Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Pharmacol ; 14: 1167934, 2023.
Article in English | MEDLINE | ID: mdl-37251332

ABSTRACT

Hachimijiogan (HJG) has originally been used to ameliorate a variety of symptoms associated with low ambient temperatures. However, its pharmacological action in metabolic organs remains unclear. We hypothesized that HJG may modulate metabolic function and have a potential therapeutic application to metabolic diseases. To test this hypothesis, we investigated metabolic action of HJG in mice. Male C57BL/6J mice chronically administered with HJG showed a reduction in adipocyte size with increased transcription of beige adipocyte-related genes in subcutaneous white adipose tissue. HJG-mixed high-fat diet (HFD)-fed mice showed alleviation of HFD-induced weight gain, adipocyte hypertrophy, liver steatosis with a significant reduction in circulating leptin and Fibroblast growth factor 21 despite no changes in food intake or oxygen consumption. Feeding an HJG-mixed HFD following 4-weeks of HFD feeding, while a limited effect on body weight, improved insulin sensitivity with a reversal of decreased circulating adiponectin. In addition, HJG improved insulin sensitivity in the leptin-deficient mice without significant effects on body weight. Treatment with n-butanol soluble extracts of HJG potentiated transcription of Uncoupling protein 1 mediated by ß3-adrenergic agonism in 3T3L1 adipocytes. These findings provide evidence that HJG modulates adipocyte function and may exert preventive or therapeutic effects against obesity and insulin resistance.

2.
Int J Mol Sci ; 23(21)2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36362372

ABSTRACT

Endoplasmic reticulum (ER) stress is a key pathogenic factor in type 1 and 2 diabetes. Glycogen synthase kinase 3 (Gsk-3) contributes to ß-cell loss in mice. However, the mechanism by which Gsk-3 leads ß-cell death remains unclear. ER stress was pharmacologically induced in mouse primary islets and insulinoma cells. We used insulinoma cells derived from Akita mice as a model of genetic ER stress. Gsk-3 activity was blocked by treating with Gsk-3 inhibitors or by introducing catalytically inactive Gsk-3ß. Gsk-3 inhibition prevented proteasomal degradation of activating transcriptional factor 4 (ATF4) and alleviated apoptosis. We found that ATF4-S214 was phosphorylated by Gsk-3, and that this was required for a binding of ATF4 with ßTrCP, which mediates polyubiquitination. The anti-apoptotic effect of Gsk-3 inhibition was attenuated by introducing DN-ATF4 or by knockdown of ATF4. Mechanistically, Gsk-3 inhibition modulated transcription targets of ATF4 and in turn facilitated dephosphorylation of eIF2α, altering the protein translational dynamism under ER stress. These observations were reproduced in the Akita mouse-derived cells. Thus, these results reveal the role of Gsk-3 in the regulation of the integrated stress response, and provide a rationale for inhibiting this enzyme to prevent ß-cell death under ER stress conditions.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Insulinoma , Pancreatic Neoplasms , Mice , Animals , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Signal Transduction , Endoplasmic Reticulum Stress , Apoptosis
3.
Diabetes Care ; 44(4): 1002-1011, 2021 04.
Article in English | MEDLINE | ID: mdl-33627367

ABSTRACT

OBJECTIVE: To elucidate the pathogenesis of postpancreatectomy diabetes mellitus (PPDM). RESEARCH DESIGN AND METHODS: Forty-eight patients without diabetes undergoing either pancreatoduodenectomy (PD) (n = 20) or distal pancreatectomy (DP) (n = 28) were included. A 75-g oral glucose tolerance test was performed every 6 months. Microbiome composition and short-chain fatty acids (SCFAs) in feces were examined before and 6 months after surgery. The association of histological characteristics of the resected pancreas with PPDM was examined. RESULTS: During follow-up (median 3.19 years), 2 of 20 PD patients and 16 of 28 DP patients developed PPDM. Proteobacteria relative abundance, plasma glucagon-like peptide 1 (GLP-1), and fecal butyrate levels increased only after PD. Postsurgical butyrate levels were correlated with postsurgical GLP-1 levels. With no significant difference in the volume of the resected pancreas between the surgical procedures, both ß-cell and α-cell areas in the resected pancreas were significantly higher in DP patients than in PD patients. In DP patients, the progressors to diabetes showed preexisting insulin resistance compared with nonprogressors, and both increased α- and ß-cell areas were predictors of PPDM. Furthermore, in DP patients, α-cell and ß-cell areas were associated with ALDH1A3 expression in islets. CONCLUSIONS: We postulate that a greater removal of ß-cells contributes to the development of PPDM after DP. Islet expansion along with preexisting insulin resistance is associated with high cellular plasticity, which may predict the development of PPDM after DP. In contrast, PD is associated with alterations of gut microbiome and increases in SCFA production and GLP-1 secretion, possibly protecting against PPDM development.


Subject(s)
Diabetes Mellitus , Islets of Langerhans , Cell Plasticity , Glucagon-Like Peptide 1 , Humans , Insulin , Pancreatectomy
4.
JCI Insight ; 6(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33427207

ABSTRACT

Dedifferentiation has been implicated in ß cell dysfunction and loss in rodent diabetes. However, the pathophysiological significance in humans remains unclear. To elucidate this, we analyzed surgically resected pancreatic tissues of 26 Japanese subjects with diabetes and 11 nondiabetic subjects, who had been overweight during adulthood but had no family history of diabetes. The diabetic subjects were subclassified into 3 disease stage categories, early, advanced, and intermediate. Despite no numerical changes in endocrine cells immunoreactive for chromogranin A (ChgA), diabetic islets showed profound ß cell loss, with an increase in α cells without an increase in insulin and glucagon double-positive cells. The proportion of dedifferentiated cells that retain ChgA immunoreactivity without 4 major islet hormones was strikingly increased in diabetic islets and rose substantially during disease progression. The increased dedifferentiated cell ratio was inversely correlated with declining C-peptide index. Moreover, a subset of islet cells converted into exocrine-like cells during disease progression. These results indicate that islet remodeling with dedifferentiation is the underlying cause of ß cell failure during the course of diabetes progression in humans.


Subject(s)
Cell Dedifferentiation , Diabetes Mellitus, Type 2/pathology , Islets of Langerhans/pathology , Adult , Age Factors , Aged , Aged, 80 and over , Case-Control Studies , Cell Dedifferentiation/physiology , Chromogranin A/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Progression , Female , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/metabolism , Male , Middle Aged , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology
5.
Diabetologia ; 61(10): 2189-2201, 2018 10.
Article in English | MEDLINE | ID: mdl-30054673

ABSTRACT

AIMS/HYPOTHESIS: Loss of functional beta cells results in a gradual progression of insulin insufficiency in Wolfram syndrome caused by recessive WFS1 mutations. However, beta cell dysfunction in Wolfram syndrome has yet to be fully characterised, and there are also no specific treatment recommendations. In this study, we aimed to characterise beta cell secretory defects and to examine the potential effects of a glucagon-like peptide-1 (GLP-1) receptor agonist on diabetes in Wolfram syndrome. METHODS: Insulin secretory function was assessed by the pancreatic perfusion method in mice used as a model of Wolfram syndrome. In addition, granule dynamics in living beta cells were examined using total internal reflection fluorescence microscopy. Acute and chronic effects of exendin-4 (Ex-4) on glucose tolerance and insulin secretion were examined in young Wfs1-/- mice without hyperglycaemia. Molecular events associated with Ex-4 treatment were investigated using pancreatic sections and isolated islets. In addition, we retrospectively observed a woman with Wolfram syndrome who had been treated with liraglutide for 24 weeks. RESULTS: Treatment with liraglutide ameliorated our patient's glycaemic control and resulted in a 20% reduction of daily insulin dose along with an off-drug elevation of fasting C-peptide immunoreactivity. Glucose-stimulated first-phase insulin secretion and potassium-stimulated insulin secretion decreased by 53% and 59%, respectively, in perfused pancreases of 10-week-old Wfs1-/- mice compared with wild-type (WT) mice. The number of insulin granule fusion events in the first phase decreased by 41% in Wfs1-/- beta cells compared with WT beta cells. Perfusion with Ex-4 increased insulin release in the first and second phases by 3.9-fold and 5.6-fold, respectively, in Wfs1-/- mice compared with perfusion with saline as a control. The physiological relevance of the effects of Ex-4 was shown by the fact that a single administration potentiated glucose-stimulated insulin secretion and improved glucose tolerance in Wfs1-/- mice. Four weeks of administration of Ex-4 resulted in an off-drug amelioration of glucose excursions after glucose loading in Wfs1-/- mice, with insulin secretory dynamics that were indistinguishable from those in WT mice, despite the fact that there was no alteration in beta cell mass. In association with the functional improvements, Ex-4 treatment reversed the increases in phosphorylated eukaryotic initiation factor (EIF2α) and thioredoxin interacting protein (TXNIP), and the decrease in phosphorylated AMP-activated kinase (AMPK), in the beta cells of the Wfs1-/- mice. Furthermore, Ex-4 treatment modulated the transcription of oxidative and endoplasmic reticulum stress-related markers in isolated islets, implying that it was able to mitigate the cellular stresses resulting from Wfs1 deficiency. CONCLUSIONS/INTERPRETATION: Our study provides deeper insights into the pathophysiology of beta cell dysfunction caused by WFS1 deficiency and implies that activation of the GLP-1 receptor signal may alleviate insulin insufficiency and aid glycaemic control in Wolfram syndrome.


Subject(s)
Glucagon-Like Peptide-1 Receptor/metabolism , Insulin-Secreting Cells/cytology , Wolfram Syndrome/metabolism , Adult , Animals , Endoplasmic Reticulum/metabolism , Exenatide/pharmacology , Female , Glucose/chemistry , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Liraglutide/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Perfusion , Retrospective Studies , Signal Transduction/drug effects
6.
J Diabetes Res ; 2017: 3605178, 2017.
Article in English | MEDLINE | ID: mdl-28168202

ABSTRACT

Type 2 diabetes mellitus (T2DM) results from pancreatic ß-cell failure in the setting of insulin resistance. In the early stages of this disease, pancreatic ß-cells meet increased insulin demand by both enhancing insulin-secretory capacity and increasing ß-cell mass. As the disease progresses, ß-cells fail to maintain these compensatory responses. This involves both extrinsic signals and mediators intrinsic to ß-cells, which adversely affect ß-cells by impairing insulin secretion, decreasing proliferative capacities, and ultimately causing apoptosis. In recent years, it has increasingly been recognized that changes in circulating levels of various factors from other organs play roles in ß-cell dysfunction and cellular loss. In this review, we discuss current knowledge of interorgan communications underlying ß-cell failure during the progression of T2DM.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Adipocytes/metabolism , Animals , Humans , Insulin Secretion
7.
Diabetologia ; 59(12): 2711-2721, 2016 12.
Article in English | MEDLINE | ID: mdl-27695899

ABSTRACT

AIMS/HYPOTHESIS: Soluble insulin receptor (sIR), the ectodomain of the insulin receptor (IR), has been detected in human plasma and its concentration paralleled that of blood glucose. We have previously developed an in vitro model using HepG2 liver-derived cells, which mimics changes in sIR levels in plasma from diabetic patients and shows that calcium-dependent proteases cleave IR extracellularly (a process known as shedding). The present study aimed to reveal the mechanisms of IR cleavage. METHODS: Using the in vitro model, we investigated the molecular mechanisms of IR cleavage, which is accelerated by high-glucose treatment. We also analysed the relationship between IR cleavage and cellular insulin resistance, and the correlation between plasma sIR levels and insulin sensitivity, which was assessed by the euglycaemic-hyperinsulinaemic clamp technique. RESULTS: Here, we determined that calpain 2, which is secreted into the extracellular space associated with exosomes, directly cleaved the ectodomain of the IRß subunit (IRß), which in turn promoted intramembrane cleavage of IRß by γ-secretase. IR cleavage impaired insulin signalling and the inhibition of IR cleavage (by knockdown of calpain 2 and γ-secretase), restored IR substrate-1 and Akt, independent of IR. Furthermore, the glucose-lowering drug, metformin, prevented IR cleavage accompanied by inhibition of calpain 2 release in exosomes, and re-established insulin signalling. In patients with type 2 diabetes, plasma sIR levels inversely correlated with insulin sensitivity. CONCLUSIONS/INTERPRETATION: Sequential cleavage of IR by calpain 2 and γ-secretase may contribute to insulin signalling in cells and its inhibition may be partly responsible for the glucose-lowering effects of metformin. Thus, IR cleavage may offer a new mechanism for the aetiology of insulin resistance.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Calpain/metabolism , Receptor, Insulin/metabolism , Amyloid Precursor Protein Secretases/genetics , Blotting, Western , Calpain/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Enzyme-Linked Immunosorbent Assay , Exosomes/metabolism , Hep G2 Cells , Humans , Immunoprecipitation , Insulin Resistance/genetics , Insulin Resistance/physiology , RNA, Small Interfering , Receptor, Insulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...