, 34.0%), and cluster 3 (high SMBG test frequency and high reference to Libra data, 48.3%). Compared with other clusters, individuals in cluster 1 were younger, those in cluster 2 had a shorter Libre duration, and individuals in cluster 3 had lower time-in-range, higher severe diabetic distress, and high intake of snacks and sweetened beverages. There were no marked differences in the incidence of diabetic complications and rate of wearing the Libre sensor among the clusters. Conclusion We stratified the patients into three subgroups with varied clinical characteristics and CGM metrics. This new substratification might help tailor diabetes management of patients with T1D using isCGM.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 1 , Humans , Young Adult , Adult , Diabetes Mellitus, Type 1/epidemiology , Blood Glucose Self-Monitoring/methods , Japan/epidemiology , Cluster Analysis , Hypoglycemic Agents
6.
Article in English | MEDLINE | ID: mdl-36593661

ABSTRACT

INTRODUCTION: Emerging evidence suggests that diabetes stigma and negative emotions associated with it may impair the quality of life of people with diabetes. Among these psychological distresses, shame is considered the most distressing of all human emotional experiences and may be a condition to which diabetes clinicians should pay attention. This epidemiological study focused on diabetes-related shame and aimed to determine the prevalence of diabetes-related shame, its factors, and its association with psychological indicators. RESEARCH DESIGN AND METHODS: A cross-sectional online survey was conducted among people with type 2 diabetes preregistered with a research firm. The questionnaire included experience of diabetes-related shame and demographic data such as age, clinical characteristic measures such as hemoglobin A1c (HbA1c), and psychological indicators, including the WHO Five Well-Being Index (WHO-5) and Problem Areas In Diabetes-5 (PAID-5). Differences in each indicator between people with diabetes who experienced shame and those who did not were analyzed with the unpaired t-test. As supplemental analysis, binomial logistic regression analysis was used to identify factors associated with the prevalence of diabetes-related shame. RESULTS: Of the 510 participants, 32.9% experienced diabetes-related shame and 17.5% concealed their disease from colleagues or friends. Those who had experienced diabetes-related shame showed significantly lower WHO-5 and higher PAID-5 scores (p<0.001). However, no significant difference was found in HbA1c (p=0.36). Binomial logistic regression revealed that women, young adults, those without a college degree, those with low self-efficacy, and those with a strong sense of financial burden or external pressure were at higher risk of experiencing diabetes-related shame. CONCLUSIONS: Among people with type 2 diabetes mellitus, diabetes-related shame was associated with diabetes-specific emotional distress and low psychological well-being. Further research and care development are needed to address diabetes-related shame and improve the quality of life of people with diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Shame , Female , Humans , Young Adult , Cross-Sectional Studies , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/psychology , Glycated Hemoglobin , Quality of Life , Stress, Psychological
7.
Diabetol Int ; 12(2): 197-206, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33786274

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors often increase the hematocrit. It remains unclear whether this increase would be observed in all patients administered SGLT2 inhibitors, however. We therefore used the data from the previous study and investigated time-dependent alterations of various outcomes related to erythrocytes, erythropoiesis, and clinical outcome in type 2 diabetes subjects (n = 89) treated with ipragliflozin for 16 weeks. Among a total of 89 participants, 71 subjects (80.0% of total participants) showed the elevation of the hematocrit and 18 subjects (20.0% of total participants) did not at 16 weeks. Although the hematocrit levels at baseline were significantly lower in hematocrit-elevated group than non-elevated group, they reached the same levels 4 weeks after the onset of treatment. Binomial logistic regression analysis demonstrated that a lower baseline hematocrit level was related to the elevation of hematocrit at 16 weeks. Optimal cutoff hematocrit levels at baseline to predict hematocrit elevation were 46.9% (male) and 41.7% (female) in ROC analysis. Random intercept model analysis revealed the serum erythropoietin level increased in both hematocrit-elevated and non-elevated groups, whereas only the former group showed an increase in the percentage of reticulocytes during the first 4 weeks. These results suggest that the ipragliflozin-induced increase in hematocrit which is affected by the baseline hematocrit level is attributable to the responsiveness to, but not to the production of, erythropoietin. Collectively, Ht elevation observed in administration of SGLT2 inhibitors can result from erythropoietin-induced erythropoiesis, which is determined by the pre-treatment Ht level. Trial registration: This trial has been registered with University Hospital Medical Information Network Clinical Trial Registry (UMIN-CTR no. 000015478).

8.
JCI Insight ; 5(9)2020 05 07.
Article in English | MEDLINE | ID: mdl-32376799

ABSTRACT

EIF2AK4, which encodes the amino acid deficiency-sensing protein GCN2, has been implicated as a susceptibility gene for type 2 diabetes in the Japanese population. However, the mechanism by which GCN2 affects glucose homeostasis is unclear. Here, we show that insulin secretion is reduced in individuals harboring the risk allele of EIF2AK4 and that maintenance of GCN2-deficient mice on a high-fat diet results in a loss of pancreatic ß cell mass. Our data suggest that GCN2 senses amino acid deficiency in ß cells and limits signaling by mechanistic target of rapamycin complex 1 to prevent ß cell failure during the consumption of a high-fat diet.


Subject(s)
Amino Acids/analysis , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Liver , Protein Serine-Threonine Kinases , Adult , Animals , Cell Line, Tumor , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Genetic Predisposition to Disease , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Middle Aged , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Rats
9.
J Diabetes Investig ; 10(5): 1254-1261, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30688412

ABSTRACT

AIMS/INTRODUCTION: Sodium-glucose cotransporter 2 (SGLT-2) inhibitors improve blood glucose control, as well as reducing bodyweight by promoting urinary glucose excretion. The weight loss is less than expected from urinary glucose loss, however, likely because of an increase in food intake. To investigate whether SGLT-2 inhibitors might increase appetite by affecting related hormones, we examined the effects of the SGLT-2 inhibitor, ipragliflozin, including those on appetite-regulating hormones, in individuals with suboptimally controlled type 2 diabetes. MATERIALS AND METHODS: The present prospective, multicenter, open-label study was carried out with 96 patients with a body mass index of ≥22 kg/m2 who were treated with ipragliflozin (50 mg/day) for 16 weeks. Parameters including glycated hemoglobin level, bodyweight, circulating leptin and active ghrelin concentrations, and appetite as assessed with a visual analog scale were measured before and during treatment. RESULTS: Both glycated hemoglobin level (from 7.9 ± 0.8 to 7.1 ± 0.7%) and bodyweight (from 75.2 ± 12.6 to 72.6 ± 12.4 kg) were significantly decreased after treatment for 16 weeks. The fasting serum leptin level was significantly decreased after 2 weeks (from 19.5 ± 13.1 to 18.1 ± 12.4 ng/mL) and remained decreased up to 16 weeks, even after adjustment for bodyweight, whereas the plasma active ghrelin level showed no significant change. The visual analog scale score for hunger was significantly increased at 2 and 8 weeks. CONCLUSIONS: The present results suggest that ipragliflozin improved glycemic control and reduced bodyweight, but also reduced serum leptin levels and might thereby have increased appetite.


Subject(s)
Appetite/drug effects , Diabetes Mellitus, Type 2/drug therapy , Ghrelin/blood , Glucosides/therapeutic use , Glycemic Index/drug effects , Leptin/blood , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Thiophenes/therapeutic use , Biomarkers/analysis , Blood Glucose/metabolism , Body Mass Index , Body Weight/drug effects , Eating/drug effects , Female , Follow-Up Studies , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Weight Loss/drug effects
10.
Endocrine ; 64(1): 43-47, 2019 04.
Article in English | MEDLINE | ID: mdl-30406884

ABSTRACT

PURPOSE: Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are categorized as short- or long-acting types, but information regarding differences in the effects of these two types on postprandial glucose disposal has been limited. We have now investigated the effects of exenatide and liraglutide (short- and long-acting GLP-1RAs, respectively) on glucose disposal during an oral glucose tolerance test (OGTT). METHODS: Fourteen healthy volunteers with normal glucose tolerance underwent three OGTTs, which were performed without pharmacological intervention or after a single administration of exenatide or liraglutide at 30 min and 10 h, respectively, before test initiation. The three OGTTs were performed with intervals of at least 7 days between successive tests and within a period of 2 months. RESULTS: Exenatide, but not liraglutide, markedly decelerated the peak of both plasma glucose and serum insulin levels during the OGTT, with the peaks of both glucose and insulin concentrations occurring at 150 min after test initiation with exenatide compared with 30 min in the control condition or with liraglutide. Exenatide and liraglutide reduced the area under the curve for plasma glucose levels during the OGTT by similar extents, whereas that for serum insulin levels was reduced only by exenatide. CONCLUSIONS: Our results suggest that exenatide decelerates the increase in plasma glucose levels through inhibition of glucose absorption and that it exerts an insulin-sparing action after glucose challenge.


Subject(s)
Blood Glucose , Exenatide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/blood , Liraglutide/pharmacology , Adult , Female , Glucagon-Like Peptide-1 Receptor/agonists , Glucose Tolerance Test , Humans , Male , Young Adult
11.
J Diabetes Investig ; 10(3): 577-590, 2019 May.
Article in English | MEDLINE | ID: mdl-30290061

ABSTRACT

AIMS/INTRODUCTION: The preservation of pancreatic ß-cell mass is an essential factor in the onset and development of type 2 diabetes mellitus. Recently, sodium-glucose cotransporter 2 inhibitors have been launched as antihyperglycemic agents, and their organ-protective effects are attracting attention. They are also reported to have favorable effects on the preservation of pancreatic ß-cell mass, but the appropriate timing for the administration of sodium-glucose cotransporter 2 inhibitors is obscure. MATERIALS AND METHODS: In the present study, we administered a sodium-glucose cotransporter 2 inhibitor, dapagliflozin, to an animal model of type 2 diabetes mellitus, db/db mice, and investigated the adequate timing and duration for its administration. We also carried out microarray analysis using pancreatic islets from db/db mice. RESULTS: We found that dapagliflozin preserved pancreatic ß-cell mass depending on the duration of administration and markedly improved blood glucose levels. If the duration was the same, the earlier administration of dapagliflozin was more effective in preserving pancreatic ß-cell mass, increasing serum insulin levels and improving blood glucose levels. From microarray analysis, we discovered that the expression of Agr2, Tff2 and Gkn3 was significantly upregulated after the early administration of dapagliflozin. This upregulated gene expression might provide a legacy effect for the preservation of pancreatic ß-cell mass. CONCLUSIONS: We expect that the early administration of dapagliflozin would provide a long-lasting effect in preserving pancreatic ß-cell mass.


Subject(s)
Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Glucosides/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Biomarkers/analysis , Blood Glucose/analysis , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/drug effects , Male , Mice
12.
Kobe J Med Sci ; 64(2): E43-E55, 2018 Sep 11.
Article in English | MEDLINE | ID: mdl-30381726

ABSTRACT

Endoplasmic reticulum (ER) stress leads to peripheral insulin resistance and the progression of pancreatic beta cell failure in type 2 diabetes. Although ER stress plays an important role in the pathogenesis of diabetes, it is indispensable for cellular activity. Therefore, when assessing the pathological significance of ER stress, it is important to monitor and quantify ER stress levels. Here, we have established a novel system to monitor ER stress levels quickly and sensitively, and using this method, we have clarified the effect of differences in glucose concentration and various fatty acids on the ER of pancreatic ß cells. First, we developed a cell system that secretes Gaussia luciferase in culture medium depending on the activation of the GRP78 promoter. This system could sensitively monitor ER stress levels that could not be detected with real-time RT-PCR and immunoblotting. This system revealed that hyperglycemia does not induce unfolded protein response (UPR) in a short period of time in MIN6 cells, a mouse pancreatic ß cell line. Physiological concentrations of palmitic acid, a saturated fatty acid, induced ER stress quickly, while physiological concentrations of oleic acid, an unsaturated fatty acid, did not. Docosahexaenoic acid, an n-3 unsaturated fatty acid, inhibited palmitic acid-induced ER stress. In this study, we have established a system that can sensitively detect ER stress levels of living cells in a short period of time. This system can be used to monitor the state of the ER in living cells and lead to the investigation of the significance of physiological or pathological ER stress levels.


Subject(s)
Docosahexaenoic Acids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Palmitic Acid/antagonists & inhibitors , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Endoplasmic Reticulum Chaperone BiP , Glucose/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Mice , Oleic Acid/toxicity , Palmitic Acid/toxicity , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
13.
Diabetes Ther ; 9(6): 2399-2406, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30341665

ABSTRACT

INTRODUCTION: Administered basal insulin markedly influences the fasting plasma glucose (FPG) level of individuals with type 1 diabetes. Insulin degludec (IDeg) and insulin glargine U300 (IGlar U300) are now available as ultra-long-acting insulin formulations, but whether or how their glucose-stabilizing effects differ remains unclear. We will compare the effects of these basal insulins on parameters related to blood glucose control, with a focus on day-to-day glycemic variability, in individuals with type 1 diabetes treated with multiple daily injections. METHODS: A multicenter, randomized, open-label, crossover, comparative study (Kobe Best Basal Insulin Study 2) will be performed at 13 participating institutions in Japan. A total of 46 C-peptide-negative adult outpatients with type 1 diabetes will be randomly assigned 1:1 by a centralized allocation process to IGlar U300 (first period)/IDeg (second period) or IDeg (first period)/IGlar U300 (second period) groups, in which subjects will be treated with the corresponding basal insulin for consecutive 4-week periods. The basal insulin will be titrated to achieve an FPG of less than 130 mg/dL initially and then less than 110 mg/dL if feasible. In the last week of each period, plasma glucose will be determined seven times a day by self-monitoring of blood glucose (SMBG) and intraday and day-to-day glucose excursions will be determined by flash glucose monitoring (FGM). The primary end point is comparison of day-to-day glycemic variability as evaluated by the standard deviation (SD) of FPG during the last week of each treatment period. Secondary end points include the coefficient of variance of FPG, the frequency of severe hypoglycemia as evaluated by SMBG, the duration of hypoglycemia as evaluated by FGM, intraday glycemic variability calculated from both SMBG and FGM data, and the administered insulin dose. PLANNED OUTCOMES: The results of the study will be submitted for publication in a peer-reviewed journal to report differences in the effects of two ultra-long-acting basal insulins, IDeg and IGlar U300. CONCLUSION: This head-to-head comparison will be the first study to compare the effects of IDeg and IGlar U300 on day-to-day FPG variability in C-peptide-negative individuals with type 1 diabetes. TRIAL REGISTRATION: Registered in University Hospital Medical Information Network (UMIN) Clinical Trials Registry as 000029630 on 20 June 2017. FUNDING: Novo Nordisk Pharma Ltd.

14.
Biochem Biophys Res Commun ; 497(1): 451-456, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29448105

ABSTRACT

During the development of type 2 diabetes, endoplasmic reticulum (ER) stress leads to pancreatic ß cell failure. CCAAT/enhancer-binding protein (C/EBP) ß is highly induced by ER stress and AMP-activated protein kinase (AMPK) suppression in pancreatic ß cells, and its accumulation reduces pancreatic ß cell mass. We investigated the phosphorylation state of C/EBPß under these conditions. Casein kinase 2 (CK2) was found to co-localize with C/EBPß in MIN6 cells. It phosphorylated S222 of C/EBPß, a previously unidentified phosphorylation site. We found that C/EBPß is phosphorylated by CK2 under AMPK suppression and ER stress, which are important from the viewpoint of the worsening pathological condition of type 2 diabetes, such as decreased insulin secretion and apoptosis of pancreatic ß cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Casein Kinase II/metabolism , Endoplasmic Reticulum Stress/physiology , Insulin-Secreting Cells/metabolism , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cell Line , Mice , Phosphorylation
15.
PLoS One ; 12(9): e0184435, 2017.
Article in English | MEDLINE | ID: mdl-28886131

ABSTRACT

Recent studies demonstrated that insulin signaling plays important roles in the regulation of pancreatic ß cell mass, the reduction of which is known to be involved in the development of diabetes. However, the mechanism underlying the alteration of insulin signaling in pancreatic ß cells remains unclear. The involvement of epigenetic control in the onset of diabetes has also been reported. Thus, we analyzed the epigenetic control of insulin receptor substrate 2 (IRS2) expression in the MIN6 mouse insulinoma cell line. We found concomitant IRS2 up-regulation and enhanced insulin signaling in MIN6 cells, which resulted in an increase in cell proliferation. The H3K9 acetylation status of the Irs2 promoter was positively associated with IRS2 expression. Treatment of MIN6 cells with histone deacetylase inhibitors led to increased IRS2 expression, but this occurred in concert with low insulin signaling. We observed increased IRS2 lysine acetylation as a consequence of histone deacetylase inhibition, a modification that was coupled with a decrease in IRS2 tyrosine phosphorylation. These results suggest that insulin signaling in pancreatic ß cells is regulated by histone deacetylases through two novel pathways affecting IRS2: the epigenetic control of IRS2 expression by H3K9 promoter acetylation, and the regulation of IRS2 activity through protein modification. The identification of the histone deacetylase isoform(s) involved in these mechanisms would be a valuable approach for the treatment of type 2 diabetes.


Subject(s)
Histone Deacetylases/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Signal Transduction , Acetylation , Animals , Cell Line, Tumor , Cell Proliferation , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Gene Expression , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Phosphorylation , Promoter Regions, Genetic , Signal Transduction/drug effects
16.
PLoS One ; 10(6): e0130757, 2015.
Article in English | MEDLINE | ID: mdl-26091000

ABSTRACT

During the development of type 2 diabetes, endoplasmic reticulum (ER) stress leads to not only insulin resistance but also to pancreatic beta cell failure. Conversely, cell function under various stressed conditions can be restored by reducing ER stress by activating AMP-activated protein kinase (AMPK). However, the details of this mechanism are still obscure. Therefore, the current study aims to elucidate the role of AMPK activity during ER stress-associated pancreatic beta cell failure. MIN6 cells were loaded with 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR) and metformin to assess the relationship between AMPK activity and CCAAT enhancer binding protein ß (C/EBPß) expression levels. The effect of C/EBPß phosphorylation on expression levels was also investigated. Vildagliptin and metformin were administered to pancreatic beta cell-specific C/EBPß transgenic mice to investigate the relationship between C/EBPß expression levels and AMPK activity in the pancreatic islets. When pancreatic beta cells are exposed to ER stress, the accumulation of the transcription factor C/EBPß lowers the AMP/ATP ratio, thereby decreasing AMPK activity. In an opposite manner, incubation of MIN6 cells with AICAR or metformin activated AMPK, which suppressed C/EBPß expression. In addition, administration of the dipeptidyl peptidase-4 inhibitor vildagliptin and metformin to pancreatic beta cell-specific C/EBPß transgenic mice decreased C/EBPß expression levels and enhanced pancreatic beta cell mass in proportion to the recovery of AMPK activity. Enhanced C/EBPß expression and decreased AMPK activity act synergistically to induce ER stress-associated pancreatic beta cell failure.


Subject(s)
AMP-Activated Protein Kinases/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Endoplasmic Reticulum Stress/physiology , AMP-Activated Protein Kinases/genetics , Adamantane/analogs & derivatives , Adamantane/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitriles/pharmacology , Phosphorylation/drug effects , Pyrrolidines/pharmacology , Ribonucleotides/pharmacology , Vildagliptin
17.
Proc Natl Acad Sci U S A ; 112(27): 8332-7, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26100882

ABSTRACT

Genetic factors are important determinants of the onset and progression of diabetes mellitus. Numerous susceptibility genes for type 2 diabetes, including potassium voltage-gated channel, KQT-like subfamily Q, member1 (KCNQ1), have been identified in humans by genome-wide analyses and other studies. Experiments with genetically modified mice have also implicated various genes in the pathogenesis of diabetes. However, the possible effects of the parent of origin for diabetes susceptibility alleles on disease onset have remained unclear. Here, we show that a mutation at the Kcnq1 locus reduces pancreatic ß-cell mass in mice by epigenetic modulation only when it is inherited from the father. The noncoding RNA KCNQ1 overlapping transcript1 (Kcnq1ot1) is expressed from the Kcnq1 locus and regulates the expression of neighboring genes on the paternal allele. We found that disruption of Kcnq1 results in reduced Kcnq1ot1 expression as well as the increased expression of cyclin-dependent kinase inhibitor 1C (Cdkn1c), an imprinted gene that encodes a cell cycle inhibitor, only when the mutation is on the paternal allele. Furthermore, histone modification at the Cdkn1c promoter region in pancreatic islets was found to contribute to this phenomenon. Our observations suggest that the Kcnq1 genomic region directly regulates pancreatic ß-cell mass and that genomic imprinting may be a determinant of the onset of diabetes mellitus.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/genetics , Epigenesis, Genetic , Insulin-Secreting Cells/metabolism , KCNQ1 Potassium Channel/genetics , Mutation , Alleles , Animals , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Gene Expression , Genomic Imprinting/genetics , Glucose/pharmacology , Glucose Tolerance Test , Immunoblotting , Inheritance Patterns , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , KCNQ1 Potassium Channel/metabolism , Male , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Biophys Res Commun ; 458(3): 681-686, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25686499

ABSTRACT

A high-fat diet (HF) is associated with obesity, insulin resistance, and hyperglycemia. Animal studies have shown compensatory mechanisms in pancreatic ß-cells after high fat load, such as increased pancreatic ß-cell mass, enhanced insulin secretion, and exocytosis. However, the effects of high fat intake on insulin synthesis are obscure. Here, we investigated whether insulin synthesis was altered in correlation with an HF diet, for the purpose of obtaining further understanding of the compensatory mechanisms in pancreatic ß-cells. Mice fed an HF diet are obese, insulin resistant, hyperinsulinemic, and glucose intolerant. In islets of mice fed an HF diet, more storage of insulin was identified. We analyzed insulin translation in mouse islets, as well as in INS-1 cells, using non-radioisotope chemicals. We found that insulin translational levels were significantly increased in islets of mice fed an HF diet to meet systemic demand, without altering its transcriptional levels. Our data showed that not only increased pancreatic ß-cell mass and insulin secretion but also elevated insulin translation is the major compensatory mechanism of pancreatic ß-cells.


Subject(s)
Diet, High-Fat/adverse effects , Hyperinsulinism/complications , Insulin-Secreting Cells/pathology , Insulin/genetics , Obesity/complications , Protein Biosynthesis , Animals , Cell Line , Hyperinsulinism/genetics , Hyperinsulinism/pathology , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/genetics , Obesity/pathology , Transcription, Genetic
19.
Pancreatology ; 14(5): 361-5, 2014.
Article in English | MEDLINE | ID: mdl-25278305

ABSTRACT

BACKGROUND/OBJECTIVES: Many patients with autoimmune pancreatitis (AIP) have an association with diabetes mellitus. It has not been clarified whether steroid therapy for AIP improves or worsens the condition of diabetes mellitus. The aim of this study was thus to investigate the relationship between pancreatic atrophy after steroid therapy and the clinical course of diabetes. METHODS: Thirty-one AIP patients, who were treated by steroid therapy, were included in this study during December 2005 to March 2013. Pancreatic atrophy 6 months after the beginning of steroid therapy was defined to be present when the width of the pancreatic body was less than 10 mm. The relationships between pancreatic atrophy and patient characteristics as well as the course of diabetes were examined. RESULTS: Steroid therapy was effective in all treated patients. Pancreatic atrophy was observed in 12 patients and not in 19 patients after the steroid therapy. AIP patients with pancreatic atrophy showed higher incidences of diabetes mellitus (p = 0.001, 9/12 vs. 2/19), diabetes control worsening (p = 0.007, 7/12 vs. 2/17), and new onset of diabetes (p = 0.02, 5/7 vs. 1/18) than those without atrophy. It was not associated with gender, other organ involvement, pattern of pancreas swelling (diffuse/focal), serum IgG4 level, alcohol intake, and pancreatic calcification on CT. Patients with new onset of diabetes needed insulin therapy, even in the maintenance therapy of AIP. CONCLUSIONS: AIP patients with pancreatic atrophy after steroid therapy have a high incidence of diabetes mellitus. New onset of diabetes is closely associated with pancreatic atrophy after steroid therapy.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Autoimmune Diseases/drug therapy , Diabetes Mellitus, Type 2/etiology , Pancreas/pathology , Pancreatitis/drug therapy , Prednisolone/therapeutic use , Adult , Aged , Anti-Inflammatory Agents/pharmacology , Atrophy , Autoimmune Diseases/complications , Autoimmune Diseases/pathology , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreas/drug effects , Pancreatitis/complications , Pancreatitis/immunology , Pancreatitis/pathology , Prednisolone/pharmacology
20.
J Diabetes Investig ; 5(1): 48-50, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24843736

ABSTRACT

To estimate the carbohydrate-to-insulin ratio (CIR), a formula dividing a constant, usually 300-500, by the total daily dose (TDD) of insulin, is widely utilized. An appropriate CIR varies for each meal of the day, however. Here, we investigate diurnal variation of CIR in hospitalized Japanese type 1 diabetic patients treated with continuous subcutaneous insulin infusion. After optimization of the insulin dose, TDD and total basal insulin dose (TBD) were 34.9 ± 10.2 and 9.3 ± 2.8 units, respectively, with a percentage of TBD to TDD of 27.3 ± 6.0%. The products of CIR and TDD at breakfast, lunch and dinner were 311 ± 63, 530 ± 161, and 396 ± 63, respectively, suggesting that in the formula estimating CIR using TDD, the constant should vary for each meal of the day, and that 300, 500, and 400 are appropriate for breakfast, lunch, and dinner, respectively.

SELECTION OF CITATIONS
SEARCH DETAIL
...