Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Endocrinol Metab ; 326(5): E663-E672, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38568150

ABSTRACT

Despite the fact that genes and the environment are known to play a central role in islet function, our knowledge of how these parameters interact to modulate insulin secretory function remains relatively poor. Presently, we performed ex vivo glucose-stimulated insulin secretion and insulin content assays in islets of 213 mice from 13 inbred mouse strains on chow, Western diet (WD), and a high-fat, carbohydrate-free (KETO) diet. Strikingly, among these 13 strains, islets from the commonly used C57BL/6J mouse strain were the least glucose responsive. Using matched metabolic phenotyping data, we performed correlation analyses of isolated islet parameters and found a positive correlation between basal and glucose-stimulated insulin secretion, but no relationship between insulin secretion and insulin content. Using in vivo metabolic measures, we found that glucose tolerance determines the relationship between ex vivo islet insulin secretion and plasma insulin levels. Finally, we showed that islet glucose-stimulated insulin secretion decreased with KETO in almost all strains, concomitant with broader phenotypic changes, such as increased adiposity and glucose intolerance. This is an important finding as it should caution against the application of KETO diet for beta-cell health. Together these data offer key insights into the intersection of diet and genetic background on islet function and whole body glucose metabolism.NEW & NOTEWORTHY Thirteen strains of mice on chow, Western diet, and high-fat, carbohydrate-free (KETO), correlating whole body phenotypes to ex vivo pancreatic islet functional measurements, were used. The study finds a huge spectrum of functional islet responses and insulin phenotypes across all strains and diets, with the ubiquitous C57Bl/6J mouse exhibiting the lowest secretory response of all strains, highlighting the overall importance of considering genetic background when investigating islet function. Ex vivo basal and stimulated insulin secretion are correlated in the islet, and KETO imparts widescale downregulation of islet insulin secretion.


Subject(s)
Diet, High-Fat , Insulin Secretion , Insulin , Islets of Langerhans , Mice, Inbred C57BL , Animals , Mice , Islets of Langerhans/metabolism , Insulin Secretion/physiology , Insulin/metabolism , Insulin/blood , Male , Diet, Western , Glucose/metabolism , Diet, Carbohydrate-Restricted , Mice, Inbred Strains , Blood Glucose/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/genetics
2.
Am J Physiol Endocrinol Metab ; 326(2): E134-E147, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38117265

ABSTRACT

Vertical sleeve gastrectomy (VSG) restores glucose homeostasis in obese mice and humans. In addition, the increased fibroblast growth factor (FGF)15/19 circulating level postsurgery has been implicated in this effect. However, the impact of FGF15/19 on pancreatic islets remains unclear. Using a diet-induced obese mice model, we demonstrate that VSG attenuates insulin hypersecretion in isolated pancreatic islets, likely due to morphological alterations in the endocrine pancreas such as reduction in islet, ß-cell, and α-cell mass. In addition, VSG relieves gene expression of endoplasmic reticulum (ER) stress and inflammation markers in islets from obese mice. Incubation of INS-1E ß-cells with serum from obese mice induced dysfunction and cell death, whereas these conditions were not induced with serum from obese mice submitted to VSG, implicating the involvement of a humoral factor. Indeed, VSG increased FGF15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E cells treated with the serum from these mice. Moreover, exposing INS-1E cells to an FGFR inhibitor abolished the effects of VSG serum on insulin secretion and cell death. Also, recombinant FGF19 prevents INS-1E cells from dysfunction and death induced by serum from obese mice. These findings indicate that the amelioration of glucose-insulin homeostasis promoted by VSG is mediated, at least in part, by FGF15/19. Therefore, approaches promoting FGF15/19 release or action may restore pancreatic islet function in obesity.NEW & NOTEWORTHY Vertical sleeve gastrectomy (VSG) decreases insulin secretion, endoplasmic reticulum (ER) stress, and inflammation in pancreatic islets from obese mice. In addition, VSG increased fibroblast growth factor (FGF)15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E ß-cells treated with the serum from these mice. Serum from operated mice protects INS-1E cells from dysfunction and apoptosis, which was mediated by FGF15/19.


Subject(s)
Insulin-Secreting Cells , Insulin , Mice , Humans , Animals , Insulin/metabolism , Mice, Obese , Insulin-Secreting Cells/metabolism , Glucose/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Gastrectomy , Inflammation/metabolism , Homeostasis
3.
Postgrad Med J ; 100(1187): 686-691, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-38646729

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the association of fasting C-peptide and glucagon with diabetic peripheral neuropathy (DPN) in patients with type 2 diabetes (T2DM). METHODS: A comprehensive evaluation was conducted on 797 patients with T2DM to assess the various risk factors affecting DPN. The subjects were categorized into short duration and long duration group according to the duration of diabetes with a threshold of 10 years. Logistic regression analysis was employed to examine the association between DPN and islet function, as well as other parameters. Receiver operating characteristic curve analysis was performed to evaluate the predictive capability of glucagon. RESULTS: The fasting C-peptide levels were significantly lower in the DPN patients with short duration of diabetes, but lost significance in the long duration group. Conversely, a decreased level of glucagon was only observed in DPN patients with long duration of diabetes. For the group with long duration of diabetes, glucagon was the sole risk factor associated with DPN. The receiver operating characteristic curve analysis revealed that glucagon in the long duration group exhibited a moderate area under the curve of 0.706. CONCLUSIONS: The serum glucagon levels in T2DM patients with DPN exhibited bidirectional changes based on the duration of diabetes. Decreased glucagon was associated with DPN in T2DM patients with long duration of diabetes.


Subject(s)
C-Peptide , Diabetes Mellitus, Type 2 , Diabetic Neuropathies , Glucagon , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/blood , Male , Female , Diabetic Neuropathies/blood , Glucagon/blood , Middle Aged , C-Peptide/blood , Risk Factors , Aged , Time Factors , ROC Curve , Fasting/blood
4.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614300

ABSTRACT

Type 2 diabetes (T2D) represents a multifactorial metabolic disease with a strong genetic predisposition. Despite elaborate efforts in identifying the genetic variants determining individual susceptibility towards T2D, the majority of genetic factors driving disease development remain poorly understood. With the aim to identify novel T2D risk genes we previously generated an N2 outcross population using the two inbred mouse strains New Zealand obese (NZO) and C3HeB/FeJ (C3H). A linkage study performed in this population led to the identification of the novel T2D-associated quantitative trait locus (QTL) Nbg15 (NZO blood glucose on chromosome 15, Logarithm of odds (LOD) 6.6). In this study we used a combined approach of positional cloning, gene expression analyses and in silico predictions of DNA polymorphism on gene/protein function to dissect the genetic variants linking Nbg15 to the development of T2D. Moreover, we have generated congenic strains that associated the distal sublocus of Nbg15 to mechanisms altering pancreatic beta cell function. In this sublocus, Cbx6, Fam135b and Kdelr3 were nominated as potential causative genes associated with the Nbg15 driven effects. Moreover, a putative mutation in the Kdelr3 gene from NZO was identified, negatively influencing adaptive responses associated with pancreatic beta cell death and induction of endoplasmic reticulum stress. Importantly, knockdown of Kdelr3 in cultured Min6 beta cells altered insulin granules maturation and pro-insulin levels, pointing towards a crucial role of this gene in islets function and T2D susceptibility.


Subject(s)
Diabetes Mellitus, Type 2 , Genetic Predisposition to Disease , Obesity , Receptors, Peptide , Animals , Mice , Diabetes Mellitus, Type 2/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice, Inbred C3H , Mice, Obese , Obesity/genetics , Receptors, Peptide/genetics
5.
Molecules ; 28(9)2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37175224

ABSTRACT

The pharmacological potential of industrial hemp (Cannabis sativa) has been widely studied. However, the majority of studies have focused on cannabidiol, isolated from the inflorescence and leaf of the plant. In the present study, we evaluated the anti-diabetic potential of hemp root water (HWE) and ethanol extracts (HEE) in streptozotocin (STZ)-induced insulin-deficient diabetic mice. The administration of HWE and HEE ameliorated hyperglycemia and improved glucose homeostasis and islet function in STZ-treated mice (p < 0.05). HWE and HEE suppressed ß-cell apoptosis and cytokine-induced inflammatory signaling in the pancreas (p < 0.05). Moreover, HWE and HEE normalized insulin-signaling defects in skeletal muscles and apoptotic response in the liver and kidney induced by STZ (p < 0.05). Gas chromatography-mass spectrometry analysis of HWE and HEE showed possible active compounds which might be responsible for the observed anti-diabetic potential. These findings indicate the possible mechanisms by which hemp root extracts protect mice against insulin-deficient diabetes, and support the need for further studies geared towards the application of hemp root as a novel bioactive material.


Subject(s)
Cannabis , Diabetes Mellitus, Experimental , Mice , Animals , Cannabis/chemistry , Insulin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/chemically induced , Plant Extracts/therapeutic use , Pancreas , Streptozocin/pharmacology
6.
Biochem Biophys Res Commun ; 619: 9-14, 2022 09 03.
Article in English | MEDLINE | ID: mdl-35728283

ABSTRACT

BACKGROUND: and Purpose: To investigate the biological role of interferon α/ß receptor 2 (IFNAR2) in type 1 diabetes (T1D). METHODS: First, IFNAR2 mRNA and protein expression levels in serum of T1D patients and healthy controls were detected by RT-qPCR and Western blot. For experimental studies, 80 male C57BL/6 mice were randomly divided into 4 groups with 20 mice in each group: the control group, the T1D group, the T1D + ad-con group and the T1D + ad-si-IFNAR2 group. The T1D mouse model was generated by multiple intraperitoneal injections of small doses of streptozotocin (STZ). Body weight and blood glucose levels were measured weekly until 6 weeks. After 6 weeks, all mice were sacrificed and the levels of insulin (Ins), tumor necrosis factor α (TNF-α), interleukin 4 (IL-4), IL-6, and type I interferon γ (IFN-γ), IFNAR2 protein expression, the number of dendritic cells (DCs), and changes in islet ß cells were assessed. RESULTS: IFNAR2 mRNA and protein expression levels in serum of T1D patients were significantly higher than those in healthy controls (P < 0.05). Furthermore, IFNAR2 protein expression, number of DCs, and IFNAR2 mRNA, blood glucose, TNF-α, and IFN-γ levels were significantly upregulated in T1D mice compared with the control group (P < 0.05), while weight, and Ins, IL-6, and IL-4 levels were decreased (P < 0.05). However, knockdown of IFNAR2 reversed these trends. There was no significant difference in markers between the T1D + ad-con group and the T1D group (P > 0.05). CONCLUSIONS: Knockdown of IFNAR2 reduced the inflammatory response and improved islet function of T1D mice.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Insulin , Interleukin-4/genetics , Interleukin-6 , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha
7.
Gynecol Endocrinol ; 38(5): 416-424, 2022 May.
Article in English | MEDLINE | ID: mdl-35348414

ABSTRACT

BACKGROUND: Maternal high-fat diet (HFD) is a detrimental factor in developing glucose intolerance, obesity, and islet dysfunction. However, the effect of artemisinin on maternal HFD and whether it is related to the alterations of islet function is seldom studied since artemisinin treatments not only attenuate insulin resistance (IR) and restore islet ß cell function in Diabetes mellitus type 2. METHODS: Female rats were randomly fed a HFD (45% kcal from fat), HFD + artemisinin, or a regular chow diet (RCD) before pregnancy and during gestation. Glucose metabolism and the ß cell phenotypes were assessed. RESULTS: Maternal HFD increased islet load in female rats, proliferation of pancreatic ß cells, increased insulinogen, and decreased insulin secretion response to high glucose stimulation with delayed insulin release, increased fasting glucose, and glucose area under the curve compared with the general diet group. HFD inhibited expression of Foxo1 and PAX6 in female rats. Under the effect of both HFD and pregnancy, islet load was further increased, insulinogen was further increased, and fasting insulin level and fasting glucose were higher than RCD fed general-pregnancy group. ALDH1a3 transdifferentiation and PAX6, Foxo1, and PDX1 expression were increased in islets of high-fat pregnant rats. When adding artemisinin in HFD treated pregnant rats, islet function was significantly improved. CONCLUSIONS: Intervention with artemisinin in maternal HFD resulted in reduced islet size, decreased number of ß-cells and improved islet microcirculation, insulin processing shear process, decreased insulinogen/insulin ratio, and restored islet function through increased expression of PC1/3.


Subject(s)
Artemisinins , Insulin Resistance , Insulin-Secreting Cells , Animals , Artemisinins/metabolism , Artemisinins/pharmacology , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Female , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Pregnancy , Rats
8.
J Biol Chem ; 295(5): 1261-1270, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31882540

ABSTRACT

MicroRNA 199 (miR-199) negatively impacts pancreatic ß-cell function and its expression is highly increased in islets from diabetic mice as well as in plasma of diabetic patients. Here we investigated how miR-199 expression is regulated in ß-cells by assessing expression of miR-199 precursors (primiR-199a1, primiR-199a2, and primiR-199b) and mature miR-199 (miR-199-3p and miR-199-5p) and promoter transcriptional activity assays in mouse islets and mouse insulinoma cells (MIN6) under different stimuli. We found that mouse islets equally express miR-199-3p and miR-199-5p. However, the primiRNA expression levels differed; although primiR-199a1 expression was about 30% greater than that of primiR-199a2, primiR-199b is barely detected in islets. We observed a 2-fold increase in primiR-199a1 and primiR-199a2 mRNA levels in mouse islets cultured in 10 mm glucose compared with 5.5 mm glucose. Similar responses to glucose were observed in MIN6 cells. Exposure to 30 mm KCl to induce membrane depolarization and calcium influx increased expression of primiR-199a2 but not of primiR-199a1 in MIN6 cells, indicating that calcium influx was involved. Transcriptional activity studies in MIN6 cells also revealed that primiR-199a2 promoter activity was enhanced by glucose and reduced by 2-deoxy-D-glucose-induced starvation. KCl and the potassium channel blocker tolbutamide also stimulated primiR-199a2 promoter activity. Calcium channel blockade by nifedipine reduced primiR-199a2 promoter activity in MIN6 cells, and diazoxide-mediated calcium influx inhibition blunted glucose up-regulation of miR-199-3p in islets. In conclusion, we uncover that glucose acutely up-regulates miR-199 family expression in ß-cells. Glucose metabolism and calcium influx are involved in primiR-199a2 expression but not primiR-199a1 expression.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Female , Male , Mice , MicroRNAs/genetics , Up-Regulation
9.
Xenotransplantation ; 28(3): e12667, 2021 05.
Article in English | MEDLINE | ID: mdl-33438288

ABSTRACT

BACKGROUND: Necrostatin-1 (Nec-1) supplementation to tissue culture media on day 3 has recently been shown to augment the insulin content, endocrine cellular composition, and insulin release of pre-weaned porcine islets (PPIs); however, its effects were only examined for the first 7 days of tissue culture. The present study examined whether the addition of Nec-1 on day 3 could further enhance the in vitro development and function of PPIs after 14 days of tissue culture. METHODS: PPIs were isolated from 8- to 15-day-old, pre-weaned Yorkshire piglets and cultured in an islet maturation media supplemented with Nec-1 on day 3. The recovery, viability, insulin content, endocrine cellular composition, GLUT2 expression in beta cells, differentiation and proliferation potential, and glucose-stimulated insulin secretion of PPIs were assessed on days 3, 7, and 14 of tissue culture (n = 5 on each day). RESULTS: Compared with day 7 of tissue culture, islets on day 14 had a lower recovery, GLUT2 expression in beta cells, proliferation capacity of endocrine cells, and glucose-induced insulin stimulation index. Prolonging the culture time to 14 days did not affect islet viability, insulin content, proportion of endocrine cells, and differentiation potential. CONCLUSION: The growth-inducing effects of Nec-1 on PPIs were most effective on day 7 of tissue culture when added on day 3. Our findings support existing evidence that the in vitro activities of Nec-1 are short-lived and encourage future studies to explore the use of other novel growth factors during prolonged islet tissue culture.


Subject(s)
Islets of Langerhans , Animals , Imidazoles , Indoles , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Swine , Transplantation, Heterologous
10.
BMC Endocr Disord ; 21(1): 203, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34663294

ABSTRACT

BACKGROUND: The aim of the study was to explore the relationship between vitamin D status and islet function in patients with type 2 diabetes mellitus. METHODS: The participants were recruited from Hebei General Hospital. Basic characteristics and blood indicators were collected after fasting overnight. The data were analyzed statistically using SPSS 22.0. Analysis of variance, a nonparametric test, or a trend Chi-square test was used for the comparisons. The association between 25-hydroxy vitamin D and modified homeostasis model assessment-ß was assessed using multivariate ordinal logistic regression. RESULTS: One hundred seventy-four patients aged 26 to 79 years with type 2 diabetes mellitus were included in this study. Patients with vitamin D deficiency had a lower modified homeostasis model assessment-ß level compared with those without vitamin D deficiency. There were differences in body mass index, diabetes course, glycosylated hemoglobin, fasting blood glucose, fasting blood C-peptide, triglyceride, and 25-hydroxy vitamin D among different modified homeostasis model assessment-ß groups based upon the tertiles. 25-hydroxy vitamin D, as continuous or categorical variables, was positively related to modified homeostasis model assessment-ß whether or not cofounding factors were adjusted. CONCLUSION: There is an association between increased 25-hydroxy vitamin D levels and improvement in modified homeostasis model assessment-ß function in patients with type 2 diabetes mellitus. TRIAL REGISTRATION: Cross-sectional trails ChiCTR2000029391 , Registration Date: 29/01/2020.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans/physiopathology , Vitamin D/analogs & derivatives , Adult , Aged , Blood Glucose/analysis , Blood Glucose/metabolism , China/epidemiology , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Female , Glycated Hemoglobin/analysis , Humans , Insulin Resistance/physiology , Islets of Langerhans/pathology , Male , Middle Aged , Vitamin D/blood , Vitamin D Deficiency/blood , Vitamin D Deficiency/complications , Vitamin D Deficiency/epidemiology , Vitamin D Deficiency/physiopathology
11.
J Clin Biochem Nutr ; 69(3): 229-237, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34857984

ABSTRACT

Objective of the present study was to evaluate the effect of vitamin D supplementation on glycose homeostasis, islet function, and diabetes progress. Literatures were searched via electronic databases, websites, and previous reviews from the earliest available time to the end of May 2020. Randomized controlled trials initially designed for diabetes and prediabetes with 25-dihydroxyvitamin D [25(OH)D]<30 ng/ml were included. All data were analyzed and presented based on the Cochrane guidelines and PRISMA guidelines. In total, 27 articles (n = 1,932) were enrolled in this study. Vitamin D supplementation significantly improved fasting blood glucose, postprandial blood glucose, and quantitative insulin sensitivity check index in diabetes and prediabetes with baseline 25(OH)D<30 ng/ml. Higher percentages regressing from prediabetes to normal glucose status [1.60 (1.19, 2.17), p = 0.002, n = 564] and lower percentage progressing from prediabetes to diabetes [0.68 (0.36, 1.27), p = 0.23, n = 569] were found in the supplementation group. The positive effects of vitamin D supplementation on body mass index, waist, HDL-C, LDL-C, and CRP were also demonstrated. In conclusion, modest improvements in vitamin D supplementation on short-term glycose homeostasis, insulin sensitivity, and disease development in diabetes and prediabetes with 25(OH)D<30 ng/ml were demonstrated, but more research needs to be conducted in the future to support the clinical application. (Register ID: CRD42020186004).

12.
Biochem Biophys Res Commun ; 524(1): 205-210, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31983426

ABSTRACT

Recent studies highlighted the significance of chronic inflammation, which is mediated in part by toll-like receptors 4 (TLR4), in islet ß cell dysfunction by high-glucose exposure. However, about it is unclear whether islet ß cell dysfunction in response to high glucose is associated with TLR4. This investigation was designed to address the effect of TLR4 deficiency on insulin secretion in mice in response to acute intravenous glucose load. Hyperglycemic clamp was used to impair insulin secretion, and intraperitoneal glucose tolerance test was carried out to analyze insulin secretion function of islet ß cells. Our results showed that TLR4 deficiency repressed insulin secretion impairment in response to acute intravenous glucose load. Compared to wild-type mice, TLR4-/- mice did not exhibit increase of IL-1ß and TNF-α level in plasma and pancreatic tissue in response to acute intravenous load of high glucose. However, recombinant IL-1ß or TNF-α administration restored insulin secretion impairment induced by high glucose in TLR4-/- mice. Taken together, our results demonstrated that TLR4 activation and subsequent IL-1ß and TNF-α production contribute to islet ß cell dysfunction in mice in response to acute intravenous load of high glucose, which may provide a theoretical basis for diabetes complication improvement by physical exercise.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , Toll-Like Receptor 4/metabolism , Administration, Intravenous , Animals , Cytokines/administration & dosage , Cytokines/blood , Glucose/administration & dosage , Insulin Secretion , Male , Mice, Inbred C57BL , Toll-Like Receptor 4/deficiency
13.
Cell Biochem Funct ; 38(8): 1129-1138, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32869325

ABSTRACT

Taurine upregulated gene 1 (Tug1) is a novel lncRNA that participates in growth, and the abnormal expression of Tug1 related to mouse islet cell dysfunction. A recent study revealed that intrauterine growth retardation (IUGR) related to the pathogenesis of diabetes. Here, we aimed to explore the role and mechanism of Tug1 in IUGR-mediated islet dysfunction. We observed that newborn IUGR mice had lower body and pancreas weight and smaller islets than newborn control mice. After IUGR mice were given a normal diet, they showed catch-up growth and abnormal glucose tolerance; however, the pancreas/body weight ratio remained low. Blood glucose, serum insulin and related gene expression showed mild recovery after overexpression of Tug1 in IUGR mice. Furthermore, Tug1 was enriched in the nuclei of MIN6 cells. Using RIP and CHIP analyses we found that Tug1 could regulate Hes1 expression by binding to EZH2 to affect insulin synthesis in MIN6 cells. These findings indicate that lncRNA Tug1 could regulate the expression of Hes1 via EZH2-driven H3K27 methylation and affect insulin production. SIGNIFICANCE OF THE STUDY: This study suggests Tug1 as a novel biomarker, as it was shown to regulate ß cell function and is worthy of further investigation due to its potential for diabetes treatment.


Subject(s)
Fetal Growth Retardation , Islets of Langerhans , RNA, Long Noncoding/metabolism , Animals , Biomarkers/metabolism , Cell Line , Disease Models, Animal , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Islets of Langerhans/embryology , Islets of Langerhans/pathology , Male , Mice
14.
J Cell Biochem ; 120(12): 19338-19344, 2019 12.
Article in English | MEDLINE | ID: mdl-31535398

ABSTRACT

OBJECTIVE: To explore the role and molecular mechanism of regulatory T (Treg) cells in type 1 diabetes (T1D). METHODS: Patients with T1D and the healthy volunteers were selected and a number of CD3+ , CD4+ , CD25+ , and CD127- T cells were determined. The rats were divided into the control, T1D model, and Treg infusion (T1D rats were infused with Treg) group. The number of CD4+ , CD8- , and CD25+ T cells in the three groups were determined by flow cytometry. Weight, blood glucose, serum insulin, peptide C, glucagon, and glucagon like peptide 1 in the three groups were also determined. The messenger RNA (mRNA) levels and contents of interleukin (IL)-10, IL-4, transforming growth factor (TGF)-ß, IL-2, IL-17, and IFN-γ in patients with T1D, healthy volunteers, streptozotocin (STZ)-induced T1D rat model, the control rat, and Treg infusion rats were determined by reverse transcription polymerase chain reaction and the enzyme-linked immunosorbent assay, respectively. RESULTS: Treg content in patients with T1D was significantly decreased compared with the control volunteers. Treg content in rats was markedly decreased after injection with STZ to induce T1D rat model, while Treg infusion weakened the decrease. The change scope of weight and blood glucose in the model and Treg group was bigger than the control group, and the change in the infusion group was lighter than the model group. T1D decreased the expressions of IL-10, IL-4, TGF-ß, and IL-2, while Treg infusion weakened the decrease. The expressions of IL-17 and IFN-γ in the T1D group was increased, while Treg infusion weakened the increase. CONCLUSION: Autologous Treg infusion can strengthen the immunologic and islet function to treat T1D which may be via regulating the expression of inflammatory factors.


Subject(s)
Cytokines/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/genetics , Interleukin-10/genetics , Interleukin-2/genetics , Interleukin-4/genetics , Male , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory , Transforming Growth Factor beta/genetics
15.
J Cell Biochem ; 119(11): 9239-9248, 2018 11.
Article in English | MEDLINE | ID: mdl-29953637

ABSTRACT

Increasing evidence indicates that long noncoding RNAs (lncRNAs) perform special biological functions by regulating gene expression through multiple pathways and molecular mechanisms. The aim of this study was to explore the expression characteristics of lncRNA uc.322 in pancreatic islet cells and its effects on the secretion function of islet cells. Bioinformatics analysis was used to detect the lncRNA uc.322 sequence, location, and structural features. Expression of lncRNA uc.322 in different tissues was detected by quantitative polymerase chain reaction analyses. Quantitative polymerase chain reaction, Western blot analysis, adenosine triphosphate determination, glucose-stimulated insulin secretion, and enzyme-linked immunosorbent assay were used to evaluate the effects of lncRNA uc.322 on insulin secretion. The results showed that the full-length of lncRNA uc.322 is 224 bp and that it is highly conserved in various species. Bioinformatics analysis revealed that lncRNA uc.322 is located on chr7:122893196-122893419 (GRCH37/hg19) within the SRY-related HMG-box 6 gene exon region. Compared with other tissues, lncRNA uc.322 is highly expressed in pancreatic tissue. Upregulation of lncRNA uc.322 expression increases the insulin transcription factors pancreatic and duodenal homeobox 1 and Forkhead box O1 expression, promotes insulin secretion in the extracellular fluid of Min6 cells, and increases the adenosine triphosphate concentration. On the other hand, knockdown of lncRNA uc.322 has opposite effects on Min6 cells. Overall, this study showed that upregulation of lncRNA uc.322 in islet ß-cells can increase the expression of insulin transcription factors and promote insulin secretion, and it may be a new therapeutic target for diabetes.


Subject(s)
Gene Expression Regulation , Islets of Langerhans/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Adenosine Triphosphate/metabolism , Animals , Arginine/pharmacology , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Glucose/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Potassium Chloride/pharmacology
16.
FASEB J ; 31(12): 5342-5355, 2017 12.
Article in English | MEDLINE | ID: mdl-28794173

ABSTRACT

Impaired pancreatic ß-cell function is the primary defect in type 2 diabetes. Glucose is an important regulator of ß-cell growth and function; however, the mechanisms that are involved in the chronic adaptation of ß cells to hyperglycemia remain largely unknown. In the present study, global gene expression patterns revealed that tryptophan hydroxylase 1 (Tph1) was the most profound of genes that are up-regulated in rat islets exposed to high glucose. Calcium and cAMP signals synergistically mediated glucose-stimulated Tph1 transcription in ß cells by activating cAMP-responsive element-binding protein and promoting its binding with a Tph1 promoter. Similar to in vitro results, in vivo infusion of high glucose also strongly induced Tph1 expression and serotonin production in rat islets, along with enhanced islet function. Inhibition or knockdown of Tph1 markedly decreased glucose-potentiated insulin secretion. In contrast, overexpression of Tph1 augmented glucose-stimulated insulin secretion in rat islets by up-regulating the expression of genes that are related to islet function. In addition, the long-acting glucagon-like peptide 1 receptor agonist, exendin-4, stimulated Tph1 expression in a glucose-dependent manner. Knockdown of Tph1 inhibited exendin-4-potentiated insulin secretion in rat islets. These findings suggest that Tph1 mediates the compensation of islet function induced by glucose, and that promoting Tph1 expression in pancreatic ß cells will provide a new strategy for the treatment of type 2 diabetes mellitus.-Zhang, Y., Deng, R., Yang, X., Xu, W., Liu, Y., Li, F., Zhang, J., Tang, H., Ji, X., Bi, Y., Wang, X., Zhou, L., Ning, G. Glucose potentiates ß-cell function by inducing Tph1 expression in rat islets.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Tryptophan Hydroxylase/metabolism , Animals , Calcium Signaling/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Cyclic AMP/metabolism , Glucose/pharmacology , Glucose Tolerance Test , Immunoblotting , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Tryptophan Hydroxylase/genetics
17.
J Sci Food Agric ; 98(6): 2350-2359, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28991371

ABSTRACT

BACKGROUND: The aim of this study was to investigate the antidiabetic effects of the crude polyphenol extract (BSCP) from black soybean seed coats (BSC) and the whole flour of BSC and illustrate the mechanism in terms of islet and renal protection. RESULTS: BSCP and BSC effectively controlled the increased blood glucose level and impaired glucose tolerance in streptozotocin (STZ)-induced diabetic rats after 8 weeks of treatment. They increased the concentrations of serum insulin, C-peptide and Glp-1 (P < 0.05) by improving the STZ-induced damage of islet ß-cells and increasing their insulin expression (P < 0.05). Lipid profiles and antioxidant activities were also improved. Moreover, BSCP and BSC tended to decrease serum creatinine (0.05 < P < 0.1), and blood urea nitrogen was decreased by BSC significantly (P < 0.05). They also led to significantly lower glomerular volume (P < 0.05). CONCLUSION: Long-term intervention with BSC at a low dose of polyphenols plays a role in controlling blood glucose and lipids levels by promoting insulin secretion and restoring islet ß-cell function, the same as BSCP. These benefits are accompanied by their potential protection of diabetic renal dysfunction. BSCP is mainly responsible for the antidiabetic effect of BSC. © 2017 Society of Chemical Industry.


Subject(s)
Diabetes Mellitus, Type 2/diet therapy , Glycine max/metabolism , Hypoglycemic Agents/metabolism , Kidney/physiopathology , Plant Extracts/metabolism , Polyphenols/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Male , Oxidative Stress , Phytotherapy , Rats , Rats, Sprague-Dawley , Rats, Wistar , Seeds/chemistry , Glycine max/chemistry , Streptozocin/adverse effects
18.
Am J Physiol Endocrinol Metab ; 311(3): E564-74, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27460898

ABSTRACT

During pregnancy, maternal ß-cells undergo compensatory changes, including increased ß-cell mass and enhanced glucose-stimulated insulin secretion. Failure of these adaptations to occur results in gestational diabetes mellitus. The secreted protein connective tissue growth factor (CTGF) is critical for normal ß-cell development and promotes regeneration after partial ß-cell ablation. During embryogenesis, CTGF is expressed in pancreatic ducts, vasculature, and ß-cells. In adult pancreas, CTGF is expressed only in the vasculature. Here we show that pregnant mice with global Ctgf haploinsufficiency (Ctgf(LacZ/+)) have an impairment in maternal ß-cell proliferation; no difference was observed in virgin Ctgf(LacZ/+) females. Using a conditional CTGF allele, we found that mice with a specific inactivation of CTGF in endocrine cells (Ctgf(ΔEndo)) develop gestational diabetes during pregnancy, but this is due to a reduction in glucose-stimulated insulin secretion rather than impaired maternal ß-cell proliferation. Moreover, virgin Ctgf(ΔEndo) females also display impaired GSIS with glucose intolerance, indicating that underlying ß-cell dysfunction precedes the development of gestational diabetes in this animal model. This is the first time a role for CTGF in ß-cell function has been reported.


Subject(s)
Cell Size , Connective Tissue Growth Factor/metabolism , Diabetes, Gestational/physiopathology , Insulin-Secreting Cells/metabolism , Aging , Alleles , Animals , Connective Tissue Growth Factor/deficiency , Connective Tissue Growth Factor/genetics , Diabetes, Gestational/metabolism , Disease Models, Animal , Embryonic Development , Endocrine Cells/metabolism , Endocrine Cells/physiology , Female , Glucose/pharmacology , Glucose Intolerance/metabolism , Glucose Tolerance Test , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/blood supply , Mice , Mice, Knockout , Pregnancy
19.
Biochem Biophys Res Commun ; 481(1-2): 84-89, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27823935

ABSTRACT

Cocaine- and amphetamine-regulated transcript (CART) is an anorexigenic peptide widely expressed in the central and peripheral nervous systems, as well as in endocrine cells. CART is markedly upregulated in the ß-cells of several rodent models of type-2 diabetes. The stimulatory effect of exogenous CART peptide on insulin secretion is cAMP dependent. Glucose is the most important regulator of islet function. However, the role of CART in glucose-potentiated insulin secretion remains unclear. Here, our results showed that glucose time- and dose-dependently elicited CART mRNA expression in rat islets. Both the glucokinase agonist GKA50 and the long-acting GLP-1 analogue exendin-4 increased CART mRNA expression. The protein kinase A (PKA) inhibitor H89 and the inactivation of cAMP response element-binding protein (CREB) suppressed forskolin-stimulated CART mRNA expression. Furthermore, CART overexpression amplified insulin secretion from rat islets in response to glucose and forskolin, and ameliorated dexamethasone-impaired insulin secretion. These findings suggest that islet-derived CART is involved, at least in part, in high glucose-potentiated pancreatic ß-cell function.


Subject(s)
Gene Expression Regulation/physiology , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Nerve Tissue Proteins/biosynthesis , Animals , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glucose/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Male , Rats , Rats, Sprague-Dawley
20.
Am J Physiol Regul Integr Comp Physiol ; 310(2): R143-55, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26561648

ABSTRACT

Neuronostatin (NST) is a recently described peptide that is produced from the somatostatin preprohormone in pancreatic δ-cells. NST has been shown to increase glucagon secretion from primary rat pancreatic islets in low-glucose conditions. Here, we demonstrate that NST increases proglucagon message in α-cells and identify a potential mechanism for NST's cellular activities, including the phosphorylation of PKA following activation of the G protein-coupled receptor, GPR107. GPR107 is abundantly expressed in the pancreas, particularly, in rodent and human α-cells. Compromise of GPR107 in pancreatic α-cells results in failure of NST to increase PKA phosphorylation and proglucagon mRNA levels. We also demonstrate colocalization of GPR107 and NST on both mouse and human pancreatic α-cells. Taken together with our group's observation that NST infusion in conscious rats impairs glucose clearance in response to a glucose challenge and that plasma levels of the peptide are elevated in the fasted compared with the fed or fasted-refed state, these studies support the hypothesis that endogenous NST regulates islet cell function by interacting with GPR107 and initiating signaling in glucagon-producing α-cells.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Glucagon-Secreting Cells/drug effects , Peptide Hormones/pharmacology , Proglucagon/genetics , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line , Glucagon-Secreting Cells/enzymology , Humans , Male , Mice , Peptide Fragments/metabolism , Peptide Hormones/metabolism , Phosphorylation , RNA Interference , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Somatostatin/metabolism , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL