460 results
Search Results
2. Regional variation of human pancreatic islets dimension and its impact on beta cells in Indian population
- Author
-
Pravash Ranjan Mishra, Suvendu Purkait, Usha Agrawal, Shree Ram Singh, Susama Patra, Madhumita Patnaik, and Praveen Kumar Ravi
- Subjects
Adult ,Male ,0301 basic medicine ,endocrine system ,medicine.medical_specialty ,endocrine system diseases ,Insulin Antibodies ,Endocrinology, Diabetes and Metabolism ,Population ,India ,030209 endocrinology & metabolism ,Biology ,Computing Methodologies ,Islets of Langerhans ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Insulin resistance ,Insulin-Secreting Cells ,Internal medicine ,Diabetes mellitus ,medicine ,Humans ,Beta (finance) ,education ,Pancreas ,geography ,education.field_of_study ,geography.geographical_feature_category ,Pancreatic islets ,Anatomy, Regional ,Islet ,medicine.disease ,Immunohistochemistry ,030104 developmental biology ,medicine.anatomical_structure ,Biological Variation, Population ,Female ,Autopsy ,Beta cell ,Research Paper - Abstract
Background & objectives: Islet of Langerhans, the endocrine pancreas plays a significant role in glucose metabolism. Obesity and insulin resistance are the major factors responsible for beta cell dysfunction. Asian Indian population has increased susceptibility to diabetes in spite of having lower BMI. The morphology of islets plays a significant role in beta cell function. The present study was designed for better understanding the morphology, composition and distribution of islets in different parts of the pancreas and its impact on beta cell proportion. Methods: We observed islet morphology and beta cell area proportion by Large-scale computer-assisted analysis in 20 adult human pancreases in non-diabetic Indian population. Immunohistochemical staining with anti-synaptophysin and anti-insulin antibody was used to detect islet and beta cells respectively. Whole slide images were analyzed using ImageJ software. Results: Endocrine proportion were heterogeneously increasing from head to tail with maximum islet and beta cell distribution in the tail region. Larger islets were predominately confined to the tail region. The islets in Indian population were relatively smaller in size, but they have more beta cells (20%) when compared to American population. Interpretation & conclusions: The beta cells of larger islets are functionally more active than the smaller islets via paracrine effect. Thus, reduction in the number of larger islets may be one of the probable reasons for increased susceptibility of Indians to diabetes even at lower BMI. Knowledge about the regional distribution of islets will help the surgeons to preserve the islet rich regions during surgery.
- Published
- 2019
3. Glyoxalase 1 Prevents Chronic Hyperglycemia Induced Heart-Explant Derived Cell Dysfunction
- Author
-
Sandrine Parent, Mary-Ellen Harper, Ghazaleh Rafatian, Pushpinder Kanda, Connor Michie, Melanie Villanueva, Darryl R. Davis, Bin Ye, Georges N. Kanaan, and Wenbin Liang
- Subjects
0301 basic medicine ,Heart disease ,Cell- and Tissue-Based Therapy ,Myocardial Infarction ,Medicine (miscellaneous) ,Mice, Transgenic ,030204 cardiovascular system & hematology ,Pharmacology ,medicine.disease_cause ,Antioxidants ,Cell therapy ,Diabetes Complications ,03 medical and health sciences ,Extracellular Vesicles ,Mice ,0302 clinical medicine ,reactive dicarbonyls ,Medicine ,Animals ,Humans ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Heart Failure ,diabetes ,business.industry ,cardiac stem cells ,Lactoylglutathione Lyase ,medicine.disease ,Streptozotocin ,3. Good health ,Mice, Inbred C57BL ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Heart failure ,Hyperglycemia ,Chronic Disease ,Beta cell ,business ,Reactive Oxygen Species ,Oxidative stress ,medicine.drug ,Blood vessel ,Research Paper - Abstract
Decades of work have shown that diabetes increases the risk of heart disease and worsens clinical outcomes after myocardial infarction. Because diabetes is an absolute contraindication to heart transplant, cell therapy is increasingly being explored as a means of improving heart function for these patients with very few other options. Given that hyperglycemia promotes the generation of toxic metabolites, the influence of the key detoxification enzyme glyoxalase 1 (Glo1) on chronic hyperglycemia induced heart explant-derived cell (EDC) dysfunction was investigated. Methods: EDCs were cultured from wild type C57Bl/6 or Glo1 over-expressing transgenic mice 2 months after treatment with the pancreatic beta cell toxin streptozotocin or vehicle. The effects of Glo1 overexpression was evaluated using in vitro and in vivo models of myocardial ischemia. Results: Chronic hyperglycemia reduced overall culture yields and increased the reactive dicarbonyl cell burden within EDCs. These intrinsic cell changes reduced the angiogenic potential and production of pro-healing exosomes while promoting senescence and slowing proliferation. Compared to intra-myocardial injection of normoglycemic cells, chronic hyperglycemia attenuated cell-mediated improvements in myocardial function and reduced the ability of transplanted cells to promote new blood vessel and cardiomyocyte growth. In contrast, Glo1 overexpression decreased oxidative damage while restoring both cell culture yields and EDC-mediated repair of ischemic myocardium. The latter was associated with enhanced production of pro-healing extracellular vesicles by Glo1 cells without altering the pro-healing microRNA cargo within. Conclusions: Chronic hyperglycemia decreases the regenerative performance of EDCs. Overexpression of Glo1 reduces dicarbonyl stress and prevents chronic hyperglycemia-induced dysfunction by rejuvenating the production of pro-healing extracellular vesicles.
- Published
- 2019
4. A functional genomic approach to identify reference genes for human pancreatic beta cell real-time quantitative RT-PCR analysis
- Author
-
Miriam Cnop, Ioannis Gkantounas, Piero Marchetti, Ângela Castela, Sandra Marín-Cañas, Florian Szymczak, Mariana Igoillo-Esteve, Decio L. Eizirik, Maikel Luis Colli, Maria Inês Alvelos, Federica Fantuzzi, Lorella Marselli, Cristina Cosentino, and Bianca Marmontel de Souza
- Subjects
Endocrinology, Diabetes and Metabolism ,Coefficient of variation ,medicine.medical_treatment ,Type 2 diabetes ,Biology ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Endocrinology ,Text mining ,0302 clinical medicine ,Rt pcr analysis ,Insulin-Secreting Cells ,Pancreatic beta Cells ,Reference genes ,Gene expression ,medicine ,Humans ,Beta (finance) ,Gene ,030304 developmental biology ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Genomics ,medicine.disease ,Molecular biology ,Cytokine ,RAB7A ,030220 oncology & carcinogenesis ,Reference genes/ beta cells/ diabetes/ RNA-sequencing/ quantitative real-time pcr ,Beta cell ,business ,030217 neurology & neurosurgery ,Research Article ,Research Paper - Abstract
Exposure of human pancreatic beta cells to pro-inflammatory cytokines or metabolic stressors is used to model events related to type 1 and type 2 diabetes, respectively. Quantitative real-time PCR is commonly used to quantify changes in gene expression. The selection of the most adequate reference gene(s) for gene expression normalization is an important pre-requisite to obtain accurate and reliable results. There are no universally applicable reference genes, and the human beta cell expression of commonly used reference genes can be altered by different stressors. Here we aimed to identify the most stably expressed genes in human beta cells to normalize quantitative real-time PCR gene expression.We used comprehensive RNA-sequencing data from the human pancreatic beta cell line EndoC-βH1, human islets exposed to cytokines or the free fatty acid palmitate in order to identify the most stably expressed genes. Genes were filtered based on their level of significance (adjusted P-value >0.05), fold-change (|fold-change| We identified a total of 264 genes stably expressed in EndoC-βH1 cells and human islets following cytokine- or palmitate-induced stress, displaying a low coefficient of variation. Validation by quantitative real-time PCR of the top five genes ARF1, CWC15, RAB7A, SIAH1 and VAPA corroborated their expression stability under most of the tested conditions. Further validation in independent samples indicated that the geometric mean of ACTB and VAPA expression can be used as a reliable normalizing factor in human beta cells.
- Published
- 2021
- Full Text
- View/download PDF
5. Characterization of a mouse model of islet transplantation using MIN-6 cells
- Author
-
Douglas O. Sobel, Larry Mitnaul, and Barath Ramasubramanian
- Subjects
0301 basic medicine ,Blood Glucose ,Endocrinology, Diabetes and Metabolism ,Islets of Langerhans Transplantation ,Mice, Nude ,030209 endocrinology & metabolism ,Mice, Transgenic ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Islets of Langerhans ,Mice ,0302 clinical medicine ,Endocrinology ,In vivo ,Mice, Inbred NOD ,Diabetes mellitus ,medicine ,Animals ,Insulin ,Beta (finance) ,Cell Line, Transformed ,geography ,Mice, Inbred BALB C ,Mice, Inbred C3H ,geography.geographical_feature_category ,Chemistry ,Glucose Tolerance Test ,medicine.disease ,Islet ,Molecular biology ,In vitro ,Transplantation ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Transformed cell ,Female ,Beta cell ,Research Paper - Abstract
Immortalized beta cells are an abundant source of insulin-producing cells. Although MIN-6 cells have similar characteristics as normal islets in vitro, the in vivo use of MIN-6 cells has not been fully described. This study characterizes in vivo mouse models of MIN-6 transplantation and rejection. Subcutaneous (sc) transplantation of MIN-6 cells in either Matrigel or HyStem-C hydrogels reduced blood sugars in nude mice and thus are good matrices for MIN-6 cells in vivo. NOD mice are good transplant recipients since they best rejected MIN-6 cells. MLR responses from BalbC, Black Webster, Swiss Black, C3H, and NOD mice correlated with mean blood glucose response suggesting the importance of allogeneic differences in the rejection of cells. Three days of cyclosporine administration caused no inhibition of MIN-6 cell rejection and 6 days resulted in a transient decrease in blood glucose, while daily administration inhibited rejection long term. Kinetic glucose tolerance (GTT) studies in nude mice demonstrated transplanted MIN-6 cells are close but not as effective as normal islets in controlling blood glucose and blood glucose set point for insulin release in MIN-6 cells decreases to hypoglycemic levels over time. To avoid hypoglycemia, the effect of MIN-6 cell irradiation was assessed. However, irradiation only delayed the development of hypoglycemia, not altering the final glucose set point for insulin release. In conclusion, we have characterized a mouse model for beta-cell transplantation using subcutaneous MIN-6 cells that can be used as a tool to study approaches to mitigate immune rejection.
- Published
- 2020
6. Inhibition of heparanase protects against pancreatic beta cell death in streptozotocin-induced diabetic mice via reducing intra-islet inflammatory cell infiltration
- Author
-
Chen-Yu Zhang, Kai Li, Ying Ding, Ming-Xuan Zhou, Xiao Han, Yun Xia, Yan Wang, Xiao-Han Jiang, Wen-Yu Song, Chen Qiu, Peng Sun, and Chong-Chong Yin
- Subjects
0301 basic medicine ,medicine.medical_specialty ,endocrine system ,endocrine system diseases ,Streptozocin ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,chemistry.chemical_compound ,Islets of Langerhans ,Mice ,0302 clinical medicine ,Internal medicine ,Insulin-Secreting Cells ,medicine ,Animals ,Insulin ,Pancreatic islet function ,Heparanase ,Insulinoma ,Glucuronidase ,Pharmacology ,geography ,geography.geographical_feature_category ,Chemistry ,Heparan sulfate ,medicine.disease ,Streptozotocin ,Islet ,Research Papers ,030104 developmental biology ,Endocrinology ,Apoptosis ,Beta cell ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background and purpose Intra-islet heparan sulfate (HS) plays an important role in the maintenance of pancreatic islet function. The aim of this study was to investigate the effect mechanism of HS loss on the functioning of islets in diabetic mice. Experimental approach The hypoglycaemic effect of a heparanase inhibitor, OGT2115, was tested in a streptozotocin-induced diabetic mouse model. The islets in pancreatic sections were also stained to reveal their morphology. An insulinoma cell line (MIN6) and primary isolated murine islets were used to investigate the effect of OGT2115 in vitro. Key results Intra-islet HS was clearly lost in streptozotocin-induced diabetic mice due to the increased heparanase expression in damaged islets. OGT2115 prevented intra-islet HS loss and improved the glucose profile and insulin secretion in streptozotocin-treated mice. The apoptosis of pancreatic beta cells and the infiltration of mononuclear macrophages, CD4- and CD8-positive T-cells in islets was reduced by OGT2115 in streptozotocin-treated mice, but OGT2115 did not alter the direct streptozotocin-induced damage in vitro. The expression of heparanase was increased in high glucose-treated isolated islets but not in response to direct streptozotocin stimulation. Further experiments showed that high glucose stimuli could decreased expression of PPARγ in cultured islets, thereby relieving the PPARγ-induced inhibition of heparanase gene expression. Conclusion and implications Hyperglycaemia could cause intra-islet HS loss by elevating the expression of heparanase, thereby aggravating inflammatory cell infiltration and islet damage. Inhibition of heparanase might provide benefit for pancreatic beta cell protection in Type 1 diabetes.
- Published
- 2020
7. Insulin acts as a repressive factor to inhibit the ability of PAR2 to induce islet cell transdifferentiation
- Author
-
Ergeng Hao, David Scharp, Fred Levine, and Seung-Hee Lee
- Subjects
0301 basic medicine ,geography ,geography.geographical_feature_category ,biology ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Transdifferentiation ,biology.organism_classification ,Islet ,Neogenesis ,Alpha cell ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,030220 oncology & carcinogenesis ,Cell Transdifferentiation ,medicine ,PAX4 ,Beta cell ,Research Paper - Abstract
Recently, we showed that pancreatitis in the context of profound β-cell deficiency was sufficient to induce islet cell transdifferentiation. In some circumstances, this effect was sufficient to result in recovery from severe diabetes. More recently, we showed that the molecular mechanism by which pancreatitis induced β-cell neogenesis by transdifferentiation was activation of an atypical GPCR called Protease-Activated Receptor 2 (PAR2). However, the ability of PAR2 to induce transdifferentiation occurred only in the setting of profound β-cell deficiency, implying the existence of a repressive factor from those cells. Here we show that the repressor from β-cells is insulin. Treatment of primary islets with a PAR2 agonist (2fLI) in combination with inhibitors of insulin secretion and signaling was sufficient to induce insulin and PAX4 gene expression. Moreover, in primary human islets, this treatment also led to the induction of bihormonal islet cells coexpressing glucagon and insulin, a hallmark of islet cell transdifferentiation. Mechanistically, insulin inhibited the positive effect of a PAR2 agonist on insulin gene expression and also led to an increase in PAX4, which plays an important role in islet cell transdifferentiation. The studies presented here demonstrate that insulin represses transdifferentiation of α- to β-cells induced by activation of PAR2. This provides a mechanistic explanation for the observation that α- to β-cell transdifferentiation occurs only in the setting of severe β-cell ablation. The mechanistic understanding of islet cell transdifferentiation and the ability to modulate that process using available pharmacological reagents represents an important step along the path towards harnessing this novel mechanism of β-cell neogenesis as a therapy for diabetes.
- Published
- 2018
8. Inhibition of Miro1 disturbs mitophagy and pancreatic β-cell function interfering insulin release via IRS-Akt-Foxo1 in diabetes
- Author
-
Yibin Yang, Jianfeng Gao, Yongchang Wei, Xiuli Men, Lingling Chen, Sijun Yang, Fuling Zhou, Lawrence W. C. Chan, Zhiwen Xie, Damien J. Keating, Jiazhong Sun, and Chunyan Liu
- Subjects
0301 basic medicine ,Gerontology ,medicine.medical_specialty ,medicine.medical_treatment ,FOXO1 ,Type 2 diabetes ,03 medical and health sciences ,Insulin resistance ,Internal medicine ,Diabetes mellitus ,insulin resistance ,Mitophagy ,Pathology Section ,Medicine ,Protein kinase B ,diabetes ,business.industry ,Insulin ,medicine.disease ,Research Paper: Pathology ,030104 developmental biology ,Endocrinology ,mitophagy ,Oncology ,Miro1 ,HFD ,Beta cell ,business - Abstract
// Lingling Chen 1,2,* , Chunyan Liu 1,* , Jianfeng Gao 1 , Zhiwen Xie 3 , Lawrence W.C. Chan 4 , Damien J. Keating 5 , Yibin Yang 6 , Jiazhong Sun 7 , Fuling Zhou 8 , Yongchang Wei 9 , Xiuli Men 10 and Sijun Yang 1 1 ABSL-3 Laboratory at the Center for Animal Experiment and Institute of Animal Model for Human Disease, Wuhan University School of Medicine, Wuhan, P. R. China 2 Department of Cell Biology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, P.R. China 3 School of Bioscience and Technology , Weifang Medical University, Weifang Shandong, P.R. China 4 Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong, Hong Kong 5 Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia 6 Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China 7 Department of Respiratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China 8 Department of Hematology and Radiation, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China 9 Department of Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China 10 School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, P.R. China * These authors have contributed equally to this work Correspondence to: Sijun Yang, email: // Keywords : Miro1, mitophagy, insulin resistance, HFD, diabetes, Pathology Section Received : June 26, 2017 Accepted : August 29, 2017 Published : September 16, 2017 Abstract Mitochondrial function is essential to meet metabolic demand of pancreatic beta cells respond to high nutrient stress. Mitophagy is an essential component to normal pancreatic β-cell function and has been associated with β-cell failure in Type 2 diabetes (T2D). Our previous studies have indicated that mitochondrial Rho (Miro) GTPase-mediated mitochondrial dysfunction under high nutrient stress leads to NOD-like receptor 3 (NLRP3)-dependent proinflammatory responses and subsequent insulin resistance. However, the in vivo mechanism by which Miro1 underlies mitophagy has not been identified. Here we show firstly that the expression of Miro is reduced in human T2D and mouse db/db islets and in INS-1 cell line exposed to high glucose and palmitate. β-cell specific ablation of Miro1 (Miro1f/f: Rip-cre mice, or (IKO) under high nutrient stress promotes the development of hyperglycemia. β-cells from IKO mice display an inhibition of mitophagy under oxidative stress and induces mitochondrial dysfunction. Dysfunctional mitophagy in IKO mice is represented by damaged islet beta cell mitochondrial and secretory capacity, unbalanced downstream MKK-JNK signalling without affecting the levels of MEK, ERK or p38 activation and subsequently, impaired insulin secretion signaling via inhibition IRS-AKT-Foxo1 pathway, leading to worsening glucose tolerance in these mice. Thus, these data suggest that Miro1 may be responsible for mitophagy deficiency and β-cell dysfunction in T2D and that strategies target Miro1 in vivo may provide a therapeutic target to enhance β-cell mitochondrial quality and insulin secretion to ameliorate complications associated with T2D.
- Published
- 2017
9. Upregulation of UCP2 in beta-cells confers partial protection against both oxidative stress and glucotoxicity
- Author
-
Pierre Maechler, Melis Karaca, and Ning Li
- Subjects
0301 basic medicine ,UCP2 ,medicine.medical_specialty ,Programmed cell death ,Clinical Biochemistry ,030209 endocrinology & metabolism ,Context (language use) ,Type 2 diabetes ,Biology ,medicine.disease_cause ,Biochemistry ,Pancreatic islet ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adenosine Triphosphate ,Downregulation and upregulation ,Internal medicine ,Insulin-Secreting Cells ,medicine ,Uncoupling protein ,Animals ,Uncoupling Protein 2 ,ddc:612 ,lcsh:QH301-705.5 ,Glucotoxicity ,Cells, Cultured ,Membrane Potential, Mitochondrial ,lcsh:R5-920 ,Cell Death ,Superoxide ,Insulin secretion ,Organic Chemistry ,Beta-cell ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Glucose ,lcsh:Biology (General) ,chemistry ,Beta cell ,lcsh:Medicine (General) ,Oxidative stress ,Research Paper - Abstract
Deterioration of pancreatic beta-cells plays a critical role in the development of type 2 diabetes. Among the various stressors contributing to these deleterious effects, glucotoxicity and superoxides have been proposed as major players. In this context, the mitochondrial uncoupling protein UCP2 is regularly associated with the stress response. In the present study, we tested the effects of UCP2 upregulation in mouse islets with beta-cell specific overexpression of UCP2 (RIP-UCP2). Islets were subjected to both chronic glucotoxicity (7 days at 30 mM glucose) and acute oxidative stress (200 µM H2O2 for 10 min). Increased UCP2 expression did not alter mitochondrial potential and ATP generation but protected against glucotoxic effects. Glucose-stimulated insulin secretion was altered by both glucotoxicity and oxidative stress, in particular through higher basal insulin release at non-stimulatory glucose concentrations. The secretory response to glucose stimulation was partially preserved in beta-cells overexpressing UCP2. The higher rate of cell death induced by chronic high glucose exposure was lower in RIP-UCP2 islets. Finally, superoxide production was reduced by high glucose, both under acute and chronic conditions, and not modified by UCP2 overexpression. In conclusion, upregulation of UCP2 conferred protective effects to the stressed beta-cell through mechanisms not directly associated with superoxide production., Graphical abstract fx1, Highlights • UCP2 upregulation protects pancreatic ß-cells against glucotoxicity. • High glucose reduces superoxide production in pancreatic islets. • UCP2 upregulation does not change superoxide production. • UCP2 upregulation protects ß-cells against oxidative stress.
- Published
- 2017
10. IER3IP1 deficiency leads to increased β-cell death and decreased β-cell proliferation
- Author
-
Decheng Ren and Juan Sun
- Subjects
0301 basic medicine ,Gene knockdown ,Programmed cell death ,endocrine system ,cell death and proliferation ,Cell growth ,Kinase ,beta-cell ,Biology ,Permanent neonatal diabetes mellitus ,medicine.disease ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Oncology ,chemistry ,medicine ,Unfolded protein response ,IER3IP1 ,Inositol ,Beta cell ,Research Paper - Abstract
Mutations in the gene for Immediate Early Response 3 Interacting Protein 1 (IER3IP1) cause permanent neonatal diabetes mellitus in human. The mechanisms involved have not been determined and the role of IER3IP1 in β-cell survival has not been characterized. In order to determine if there is a molecular link between IER3IP1 deficiency and β-cell survival and proliferation, we knocked down Ier3ip1 gene expression in mouse MIN6 insulinoma cells. IER3IP1 suppression induced apoptotic cell death which was associated with an increase in Bim and a decrease in Bcl-xL. Knockdown of Bim reduced apoptotic cell death in MIN6 cells induced by IER3IP1 suppression. Overexpression of the anti-apoptotic molecule Bcl-xL prevents cell death induced by IER3IP1 suppression. Moreover, IER3IP1 also regulates activation of the unfolded protein response (UPR). IER3IP1 suppression impairs the Inositol Requiring 1 (IRE1) and PKR-like ER kinase (PERK) arms of UPR. The cell proliferation of MIN6 cells was also decreased in IER3IP1 deficient cells. These results suggest that IER3IP1 suppression induces an increase in cell death and a decrease in cell proliferation in MIN6 cells, which may be the mechanism that mutations in IER3IP1 lead to diabetes.
- Published
- 2017
11. Rac1-NADPH oxidase signaling promotes CD36 activation under glucotoxic conditions in pancreatic beta cells
- Author
-
Inkyu Lee, Kyu Chang Won, Udayakumar Karunakaran, Jun Sung Moon, and Suma Elumalai
- Subjects
CD36 Antigens ,rac1 GTP-Binding Protein ,0301 basic medicine ,Programmed cell death ,Clinical Biochemistry ,Apoptosis ,RAC1 ,Mitochondrion ,Biology ,Biochemistry ,03 medical and health sciences ,Insulin-Secreting Cells ,Rac1 ,NADPH oxidase ,Beta-cell dysfunction ,CD36 ,Oxidative stress ,Humans ,lcsh:QH301-705.5 ,lcsh:R5-920 ,Oxidase test ,Membrane Glycoproteins ,Organic Chemistry ,NADPH Oxidases ,Mitochondria ,Cell biology ,Oxidative Stress ,Glucose ,Pyrimidines ,030104 developmental biology ,lcsh:Biology (General) ,Diabetes Mellitus, Type 2 ,Aminoquinolines ,biology.protein ,Signal transduction ,Beta cell ,lcsh:Medicine (General) ,Reactive Oxygen Species ,Signal Transduction ,Research Paper - Abstract
We recently reported that cluster determinant 36 (CD36), a fatty acid transporter, plays a pivotal role in glucotoxicity-induced β-cell dysfunction. However, little is known about how glucotoxicity influences CD36 expression. Emerging evidence suggests that the small GTPase Rac1 is involved in the pathogenesis of beta cell dysfunction in type 2 diabetes (T2D). The primary objective of the current study was to determine the role of Rac1 in CD36 activation and its impact on β-cell dysfunction in diabetes mellitus. To address this question, we subjected INS-1 cells and human beta cells (1.1B4) to high glucose conditions (30 mM) in the presence or absence of Rac1 inhibition either by NSC23766 (Rac1 GTPase inhibitor) or small interfering RNA. High glucose exposure in INS-1 and human beta cells (1.1b4) resulted in the activation of Rac1 and induced cell apoptosis. Rac1 activation mediates NADPH oxidase (NOX) activation leading to elevated ROS production in both cells. Activation of the Rac1-NOX complex by high glucose levels enhanced CD36 expression in INS-1 and human 1.1b4 beta cell membrane fractions. The inhibition of Rac1 by NSC23766 inhibited NADPH oxidase activity and ROS generation induced by high glucose concentrations in INS-1 & human 1.1b4 beta cells. Inhibition of Rac1-NOX complex activation by NSC23766 significantly reduced CD36 expression in INS-1 and human 1.1b4 beta cell membrane fractions. In addition, Rac1 inhibition by NSC23766 significantly reduced high glucose-induced mitochondrial dysfunction. Furthermore, NADPH oxidase inhibition by VAS2870 also attenuated high glucose-induced ROS generation and cell apoptosis. These results suggest that Rac1-NADPH oxidase dependent CD36 expression contributes to high glucose-induced beta cell dysfunction and cell death., Graphcal abstract Rac1- NADPH oxidase induces CD36 expression under high glucose. High glucose induced Rac1-NADPH oxidase complex induced CD36 trafficking to the plasma membrane. Increased plasma membrane expression of CD36 may increase influx of free fatty acids into the cell and increase the dysfunction of beta cells. High levels of ROS enhance the oxidative damage and exacerbate the beta cell apoptosis by mitochondrial dysfunction.fx1, Highlights • High glucose induce β-cell damage by Rac1 and NADPH oxidase activation. • High glucose induced Rac1-NADPH oxidase mediate CD36 expression and mitochondrial dysfunction. • Inhibition of Rac1 suppressed high glucose induced NADPH oxidase activity and downregulated CD36 expression. • Inhibition of Rac1 or NADPH oxidase prevent high glucose induced mitochondrial dysfunction and β-cell apoptosis.
- Published
- 2017
12. The role of ORMDL3/ATF6 in compensated beta cell proliferation during early diabetes
- Author
-
Xiangwei Xiao, Shane Fischbach, Feifei Sheng, Baolan Sun, and Weixia Yang
- Subjects
Blood Glucose ,Aging ,medicine.medical_specialty ,Adolescent ,Activating transcription factor ,Mitochondrial Proteins ,Mice ,Mice, Inbred NOD ,Internal medicine ,Diabetes mellitus ,Cell Line, Tumor ,Insulin-Secreting Cells ,medicine ,Diabetes Mellitus ,Leukocytes ,Animals ,Humans ,Insulin ,unfolded protein response (UPR) ,ATF6 ,NOD mice ,Cell Proliferation ,Glycated Hemoglobin ,Type 1 diabetes ,Chemistry ,Endoplasmic reticulum ,ORMDL3 ,Membrane Proteins ,beta cell proliferation ,Cell Biology ,medicine.disease ,Activating Transcription Factor 6 ,Endocrinology ,Diabetes Mellitus, Type 1 ,Case-Control Studies ,Unfolded protein response ,Female ,Insulinoma ,Beta cell ,Apoptosis Regulatory Proteins ,ER stress ,Research Paper - Abstract
Endoplasmic reticulum (ER) stress in beta cells induces a signaling network called the unfolded protein response (UPR), which plays a dual role in diabetes. A key regulator of ER-stress and UPR, the orosomucoid 1-like protein 3 (ORMDL3), has been shown to regulate airway remodeling through a major UPR protein, activating transcription factor 6 (ATF6), but the contribution of this regulatory axis to compensatory pancreatic beta cell proliferation in diabetes has not been studied. Here, we detected significantly lower levels of ORMDL3 mRNA in leukocytes of peripheral blood specimens from type 1 diabetes (T1D) children, compared to normal children. Moreover, these ORMDL3 levels in T1D children exhibited further decreases upon follow-up. ORMDL3 levels in islets from NOD mice, a mouse model for T1D in humans, showed a mild increase before diabetes onset, but a gradual decrease subsequently. In high glucose culture, beta cell proliferation, but not apoptosis, was increased by overexpression of ORMDL3 levels, likely mediated by its downstream factor ATF6. Mechanistically, ORMDL3 transcriptionally activated ATF6, which was confirmed in a promoter reporter assay. Together, our data suggest that ORMDL3 may increase beta cell proliferation through ATF6 as an early compensatory change in response to diabetes.
- Published
- 2019
13. Characterization of host defense molecules in the human pancreas
- Author
-
Anton Stenwall, Olle Korsgren, Oskar Skog, and Sofie Ingvast
- Subjects
0301 basic medicine ,Male ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Gene Expression ,Pancreatitis-Associated Proteins ,Cathelicidin ,Defensins ,0302 clinical medicine ,Endocrinology ,pancreas ,Child ,bacteria ,Defensin ,diabetes ,Immunohistochemistry ,Tissue Donors ,3. Good health ,medicine.anatomical_structure ,Child, Preschool ,Endokrinologi och diabetes ,Female ,Beta cell ,Pancreas ,Research Paper ,Adult ,Adolescent ,030209 endocrinology & metabolism ,Biology ,Endocrinology and Diabetes ,GPI-Linked Proteins ,03 medical and health sciences ,Young Adult ,medicine ,Humans ,Islet of Langerhans ,Innate immune system ,Infant ,Immunology in the medical area ,medicine.disease ,beta cell ,030104 developmental biology ,Secretory protein ,Diabetes Mellitus, Type 1 ,Case-Control Studies ,Immunologi inom det medicinska området ,Immunology ,Pancreatitis ,defensin - Abstract
The gut microbiota can play a role in pancreatitis and, likely, in the development of type 1 diabetes (T1D). Anti-microbial peptides and secretory proteins are important mediators of the innate immune response against bacteria but their expression in the human pancreas is not fully known. In this study, immunohistochemistry was used to analyze the expression of seven anti-microbial peptides (Defensin alpha 1, alpha 4, beta 1-4 and Cathelicidin) and two secretory proteins with known antimicrobial properties (REG3A and GP2) in pancreatic and duodenal biopsies from 10 non-diabetic organ donors and one organ donor that died at onset of T1D. Immunohistochemical data was compared with previously published whole-transcriptome data sets. Seven (Defensin alpha 1, beta 2, beta 3, alpha 4, GP2, Cathelicidin, and REG3A) host defense molecules showed positive staining patterns in most non-diabetic organ donors, whereas two (Defensin beta 1 and beta 4) were negative in all non-diabetic donors. Two molecules (Defensin alpha 1 and GP2) were restricted to the exocrine pancreas whereas two (Defensin beta 3, alpha 4) were only expressed in islet tissue. Cathelicidin, beta 2, and REG3A were expressed in both islets and exocrine tissue. The donor that died at onset of T1D had generally less positivity for the host defense molecules, but, notably, this pancreas was the only one where defensin beta 1 was found. Neither donor age, immune-cell infiltration, nor duodenal expression correlated to the pancreatic expression of host defense molecules. In conclusion, these findings could have important implications for the inflammatory processes in diabetes and pancreatitis as we find several host defense molecules expressed by the pancreatic tissue.
- Published
- 2019
14. Dual Effect of Rosuvastatin on Glucose Homeostasis Through Improved Insulin Sensitivity and Reduced Insulin Secretion
- Author
-
Anna Wendt, Thomas Reinbothe, Jones K. Ofori, Inês G. Mollet, Karin G. Stenkula, Jenny Vikman, Lena Eliasson, Helena Anna Malm, Jonathan L.S. Esguerra, and Vishal A. Salunkhe
- Subjects
Medicin och hälsovetenskap ,Glucose uptake ,medicine.medical_treatment ,lcsh:Medicine ,030204 cardiovascular system & hematology ,Glucose homeostasis ,Medical and Health Sciences ,Mice ,0302 clinical medicine ,Insulin-Secreting Cells ,Insulin Secretion ,Adipocytes ,Medicine ,Homeostasis ,Insulin ,Ca2+ measurements ,Rosuvastatin Calcium ,lcsh:R5-920 ,Insulin secretion ,Ca(2+) measurements ,General Medicine ,Beta cell ,Muscle ,Female ,lcsh:Medicine (General) ,medicine.drug ,Research Paper ,Islet ,medicine.medical_specialty ,Adipose tissue ,030209 endocrinology & metabolism ,Diet, High-Fat ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Insulin resistance ,Diabetes mellitus ,Internal medicine ,OGTT ,Animals ,Rosuvastatin ,Calcium Signaling ,business.industry ,lcsh:R ,nutritional and metabolic diseases ,Statin ,medicine.disease ,Diet ,Ca2 + measurements ,High-Fat ,Endocrinology ,Glucose ,Calcium ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Insulin Resistance ,business ,Transmission electron microscopy - Abstract
Statins are beneficial in the treatment of cardiovascular disease (CVD), but these lipid-lowering drugs are associated with increased incidence of new on-set diabetes. The cellular mechanisms behind the development of diabetes by statins are elusive. Here we have treated mice on normal diet (ND) and high fat diet (HFD) with rosuvastatin. Under ND rosuvastatin lowered blood glucose through improved insulin sensitivity and increased glucose uptake in adipose tissue. In vitro rosuvastatin reduced insulin secretion and insulin content in islets. In the beta cell Ca2 + signaling was impaired and the density of granules at the plasma membrane was increased by rosuvastatin treatment. HFD mice developed insulin resistance and increased insulin secretion prior to administration of rosuvastatin. Treatment with rosuvastatin decreased the compensatory insulin secretion and increased glucose uptake. In conclusion, our data shows dual effects on glucose homeostasis by rosuvastatin where insulin sensitivity is improved, but beta cell function is impaired., Highlights • Rosuvastatin lowered blood glucose in vivo most likely due to improved glucose uptake. • Rosuvastatin reduced insulin content and impaired Ca2 + signaling in beta cells leading to reduced insulin secretion. • Dual effects of rosuvastatin in HFD mice though decreased compensatory insulin secretion and increased glucose uptake. Statins are a group of drugs used to lower blood cholesterol in individuals with a risk of developing cardiovascular disease. It has been shown in several studies that statins increase the risk of developing type 2 diabetes. This increased risk has not yet been explained. We have investigated the effect of rosuvastatin on blood glucose regulation in mice. We found that rosuvastatin has a beneficial effect on glucose uptake in muscles which results in lowered blood glucose. However, in the insulin producing beta cells rosuvastatin altered normal cell function something that might increase the risk of developing type 2 diabetes.
- Published
- 2016
15. Redox homeostasis and cell cycle activation mediate beta-cell mass expansion in aged, diabetes-prone mice under metabolic stress conditions: Role of thioredoxin-interacting protein (TXNIP)
- Author
-
Annette Schürmann, Markus Jähnert, Annika Höhn, Tanina Flore, Tilman Grune, Wenke Jonas, Richard Kehm, Tobias Jung, Jeannette König, Mandy Stadion, and Stefanie Deubel
- Subjects
0301 basic medicine ,Aging ,Beta-cells ,medicine.medical_specialty ,Thioredoxin-Interacting Protein ,medicine.medical_treatment ,Clinical Biochemistry ,Mice, Obese ,Type 2 diabetes ,Biology ,Biochemistry ,Mice ,03 medical and health sciences ,Thioredoxins ,0302 clinical medicine ,Insulin resistance ,Stress, Physiological ,Insulin-Secreting Cells ,Diabetes mellitus ,Internal medicine ,medicine ,Thioredoxin-interacting protein ,Animals ,Homeostasis ,Redox homeostasis ,Cell cycle ,Metabolic stress ,lcsh:QH301-705.5 ,lcsh:R5-920 ,Insulin ,Cell Cycle ,Organic Chemistry ,medicine.disease ,030104 developmental biology ,Endocrinology ,lcsh:Biology (General) ,Diabetes Mellitus, Type 2 ,Beta cell ,lcsh:Medicine (General) ,Carrier Proteins ,Oxidation-Reduction ,030217 neurology & neurosurgery ,TXNIP ,Research Paper - Abstract
Overnutrition contributes to insulin resistance, obesity and metabolic stress, initiating a loss of functional beta-cells and diabetes development. Whether these damaging effects are amplified in advanced age is barely investigated. Therefore, New Zealand Obese (NZO) mice, a well-established model for the investigation of human obesity-associated type 2 diabetes, were fed a metabolically challenging diet with a high-fat, carbohydrate restricted period followed by a carbohydrate intervention in young as well as advanced age. Interestingly, while young NZO mice developed massive hyperglycemia in response to carbohydrate feeding, leading to beta-cell dysfunction and cell death, aged counterparts compensated the increased insulin demand by persistent beta-cell function and beta-cell mass expansion. Beta-cell loss in young NZO islets was linked to increased expression of thioredoxin-interacting protein (TXNIP), presumably initiating an apoptosis-signaling cascade via caspase-3 activation. In contrast, islets of aged NZOs exhibited a sustained redox balance without changes in TXNIP expression, associated with higher proliferative potential by cell cycle activation. These findings support the relevance of a maintained proliferative potential and redox homeostasis for preserving islet functionality under metabolic stress, with the peculiarity that this adaptive response emerged with advanced age in diabetes-prone NZO mice., Graphical abstract Image 1, Highlights • Differential expression of redox and cell cycle genes in young and aged islets. • Increased TXNIP expression is associated with the induction of beta-cell apoptosis. • Islets of aged mice maintained redox homeostasis and proliferative potential. • Aging under diet-induced metabolic stress does not amplify beta-cell failure.
- Published
- 2020
16. PDL1 is expressed in the islets of people with type 1 diabetes and is up-regulated by interferons-α and-γ via IRF1 induction
- Author
-
Anne Op De Beeck, Sarah J. Richardson, Piero Marchetti, Lorella Marselli, Decio L. Eizirik, Pia Leete, Maikel Luis Colli, Knut Dahl-Jørgensen, Reinaldo Sousa Dos Santos, Alexandra Coomans de Brachène, Lars Krogvold, Jessica R. Chaffey, Noel G. Morgan, Flavia M.M. Paula, Angela Castela, Jessica L. E. Hill, and Laura Marroquí
- Subjects
0301 basic medicine ,Genetics and Molecular Biology (all) ,Research paper ,endocrine system diseases ,Immune checkpoint inhibitor ,Biochemistry ,B7-H1 Antigen ,immune system diseases ,Insulin-Secreting Cells ,Interferon gamma ,STAT1 ,Child ,biology ,General Medicine ,Sciences bio-médicales et agricoles ,Middle Aged ,3. Good health ,medicine.anatomical_structure ,Type 1 diabetes ,Child, Preschool ,Antibody ,Beta cell ,medicine.drug ,Type 1 ,Adult ,endocrine system ,Adolescent ,Pancreatic islets ,Pancreatic beta cells ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Interferon-gamma ,Islets of Langerhans ,Young Adult ,Downregulation and upregulation ,PDL1 ,medicine ,Diabetes Mellitus ,Gene silencing ,CD274 ,Humans ,Preschool ,PDL-1 ,IRF1 ,Biomarkers ,Diabetes Mellitus, Type 1 ,Interferon Regulatory Factor-1 ,Interferon-alpha ,Gene Expression Regulation ,Biochemistry, Genetics and Molecular Biology (all) ,nutritional and metabolic diseases ,030104 developmental biology ,Cell culture ,Cancer research ,biology.protein - Abstract
Antibodies targeting PD-1 and its ligand PDL1 are used in cancer immunotherapy but may lead to autoimmune diseases, including type 1 diabetes (T1D). It remains unclear whether PDL1 is expressed in pancreatic islets of people with T1D and how is it regulated., “Innovative Medicines Initiative 2”;“European Union(EU)”“Horizon2020”; and EFPIA”,“JDRF International” and The Leona M.and Harry B.Helmsley Charitable Trust”, SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2018
- Full Text
- View/download PDF
17. Glucagon-Like Peptide-1 Receptor Agonist and Glucagon Increase Glucose-Stimulated Insulin Secretion in Beta Cells via Distinct Adenylyl Cyclases
- Author
-
Young-Sun Lee and Hee-Sook Jun
- Subjects
0301 basic medicine ,Glucagon-like peptide-1 ,medicine.medical_specialty ,endocrine system ,insulin secretion ,medicine.medical_treatment ,Mice, Obese ,Diet, High-Fat ,Glucagon ,Alpha cell ,Glucagon-Like Peptide-1 Receptor ,Cell Line ,03 medical and health sciences ,Islets of Langerhans ,Mice ,Insulin resistance ,Internal medicine ,Insulin-Secreting Cells ,medicine ,Cyclic AMP ,Diabetes Mellitus ,Animals ,Humans ,Insulin ,education ,alpha cell ,Glucagon-like peptide 1 receptor ,education.field_of_study ,Chemistry ,Venoms ,Glucagon secretion ,General Medicine ,Soluble adenylyl cyclase ,medicine.disease ,beta cell ,030104 developmental biology ,Endocrinology ,Glucose ,Adenylyl Cyclase Inhibitors ,Dideoxyadenosine ,Exenatide ,Beta cell ,Insulin Resistance ,Peptides ,Adenylyl Cyclases ,Research Paper - Abstract
Diabetes mellitus is a chronic disease in which the pancreas no longer produces enough insulin. Pancreatic alpha cell mass increases in response to insufficient insulin secretion. However, the reason for this increase is not clear. It is possible that the increased alpha-cells may stimulate compensatory insulin release in response to the insufficient insulin such as insulin resistance. In this study, we investigated whether glucagon and glucagon-like peptide-1 (GLP-1), hormones produced by alpha cells, contribute to insulin secretion in INS-1 cells, a beta cell line. We confirmed that alpha cell area in the pancreatic islets and glucagon secretion were increased in HFD-induced obese mice. Co-treatment with glucagon and exendin-4 (Ex-4), a GLP-1 receptor agonist, additively increased glucose-stimulated insulin secretion in INS-1 cells. In parallel, cAMP production was also additively increased by co-treatment with these hormones. The increase of insulin secretion by Ex-4 in the presence of high glucose was inhibited by 2'5'-dideoxyadenosine, a transmembrane adenylyl cyclase inhibitor, but not by KH-7, a soluble adenylyl cyclase inhibitor. The increase of insulin secretion by glucagon in INS-1 cells was inhibited by both 2'5'-dideoxyadenosine and KH-7. We suggest that glucagon and GLP-1 produced from alpha cells additively increase cAMP and insulin secretion in the presence of high glucose via distinct adenylyl cyclases in INS-1 cells, and this may contribute to the compensatory increase of insulin secretion by an increase of pancreatic alpha cell mass under conditions of insulin resistance.
- Published
- 2017
18. HX-1171 attenuates pancreatic β-cell apoptosis and hyperglycemia-mediated oxidative stress via Nrf2 activation in streptozotocin-induced diabetic model
- Author
-
Su-Hyun Shin, Jimin Kim, Jong-Koo Kang, and Jae Wha Kim
- Subjects
0301 basic medicine ,endocrine system diseases ,Cell ,030209 endocrinology & metabolism ,Pharmacology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Diabetes mellitus ,medicine ,HX-1171 ,Nrf2 activator ,diabetes ,Activator (genetics) ,business.industry ,Pancreatic islets ,Streptozotocin ,medicine.disease ,beta cell ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,hyperglycemia ,Beta cell ,business ,Oxidative stress ,medicine.drug ,Research Paper - Abstract
// Jimin Kim 1, 2 , Su-Hyun Shin 1, 2 , Jong-Koo Kang 3 and Jae Wha Kim 1, 2 1 Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea 2 Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, Republic of Korea 3 Department of Laboratory Animal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea Correspondence to: Jae Wha Kim, email: wjkim@kribb.re.kr Keywords: HX-1171; Nrf2 activator; beta cell; diabetes; hyperglycemia Received: July 17, 2017 Accepted: September 04, 2017 Epub: March 23, 2018 Published: May 11, 2018 ABSTRACT Streptozotocin (STZ) acts specifically on pancreatic beta cells, inducing cell destruction and cell dysfunction, resulting in diabetes. Many studies have reported that nuclear factor-erythroid 2-related factor 2 (Nrf2), a main regulator of antioxidant expression, prevents and improves diabetes-related diseases. In this study, we investigated the antidiabetic effect of the newly discovered Nrf2 activator, HX-1171, in the STZ-induced diabetic mouse model. HX-1171 enhanced insulin secretion by reducing STZ-induced cell apoptosis, and decreased intracellular reactive oxygen species (ROS) generation by upregulating the expression of antioxidant enzymes through Nrf2 activation in INS-1 pancreatic beta cells. In STZ-induced diabetic mice, HX-1171 administration significantly lowered blood glucose levels and restored blood insulin levels. In the STZ-only injected mice, the pancreatic islets showed morphological changes and loss of function, whereas the HX-1171-treated group was similar to that of the control group. These results suggest that HX-1171 may be developed as a promising therapeutic agent for diabetes-related diseases.
- Published
- 2017
19. δ-cells and β-cells are electrically coupled and regulate α-cell activity via somatostatin
- Author
-
Briant, L. J. B., Reinbothe, T. M., Spiliotis, I., Miranda, C., Rodriguez, B., and Rorsman, P.
- Subjects
Male ,endocrine system ,Somatostatin-Secreting Cells ,Cell Communication ,Mice ,computer modelling ,Insulin-Secreting Cells ,Animals ,Insulin ,Computer Simulation ,alpha cell ,optogenetics ,Cells, Cultured ,delta cell ,Gap Junctions ,electrophysiology ,Glucagon ,beta cell ,Glucose ,Glucagon-Secreting Cells ,Islet cell ,Calcium ,Female ,Endocrine, Nutrition and Metabolism ,Somatostatin ,Research Paper ,Perspectives - Abstract
Key points We used a mouse expressing a light‐sensitive ion channel in β‐cells to understand how α‐cell activity is regulated by β‐cells.Light activation of β‐cells triggered a suppression of α‐cell activity via gap junction‐dependent activation of δ‐cells.Mathematical modelling of human islets suggests that 23% of the inhibitory effect of glucose on glucagon secretion is mediated by β‐cells via gap junction‐dependent activation of δ‐cells/somatostatin secretion. Abstract Glucagon, the body's principal hyperglycaemic hormone, is released from α‐cells of the pancreatic islet. Secretion of this hormone is dysregulated in type 2 diabetes mellitus but the mechanisms controlling secretion are not well understood. Regulation of glucagon secretion by factors secreted by neighbouring β‐ and δ‐cells (paracrine regulation) have been proposed to be important. In this study, we explored the importance of paracrine regulation by using an optogenetic strategy. Specific light‐induced activation of β‐cells in mouse islets expressing the light‐gated channelrhodopsin‐2 resulted in stimulation of electrical activity in δ‐cells but suppression of α‐cell activity. Activation of the δ‐cells was rapid and sensitive to the gap junction inhibitor carbenoxolone, whereas the effect on electrical activity in α‐cells was blocked by CYN 154806, an antagonist of the somatostatin‐2 receptor. These observations indicate that optogenetic activation of the β‐cells propagates to the δ‐cells via gap junctions, and the consequential stimulation of somatostatin secretion inhibits α‐cell electrical activity by a paracrine mechanism. To explore whether this pathway is important for regulating α‐cell activity and glucagon secretion in human islets, we constructed computational models of human islets. These models had detailed architectures based on human islets and consisted of a collection of >500 α‐, β‐ and δ‐cells. Simulations of these models revealed that this gap junctional/paracrine mechanism accounts for up to 23% of the suppression of glucagon secretion by high glucose., Key points We used a mouse expressing a light‐sensitive ion channel in β‐cells to understand how α‐cell activity is regulated by β‐cells.Light activation of β‐cells triggered a suppression of α‐cell activity via gap junction‐dependent activation of δ‐cells.Mathematical modelling of human islets suggests that 23% of the inhibitory effect of glucose on glucagon secretion is mediated by β‐cells via gap junction‐dependent activation of δ‐cells/somatostatin secretion.
- Published
- 2017
20. Intermittent fasting preserves beta-cell mass in obesity-induced diabetes via the autophagy-lysosome pathway
- Author
-
Fumihiko Urano, Rebecca J. Godar, Maria S. Remedi, Paul Saftig, John T. Murphy, Krzysztof L. Hyrc, Nidhi Rohatgi, Abhinav Diwan, Xiucui Ma, Babak Razani, Haiyan Liu, Jana Mahadevan, Ali Javaheri, Michael L. McDaniel, and Connie A. Marshall
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Research Paper - Basic Science ,Nerve Tissue Proteins ,030209 endocrinology & metabolism ,Biology ,Diabetes Mellitus, Experimental ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Downregulation and upregulation ,Insulin-Secreting Cells ,Lysosomal-Associated Membrane Protein 2 ,Internal medicine ,Diabetes mellitus ,Lysosome ,Intermittent fasting ,Autophagy ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Insulin ,Obesity ,Molecular Biology ,Fasting ,Cell Biology ,BECN1 ,medicine.disease ,Mice, Mutant Strains ,Mitochondria ,Up-Regulation ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,Beclin-1 ,Beta cell ,Lysosomes - Abstract
Obesity-induced diabetes is characterized by hyperglycemia, insulin resistance, and progressive beta cell failure. In islets of mice with obesity-induced diabetes, we observe increased beta cell death and impaired autophagic flux. We hypothesized that intermittent fasting, a clinically sustainable therapeutic strategy, stimulates autophagic flux to ameliorate obesity-induced diabetes. Our data show that despite continued high-fat intake, intermittent fasting restores autophagic flux in islets and improves glucose tolerance by enhancing glucose-stimulated insulin secretion, beta cell survival, and nuclear expression of NEUROG3, a marker of pancreatic regeneration. In contrast, intermittent fasting does not rescue beta-cell death or induce NEUROG3 expression in obese mice with lysosomal dysfunction secondary to deficiency of the lysosomal membrane protein, LAMP2 or haplo-insufficiency of BECN1/Beclin 1, a protein critical for autophagosome formation. Moreover, intermittent fasting is sufficient to provoke beta cell death in nonobese lamp2 null mice, attesting to a critical role for lysosome function in beta cell homeostasis under fasting conditions. Beta cells in intermittently-fasted LAMP2- or BECN1-deficient mice exhibit markers of autophagic failure with accumulation of damaged mitochondria and upregulation of oxidative stress. Thus, intermittent fasting preserves organelle quality via the autophagy-lysosome pathway to enhance beta cell survival and stimulates markers of regeneration in obesity-induced diabetes.
- Published
- 2017
- Full Text
- View/download PDF
21. Time-dependent Mechanisms in Beta-cell Glucose Sensing
- Author
-
Morten Colding-Jørgensen and Thomas Vagn Korsgaard
- Subjects
chemistry.chemical_classification ,medicine.medical_specialty ,Pulse (signal processing) ,Glucokinase ,Insulin ,medicine.medical_treatment ,Biophysics ,Cell Biology ,Carbohydrate metabolism ,Biology ,Atomic and Molecular Physics, and Optics ,Enzyme ,Endocrinology ,chemistry ,Negative feedback ,Internal medicine ,medicine ,Glucose homeostasis ,Beta cell ,Molecular Biology ,Research Paper - Abstract
The relation between plasma glucose and insulin release from pancreatic beta-cells is not stationary in the sense that a given glucose concentration leads to a specific rate of insulin secretion. A number of time-dependent mechanisms appear to exist that modify insulin release both on a short and a longer time scale. Typically, two phases are described. The first phase, lasting up to 10 min, is a pulse of insulin release in response to fast changes in glucose concentration. The second phase is a more steady increase of insulin release over minutes to hours, if the elevated glucose concentration is sustained. The paper describes the glucose sensing mechanism via the complex dynamics of the key enzyme glucokinase, which controls the first step in glucose metabolism: phosphorylation of glucose to glucose-6-phosphate. Three time-dependent phenomena (mechanisms) are described. The fastest, corresponding to the first phase, is a delayed negative feedback regulating the glucokinase activity. Due to the delay, a rapid glucose increase will cause a burst of activity in the glucose sensing system, before the glucokinase is down-regulated. The second mechanism corresponds to the translocation of glucokinase from an inactive to an active form. As the translocation is controlled by the product(s) of the glucokinase reaction rather than by the substrate glucose, this mechanism gives a positive, but saturable, feedback. Finally, the release of the insulin granules is assumed to be enhanced by previous glucose exposure, giving a so-called glucose memory to the beta-cells. The effect depends on the insulin release of the cells, and this mechanism constitutes a second positive, saturable feedback system. Taken together, the three phenomena describe most of the glucose sensing behaviour of the beta-cells. The results indicate that the insulin release is not a precise function of the plasma glucose concentration. It rather looks as if the beta-cells just increase the insulin production, until the plasma glucose has returned to normal. This type of integral control has the advantage that the precise glucose sensitivity of the beta-cells is not important for normal glucose homeostasis.
- Published
- 2006
22. Imeglimin lowers glucose primarily by amplifying glucose-stimulated insulin secretion in high-fat-fed rodents
- Author
-
Max C. Petersen, Sébastien Bolze, Rebecca L. Cardone, Gerald I. Shulman, K F Petersen, Pascale Fouqueray, Sophie Hallakou-Bozec, Dongyan Zhang, Richard G. Kibbey, and Rachel J. Perry
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,medicine.medical_specialty ,Imeglimin ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Diet, High-Fat ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Insulin resistance ,Physiology (medical) ,Internal medicine ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Animals ,Hypoglycemic Agents ,Insulin ,Insulin secretion ,geography ,geography.geographical_feature_category ,business.industry ,Triazines ,Fasting ,Glucose clamp technique ,medicine.disease ,Islet ,Postprandial Period ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Glucose ,chemistry ,Liver ,Call for Papers ,Glucose Clamp Technique ,Hemoglobin ,Beta cell ,Insulin Resistance ,business - Abstract
Imeglimin is a promising new oral antihyperglycemic agent that has been studied in clinical trials as a possible monotherapy or add-on therapy to lower fasting plasma glucose and improve hemoglobin A1c(1–3, 9). Imeglimin was shown to improve both fasting and postprandial glycemia and to increase insulin secretion in response to glucose during a hyperglycemic clamp after 1-wk of treatment in type 2 diabetic patients. However, whether the β-cell stimulatory effect of imeglimin is solely or partially responsible for its effects on glycemia remains to be fully confirmed. Here, we show that imeglimin directly activates β-cell insulin secretion in awake rodents without affecting hepatic insulin sensitivity, body composition, or energy expenditure. These data identify a primary amplification rather than trigger the β-cell mechanism that explains the acute, antidiabetic activity of imeglimin.
- Published
- 2016
23. Human placental lactogen (hPL-A) activates signaling pathways linked to cell survival and improves insulin secretion in human pancreatic islets
- Author
-
Massimo Federici, Renato Lauro, Barbara Bartolini, Luca Bova, Marco F. Lombardo, Davide Lauro, Fabiana De Angelis, Federico Bertuzzi, Barbara Capuani, Giulia Donadel, and Rita Nano
- Subjects
endocrine system ,medicine.medical_specialty ,Settore MED/09 - Medicina Interna ,Cell Survival ,MAP Kinase Signaling System ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Settore MED/50 - Scienze Tecniche Mediche Applicate ,Apoptosis ,Biology ,p38 Mitogen-Activated Protein Kinases ,Settore MED/13 - Endocrinologia ,Mice ,Endocrinology ,Human placental lactogen ,Cell Line, Tumor ,Insulin-Secreting Cells ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Humans ,Insulin ,Settore MED/49 - Scienze Tecniche Dietetiche Applicate ,Extracellular Signal-Regulated MAP Kinases ,Insulinoma ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Settore MED/04 - Patologia Generale ,Human Growth Hormone ,Pancreatic islets ,Placental Lactogen ,medicine.disease ,Prolactin ,Rats ,Pancreatic Neoplasms ,medicine.anatomical_structure ,Beta cell ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Research Paper - Abstract
The search for factors either promoting islets proliferation or survival during adult life is a major issue for both type 1 and 2 diabetes mellitus. Among factors with mitogenic activity on pancreatic β-cells, human placental lactogen (hPL) showed stronger activity when compared to the other lactogen hormones: growth hormone (GH) and prolactin (PRL). The aim of the present work is to elucidate the biological and molecular events of hPL isoform A (hPL-A) activity on human cultured islets. We used pure human pancreatic islets and insulinoma cell lines (βTC-1 and RIN, murine and rat respectively) stimulated with hPL-A recombinant protein and we compared hPL-A activity with that of hGH. We showed that hPL-A inhibits apoptosis, both in insulinoma and human islets, by the phosphorylation of AKT protein. Indeed, the antiapoptotic role of hPL-A was mediated by PI3K, p38 and it was independent by PKA, Erk1/2. Compared with hGH, hPL-A modulated at different intervals and/or intensity by the phosphorylation of JAKs/STATs and MAPKinases. Moreover, hPL-A induced PDX-1 intracellular expression, improving beta cell activity and ameliorating insulin secretion in response to high glucose stimulation. Our data support the idea that hPL-A is involved in the regulation of beta cells activity. Importantly, we found that hPL-A can preserve and improve the ability of purified human pancreatic islets cultured to secrete insulin in vitro.
- Published
- 2011
24. Pim3 negatively regulates glucose-stimulated insulin secretion
- Author
-
Gene C. Webb, Gregory Vlacich, Martijn C. Nawijn, Donald F. Steiner, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Male ,insulin secretion ,Endocrinology, Diabetes and Metabolism ,Cell ,beta-cell ,Suppressor of Cytokine Signaling Proteins ,PHOSPHOINOSITIDE 3-KINASE ,Mice ,PHOSPHORYLATES BAD ,Endocrinology ,Insulin-Secreting Cells ,Insulin ,SOCS6 ,Receptor ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,biology ,ACTIVATED PROTEIN-KINASE ,ERK ,medicine.anatomical_structure ,Organ Specificity ,B-CELLS ,Beta cell ,Signal transduction ,signal transduction ,Research Paper ,Pim3 ,EXPRESSION ,medicine.medical_specialty ,MAP Kinase Signaling System ,Protein Serine-Threonine Kinases ,MICE LACKING ,Cell Line ,Islets of Langerhans ,Organ Culture Techniques ,Proto-Oncogene Proteins ,Internal medicine ,medicine ,Animals ,Secretion ,RNA, Messenger ,Insulinoma ,Cell Size ,LANGERHANS ,Phosphoinositide 3-kinase ,GENE-TRANSCRIPTION ,RECEPTOR ,Gene Expression Profiling ,medicine.disease ,Gene Expression Regulation ,Hyperglycemia ,biology.protein ,PANCREATIC BETA-CELLS ,Insulin Resistance - Abstract
Pancreatic beta-cell response to glucose stimulation is governed by tightly regulated signaling pathways which have not been fully characterized. A screen for novel signaling intermediates identified Pim3 as a glucose-responsive gene in the beta-cell, and here, we characterize its role in the regulation of beta-cell function. Pim3 expression in the beta-cell was first observed through microarray analysis on glucose-stimulated murine insulinoma (MIN6) cells where expression was strongly and transiently induced. In the pancreas, Pim3 expression exhibited similar dynamics and was restricted to the beta-cell. Perturbation of Pim3 function resulted in enhanced glucose-stimulated insulin secretion, both in MIN6 cells and in isolated islets from Pim3(-/-) mice, where the augmentation was specifically seen in the second phase of secretion. Consequently, Pim3(-/-) mice displayed an increased glucose tolerance in vivo. Interestingly, Pim3(-/-) mice also exhibited increased insulin sensitivity. Glucose stimulation of isolated Pim3(-/-) islets resulted in increased phosphorylation of ERK1/2, a kinase involved in regulating beta-cell response to glucose. Pim3 was also found to physically interact with SOCS6 and SOCS6 levels were strongly reduced in Pim3(-/-) islets. Overexpression of SOCS6 inhibited glucose-induced ERK1/2 activation, strongly suggesting that Pim3 regulates ERK1/2 activity through SOCS6. These data reveal that Pim3 is a novel glucose-responsive gene in the beta-cell that negatively regulates insulin secretion by inhibiting the activation of ERK1/2, and through its effect on insulin sensitivity, has potentially a more global function in glucose homeostasis.
- Published
- 2010
25. Sulfonylurea Receptor Knockout Causes Glucose Intolerance in Mice That is Not Alleviated by Concomitant Somatostatin Subtype Receptor 5 Knockout
- Author
-
Xiao-Ping Wang, Michael A. Norman, Stefan Moldovan, Francesco J. DeMayo, F. Charles Brunicardi, and Victor Seghers
- Subjects
Blood Glucose ,medicine.medical_specialty ,Potassium Channels ,Receptors, Drug ,medicine.medical_treatment ,Fluorescent Antibody Technique ,Sulfonylurea Receptors ,Polymerase Chain Reaction ,Mice ,Internal medicine ,Glucose Intolerance ,Insulin Secretion ,Scientific Papers ,Animals ,Insulin ,Medicine ,Receptors, Somatostatin ,Potassium Channels, Inwardly Rectifying ,Pancreas ,Mice, Knockout ,Delta cell ,biology ,business.industry ,Calcium channel ,Glucose Tolerance Test ,Immunohistochemistry ,Potassium channel ,Insulin oscillation ,Perfusion ,Insulin receptor ,Endocrinology ,Models, Animal ,biology.protein ,Sulfonylurea receptor ,ATP-Binding Cassette Transporters ,Surgery ,Beta cell ,business - Abstract
The endocrine pancreas consists of the islets of Langerhans, which are in turn made up of several different cell types. The most common cell type in the islets are the beta cells, which make up 70% of the volume of the islet and are responsible for the secretion of insulin. Other cell types include the alpha cells, which produce glucagon, and the delta cells, which produce somatostatin. These two groups make up approximately 20% and 5%, respectively, of the volume of the islet. 1,2 Insulin secreted from the beta cell acts to regulate the blood glucose levels during fasting and fed states. Somatostatin is an inhibitory peptide discovered in the early 1970s. 3 Further work showed it to be present in a variety of tissues, 4 and as techniques became more sophisticated, it was found in more tissues. 5–7 In the early 1990s, the receptors for somatostatin were isolated, cloned, and characterized. 8–10 The somatostatin receptors (SSTRs) were found to belong to a family of G protein-coupled receptors that affect potassium and calcium channels, cAMP, and protein phosphatases. 11 Further studies suggested that SSTR5 was a likely candidate to be involved with insulin inhibition and that it was present on the beta cell. 12–18 To further characterize the role of SSTR5 on insulin secretion, an SSTR5 knockout (KO) mouse model was created in our laboratory. These mice were viable and fertile and had a subtle aging phenotype in which there was an increased insulin secretion response to glucose stimulation in year-old mice. ATP-sensitive potassium (KATP) channels consist of two different subunits: a sulfonylurea receptor (Sur), which belongs to the ATP-binding cassette family, which as a group function to transport substances across cell membranes, and an inwardly rectifying potassium channel (Kir). 19,20 Together these subunits form an ion channel (Kir) that is regulated by ATP binding to the Sur receptor, and this controls the flux of potassium out of the cell. The sulfonylurea receptor functions as the sensor for the channel and affects opening and closure, which sets the resting membrane of the cell below the threshold for calcium channel activation. 21 During the fed state, glucose is transported into the beta cell via the GLUT2 transporter and metabolized in the cell to ATP. This changes the ATP/ADP ratio in the cell and causes the KATP channel to close. This prevents the exit of potassium from the cell, raising the membrane potential and depolarizing the cell. Once the cell reaches the threshold for calcium channels, voltage-dependent calcium channels open and calcium is released into the cell. The influx of calcium into the cytosol allows calcium to bind to secretory granules, triggering exocytosis and the release of insulin. 22 To support these hypotheses, evidence obtained from Sur1 KO mice showed that the genetic ablation of Sur1 gene results in impaired glucose-stimulated insulin secretion at 3 months of age, characterized by a lack of first-phase insulin secretion and impaired second-phase secretion. These mice also show prolonged glucose levels after glucose stimulation. 23 The purposes of this study were to examine the long-term effects of SSTR5 KO, Sur KO, and Sur/SSTR5 double KO on insulin secretion and glucose levels, and to determine whether the increased secretion of insulin seen with the SSTR5 KO mice would alter the blunted insulin response of the Sur KO mice.
- Published
- 2002
26. VHL-mediated disruption of Sox9 activity compromises β-cell identity and results in diabetes mellitus
- Author
-
Matthias Hebrok, Sapna Puri, and Haruhiko Akiyama
- Subjects
Male ,endocrine system diseases ,1.1 Normal biological development and functioning ,β cell ,Autoimmune Disease ,Medical and Health Sciences ,Cell Line ,Mice ,Underpinning research ,Insulin-Secreting Cells ,Diabetes Mellitus ,Genetics ,Glucose homeostasis ,Animals ,Humans ,2.1 Biological and endogenous factors ,Progenitor cell ,Aetiology ,Metabolic and endocrine ,Regulation of gene expression ,biology ,hypoxia ,dedifferentiation ,Diabetes ,Psychology and Cognitive Sciences ,Wnt signaling pathway ,von-Hippel Lindau ,SOX9 Transcription Factor ,Biological Sciences ,Stem Cell Research ,Hedgehog signaling pathway ,Cell Hypoxia ,beta cell ,Ubiquitin ligase ,Gene Expression Regulation ,Von Hippel-Lindau Tumor Suppressor Protein ,biology.protein ,Cancer research ,Ectopic expression ,TCF7L2 ,Gene Deletion ,Research Paper ,Developmental Biology - Abstract
Precise functioning of the pancreatic β cell is paramount to whole-body glucose homeostasis, and β-cell dysfunction contributes significantly to diabetes mellitus. Using transgenic mouse models, we demonstrate that deletion of the von Hippel-Lindau (Vhlh) gene (encoding an E3 ubiquitin ligase implicated in, among other functions, oxygen sensing in pancreatic β cells) is deleterious to canonical β-cell gene expression. This triggers erroneous expression of factors normally active in progenitor cells, including effectors of the Notch, Wnt, and Hedgehog signaling cascades. Significantly, an up-regulation of the transcription factor Sox9, normally excluded from functional β cells, occurs upon deletion of Vhlh. Sox9 plays important roles during pancreas development but does not have a described role in the adult β cell. β-Cell-specific ectopic expression of Sox9 results in diabetes mellitus from similar perturbations in β-cell identity. These findings reveal that assaults on the β cell that impact the differentiation state of the cell have clear implications toward our understanding of diabetes mellitus.
- Published
- 2013
27. Beta cell specific probing with fluorescent exendin-4 is progressively reduced in type 2 diabetic mouse models
- Author
-
Jonas Ahnfelt-Rønne, Morten Grønbech Rasch, Jacob Hecksher-Sørensen, Lauge Schäffer, and Janne Lehtonen
- Subjects
0301 basic medicine ,Agonist ,endocrine system ,medicine.medical_specialty ,Cell type ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Cell ,Mice, Obese ,030209 endocrinology & metabolism ,Biology ,Diabetes Mellitus, Experimental ,Islets of Langerhans ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Glucagon-Like Peptide 1 ,In vivo ,Insulin-Secreting Cells ,Internal medicine ,Receptors, Glucagon ,medicine ,Animals ,Hypoglycemic Agents ,Venoms ,Pancreatic islets ,digestive, oral, and skin physiology ,Glucagon-like peptide-1 ,Molecular biology ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,Disease Progression ,Exenatide ,Beta cell ,Peptides ,hormones, hormone substitutes, and hormone antagonists ,Ex vivo ,Research Paper - Abstract
Probes based on GLP-1R agonist exendin-4 have shown promise as in vivo β cell tracers. However, questions remain regarding the β cell specificity of exendin-4 probes, and it is unclear if the expression levels of the GLP-1R are affected in a type 2 diabetic state. Using in vivo probing followed by ex vivo imaging we found fluorescent exendin-4 probes to distinctly label the pancreatic islets in mice in a Glp-1r dependent manner. Furthermore, a co-localization study revealed a near 100 percent β cell specificity with less than one percent probing in other analyzed cell types. We then tested if probing was affected in models of type 2 diabetes using the Lepr(db/db) (db/db) and the Diet-Induced Obese (DIO) mouse. Although nearly all β cells continued to be probed, we observed a progressive decline in probing intensity in both models with the most dramatic reduction seen in db/db mice. This was paralleled by a progressive decrease in Glp-1r protein expression levels. These data confirm β cell specificity for exendin-4 based probes in mice. Furthermore, they also suggest that GLP-1R targeting probes may provide a tool to monitor β cell function rather than mass in type 2 diabetic mouse models.
- Published
- 2015
28. No non-redundant function of suppressor of cytokine signaling 2 in insulin producing β-cells
- Author
-
Petra Dames, Andreas Lechner, Ramona Puff, M Weise, Burkhard Göke, and Klaus G. Parhofer
- Subjects
inorganic chemicals ,Blood Glucose ,Male ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Autoimmunity ,Suppressor of Cytokine Signaling Proteins ,Biology ,behavioral disciplines and activities ,Streptozocin ,Diabetes Mellitus, Experimental ,Mice ,Endocrinology ,Internal medicine ,Insulin-Secreting Cells ,medicine ,otorhinolaryngologic diseases ,Tumor Cells, Cultured ,Animals ,SOCS3 ,RNA, Small Interfering ,Mice, Knockout ,Dose-Response Relationship, Drug ,Suppressor of cytokine signaling 1 ,Pancreatic islets ,Streptozotocin ,Research Papers ,Rats ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cytokine ,Organ Specificity ,Gene Knockdown Techniques ,STAT protein ,Female ,sense organs ,Beta cell ,Janus kinase ,psychological phenomena and processes ,medicine.drug - Abstract
The members of the Suppressor of Cytokine Signaling (SOCS) protein family mainly modulate the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. SOCS-1 and SOCS-3 have already been shown to influence growth and apoptosis of pancreatic beta cells. We hypothesized that SOCS-2, which is expressed in pancreatic islets, also contributes to β-cell physiology. We tested this hypothesis in vivo in SOCS-2-/- knockout mice and in vitro in Ins-1E rat insulinoma cells. We found that SOCS-2-/- mice have normal islet insulin secretion and unchanged glucose and insulin tolerance compared to wildtype controls. SOCS-2-/- are bigger than wildtype mice but body weight-corrected β-cell mass and islet morphology were normal. Growth hormone-induced proliferation of Ins-1E cells was not affected by either siRNA-mediated SOCS-2 knockdown or stable SOCS-2 overexpression. Interleukin-1β mediated cell death in vitro was unchanged after SOCS-2 knockdown. Similarly, autoimmune destruction of beta cells in vivo after multiple low-dose injections of streptozotocin (STZ) was not altered in SOCS-2-/- mice. In summary, SOCS-2-/- knockout mice have a normal function of insulin-producing pancreatic β-cells, a fully adapted beta cell mass and a normal morphology of the endocrine islets. Based on in vitro evidence, the increased β-cell mass in the mutants is likely due to indirect adaptive mechanisms and not the result of altered growth hormone signaling within the β-cells. Immune mediated β-cell destruction is also not affected by SOCS-2 ablation in vitro and in vivo.
- Published
- 2010
29. Dual radiotracer analysis of cholinergic neuronal changes in prediabetic mouse pancreas
- Author
-
Jorge Calles-Escandon, Nicole S. Burbank, Sudha Garg, Allen W. Elster, Paige B. Clark, Jonathan Kraas, Pradeep Garg, Janice D. Wagner, Kathryn A. Morton, Michael J. Plaza, and H. Donald Gage
- Subjects
medicine.medical_specialty ,Fluorine Radioisotopes ,Endocrinology, Diabetes and Metabolism ,Tritium ,Prediabetic State ,chemistry.chemical_compound ,Mice ,Endocrinology ,Piperidines ,Internal medicine ,Insulin-Secreting Cells ,Muscarinic acetylcholine receptor ,medicine ,Animals ,Cholinergic neuron ,Neurotransmitter ,Receptor ,Pancreas ,business.industry ,Parasympatholytics ,Original Papers ,Fluorobenzenes ,Radiography ,Medical Laboratory Technology ,medicine.anatomical_structure ,chemistry ,Cholinergic ,Beta cell ,business ,Acetylcholine ,medicine.drug - Abstract
Pancreatic neuronal changes associated with beta cell loss in type 1 diabetes mellitus are complex, involving, in part, parasympathetic mechanisms to compensate for preclinical hyperglycemia. The parasympathetic neurotransmitter acetylcholine (ACh) mediates insulin release via M3 muscarinic receptors on islet beta cells. The vesicular ACh transporter (VAChT) receptor has been shown to be a useful marker of cholinergic activity in vivo. The positron emission tomography (PET) radiotracer (+)-4-[(18)F]fluorobenzyltrozamicol ([(18)F]FBT) binds to the VAChT receptor on presynaptic cholinergic neurons and can be quantified by PET. The compound 4-diphenylacetoxy-N-methylpiperidine (4-DAMP), available in a tritiated form, binds to M3 muscarinic receptors on beta cells and is a potential target for assessing pancreatic beta cell mass. In this study, we investigate the feasibility of dual radiotracer analysis in identifying neurofunctional changes that may signify type 1 diabetes mellitus in its early preclinical state.Ex vivo determinations of pancreatic uptake were performed in prediabetic nonobese diabetic mice and controls after intravenous injection of [(18)F]FBT or 4-[(3)H]DAMP. Beta cell loss in prediabetic mice was confirmed using immunohistochemical methods.[(18)F]FBT uptake was significantly higher in prediabetic pancreata than controls: 3.22 +/- 0.81 and 2.51 +/- 1.04, respectively (P0.03). 4-[(3)H]DAMP uptake was significantly lower in prediabetic pancreata than controls: 0.612 +/- 0.161 and 0.968 +/- 0.364, respectively (P = 0.01).These data suggest that a combination of radiotracer imaging agents that bind to neuronal elements intimately involved in insulin production may be an effective method of evaluating changes associated with early beta cell loss using PET.
- Published
- 2009
30. Normalization of glucose post-transplantation into diabetic rats of pig pancreatic primordia preserved in vitro
- Author
-
Marc R. Hammerman and Sharon A. Rogers
- Subjects
Transplantation ,Embryology ,medicine.medical_specialty ,Xenotransplantation ,medicine.medical_treatment ,fungi ,Biomedical Engineering ,food and beverages ,Biology ,medicine.disease ,Embryonic stem cell ,Endocrinology ,medicine.anatomical_structure ,surgical procedures, operative ,Internal medicine ,Diabetes mellitus ,medicine ,Endocrine system ,Mesenteric lymph nodes ,Primordium ,Beta cell ,Developmental Biology ,Research Paper - Abstract
Embryonic day (E) 28 (E28) pig pancreatic primordia transplanted into the mesentery of non-immunosuppresed steptozotocin (STZ)-diabetic Lewis rats normalize levels of circulating glucose within 2-4 weeks. Exocrine tissue does not differentiate after transplantation of pancreatic primordia. Rather individual endocrine (beta) cells engraft within the mesentery.To determine whether transplanted pig pancreatic primordia engraft, differentiate and function in rat hosts after preservation in vitro, we implanted pig pancreatic primordia into STZ-diabetic rats either directly or after 24 hours of suspension in ice-cold University of Wisconsin (UW) preservation solution with added growth factors. Here we show engraftment in mesentery and mesenteric lymph nodes and normalization of glucose levels in STZ-diabetic rat hosts following transplantation of preserved E28 pig pancreatic primordia comparable to glucose normalization after transplantation of non-preserved E28 pancreatic primordia.
- Published
- 2007
31. Tissue-specific deletion of Foxa2 in pancreatic beta cells results in hyperinsulinemic hypoglycemia
- Author
-
Doris A. Stoffers, Siew-Lan Ang, Melissa Casey, Marko Z. Vatamaniuk, Newman J. Sund, Franz M. Matschinsky, Mark A. Magnuson, and Klaus H. Kaestner
- Subjects
medicine.medical_specialty ,Candidate gene ,Potassium Channels ,medicine.medical_treatment ,Biology ,Hypoglycemia ,medicine.disease_cause ,Islets of Langerhans ,Mice ,Adenosine Triphosphate ,Internal medicine ,Hyperinsulinism ,Insulin Secretion ,Genetics ,medicine ,Animals ,Humans ,Insulin ,Cell Lineage ,Hyperinsulinemic hypoglycemia ,Transcription factor ,Mice, Knockout ,Nuclear Proteins ,medicine.disease ,DNA-Binding Proteins ,Endocrinology ,embryonic structures ,Hepatocyte Nuclear Factor 3-beta ,FOXA2 ,Beta cell ,Developmental Biology ,Transcription Factors ,Research Paper - Abstract
We have used conditional gene ablation to uncover a dramatic and unpredicted role for the winged-helix transcription factor Foxa2 (formerly HNF-3β) in pancreatic β-cell differentiation and metabolism. Mice that lack Foxa2 specifically in β cells (Foxa2loxP/loxP; Ins.Cre mice) are severely hypoglycemic and show dysregulated insulin secretion in response to both glucose and amino acids. This inappropriate hypersecretion of insulin in the face of profound hypoglycemia mimics pathophysiological and molecular aspects of familial hyperinsulinism. We have identified the two subunits of the β-cell ATP-sensitive K+ channel (KATP), the most frequently mutated genes linked to familial hyperinsulinism, as novel Foxa2 targets in islets. TheFoxa2loxP/loxP; Ins.Cre mice will serve as a unique model to investigate the regulation of insulin secretion by the β cell and suggest the human FOXA2 as a candidate gene for familial hyperinsulinism.
- Published
- 2001
32. Okadaic acid-induced decrease in the magnitude and efficacy of the Ca2+ signal in pancreatic beta cells and inhibition of insulin secretion
- Author
-
Pascal Mariot, Philippe Detimary, Yoshihiko Sato, Jean-Claude Henquin, Patrick Gilon, UCL - MD/FSIO - Département de physiologie et pharmacologie, UCL - MD/NOPS - Département de neurologie et de psychiatrie, and UCL - (SLuc) Service de psychiatrie adulte
- Subjects
medicine.medical_specialty ,Cytoplasm ,medicine.medical_treatment ,Phosphatase ,Biology ,In Vitro Techniques ,Exocytosis ,Dephosphorylation ,chemistry.chemical_compound ,Islets of Langerhans ,Mice ,Tolbutamide ,Adenosine Triphosphate ,Ethers, Cyclic ,Internal medicine ,Insulin Secretion ,Okadaic Acid ,medicine ,Phosphoprotein Phosphatases ,Animals ,Insulin ,Enzyme Inhibitors ,Oxazoles ,Pharmacology ,Kinase ,Pancreatic islets ,Okadaic acid ,Adenosine Diphosphate ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Papers ,Calcium ,Female ,Marine Toxins ,Calcium Channels ,Beta cell ,Energy Metabolism ,medicine.drug - Abstract
1. Phosphorylation by kinases and dephosphorylation by phosphatases markedly affect the biological activity of proteins involved in stimulus-response coupling. In this study, we have characterized the effects of okadaic acid, an inhibitor of protein phosphatases 1 and 2A, on insulin secretion. Mouse pancreatic islets were preincubated for 60 min in the presence of okadaic acid before their function was studied. 2. Okadaic acid dose-dependently (IC50 approximately 200 nM) inhibited insulin secretion induced by 15 mM glucose. At 0.5 microM, okadaic acid also inhibited insulin secretion induced by tolbutamide, ketoisocaproate and high K+, and its effects were not reversed by activation of protein kinases A or C. 3. The inhibition of insulin secretion did not result from an alteration of glucose metabolism (estimated by the fluorescence of endogenous pyridine nucleotides) or a lowering of the ATP/ADP ratio in the islets. 4. Okadaic acid treatment slightly inhibited voltage-dependent Ca2+ currents in beta cells (perforated patch technique), which diminished the rise in cytoplasmic Ca2+ (fura-2 method) that glucose and high K+ produce in islets. However, this decrease (25%), was insufficient to explain the corresponding inhibition of insulin secretion (90%). Moreover, mobilization of intracellular Ca2+ by acetylcholine was barely affected by okadaic acid, whereas the concomitant insulin response was decreased by 85%. 5. Calyculin A, another inhibitor of protein phosphatases 1 and 2A largely mimicked the effects of okadaic acid, whereas 1-norokadaone, an inactive analogue of okadaic acid on phosphatases, did not alter beta cell function. 6. In conclusion, okadaic acid inhibits insulin secretion by decreasing the magnitude of the Ca2+ signal in beta cells and its efficacy on exocytosis. The results suggest that, contrary to current concepts, both phosphorylation and dephosphorylation of certain beta cell proteins may be involved in the regulation of insulin secretion.
- Published
- 1998
33. Mechanisms involved in the effect of nitric oxide synthase inhibition on L-arginine-induced insulin secretion
- Author
-
Pellegrino Masiello, M. Roye, René Gross, M. Manteghetti, Gérard Ribes, Christophe Broca, and Dominique Hillaire-Buys
- Subjects
Male ,Nitroprusside ,medicine.medical_specialty ,Arginine ,Vasodilator Agents ,medicine.medical_treatment ,L-arginine ,In Vitro Techniques ,Carbohydrate metabolism ,L-citrulline ,Nitric oxide ,Islets of Langerhans ,chemistry.chemical_compound ,Insulin secretion,glucose,L-arginine,nitric oxide synthase inhibitor,N∞-nitro-L-arginine methyl ester,L-citrulline,NG-hydroxy-L-arginine,sodium nitroprusside ,Internal medicine ,nitric oxide synthase inhibitor ,medicine ,Citrulline ,Animals ,Insulin ,Enzyme Inhibitors ,Rats, Wistar ,glucose ,Pharmacology ,sodium nitroprusside ,Arginase ,biology ,Insulin secretion ,NG-hydroxy-L-arginine ,Rats ,Nitric oxide synthase ,NG-Nitroarginine Methyl Ester ,Endocrinology ,chemistry ,N∞-nitro-L-arginine methyl ester ,Papers ,biology.protein ,Sodium nitroprusside ,Nitric Oxide Synthase ,Beta cell ,medicine.drug - Abstract
1. A constitutive nitric oxide synthase (NOSc) pathway negatively controls L-arginine-stimulated insulin release by pancreatic beta cells. We investigated the effect of glucose on this mechanism and whether it could be accounted for by nitric oxide production. 2. NOSc was inhibited by N omega-nitro-L-arginine methyl ester (L-NAME), and sodium nitroprusside (SNP) was used as a palliative NO donor to test whether the effects of L-NAME resulted from decreased NO production. 3. In the rat isolated perfused pancreas, L-NAME (5 mM) strongly potentiated L-arginine (5 mM)-induced insulin secretion at 5 mM glucose, but L-arginine and L-NAME exerted only additive effects at 8.3 mM glucose. At 11 mM glucose, L-NAME significantly inhibited L-arginine-induced insulin secretion. Similar data were obtained in rat isolated islets. 4. At high concentrations (3 and 300 microM), SNP increased the potentiation of arginine-induced insulin output by L-NAME, but not at lower concentrations (3 or 30 nM). 5. L-Arginine (5 mM) and L-ornithine (5 mM) in the presence of 5 mM glucose induced monophasic beta cell responses which were both significantly reduced by SNP at 3 nM but not at 30 nM; in contrast, the L-ornithine effect was significantly increased by SNP at 3 microM. 6. Simultaneous treatment with L-ornithine and L-arginine provoked a biphasic insulin response. 7. At 5 mM glucose, L-NAME (5 mM) did not affect the L-ornithine secretory effect, but the amino acid strongly potentiated the alteration by L-NAME of L-arginine-induced insulin secretion. 8. L-Citrulline (5 mM) significantly reduced the second phase of the insulin response to L-NAME (5 mM) + L-arginine (5 mM) and to L-NAME + L-arginine + SNP 3 microM. 9. The intermediate in NO biosynthesis, NG-hydroxy-L-arginine (150-300 microM) strongly counteracted the potentiation by L-NAME of the secretory effect of L-arginine at 5 mM glucose. 10. We conclude that the potentiation of L-arginine-induced insulin secretion resulting from the blockade of NOSc activity in the presence of a basal glucose concentration (1) is strongly modulated by higher glucose concentrations, (2) is not due to decreased NO production but (3) is probably accounted for by decreased levels of NG-hydroxy-L-arginine or L-citrulline, resulting in the attenuation of an inhibitory effect on arginase activity.
- Published
- 1997
34. A Practical Guide to Rodent Islet Isolation and Assessment Revisited.
- Author
-
Corbin, Kathryn L., West, Hannah L., Brodsky, Samantha, Whitticar, Nicholas B., Koch, William J., and Nunemaker, Craig S.
- Subjects
DIGESTIVE enzymes ,ISLANDS of Langerhans ,TYPE 1 diabetes ,TYPE 2 diabetes - Abstract
Insufficient insulin secretion is a key component of both type 1 and type 2 diabetes. Since insulin is released by the islets of Langerhans, obtaining viable and functional islets is critical for research and transplantation. The effective and efficient isolation of these small islands of endocrine cells from the sea of exocrine tissue that is the rest of the pancreas is not necessarily simple or quick. Choosing and administering the digestive enzyme, separation of the islets from acinar tissue, and culture of islets are all things that must be considered. The purpose of this review is to provide a history of the development of islet isolation procedures and to serve as a practical guide to rodent islet research for newcomers to islet biology. We discuss key elements of mouse islet isolation including choosing collagenase, the digestion process, purification of islets using a density gradient, and islet culture conditions. In addition, this paper reviews techniques for assessing islet viability and function such as visual assessment, glucose-stimulated insulin secretion and intracellular calcium measurements. A detailed protocol is provided that describes a common method our laboratory uses to obtain viable and functional mouse islets for in vitro study. This review thus provides a strong foundation for successful procurement and purification of high-quality mouse islets for research purposes. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. A Silicon Pancreatic Beta Cell for Diabetes.
- Author
-
Georgiou, P. and Toumazou, C.
- Abstract
The paper will consider how silicon devices such as ion-sensitive field effect transistors can be used to model metabolic functions in biology. In a first example, a biologically inspired silicon beta cell is presented to serve as the main building block of an artificial pancreas. This is to be used for real-time glucose sensing and insulin release for diabetics. This system presents the first silicon implementation of a metabolic cell capable of exhibiting variable bursting behavior upon glucose stimulation. Based on the Hodgkin and Huxley formalism, this approach achieves dynamics similar to that of biological beta cells by using devices biased in the subthreshold regime. In addition to mimicking the physiological behavior of the beta cell, the circuit achieves good power efficiency, measured to be 4.5 muW [ABSTRACT FROM PUBLISHER]
- Published
- 2007
- Full Text
- View/download PDF
36. Cure of acromegaly by operative removal of an islet cell tumor of the pancreas
- Author
-
Anthony S. Pagliara, Raymond V. Randall, Kalman Kovacs, R. Mario Abellera, Lynn Koob, and Robert H. Caplan
- Subjects
Adult ,medicine.medical_specialty ,Pituitary gland ,Adenoma ,Internal medicine ,Acromegaly ,medicine ,Humans ,Electrophoresis, Paper ,Hormone metabolism ,Pancreas ,Pancreatic Proteolytic Enzymes ,business.industry ,Radioimmunoassay ,General Medicine ,Glucose Tolerance Test ,Adenoma, Islet Cell ,medicine.disease ,Hormones ,Pancreatic Neoplasms ,Endocrinology ,medicine.anatomical_structure ,Growth Hormone ,Female ,Beta cell ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
We studied a 30 year old woman in whom acromegaly was cured by operative removal of a large cystic beta cell adenoma of the pancreas. We detected substantial amounts of immunoreactive human growth hormone (hGH)-like activity in a tumor tissue extract. Extracts of the tumor and a normal human pituitary gland eluted from a Sephadex G-75 column in two identical peaks. Serial dilutions of the tumor extract displaced radioactive 125I hGH parallel to a standard curve. Surprisingly, an extract of a normal human pancreas contained large amounts of hGH-like activity and gave results similar to those of the tumor extract on gel chromatography and on serial dilution displacement in the growth hormone immunoassay. Paper electrophoretic studies of 125I hGH after incubation with normal pancreatic and tumor extracts with and without enzyme inhibitors suggested that pancreatic proteolytic enzymes damaged the 125I hGH used in growth hormone radioimmunoassay and produced a false detection of hGH.
- Published
- 1978
37. Beta cell-specific PAK1 enrichment ameliorates diet-induced glucose intolerance in mice by promoting insulin biogenesis and minimising beta cell apoptosis
- Author
-
Ahn, Miwon, Dhawan, Sangeeta, McCown, Erika M., Garcia, Pablo A., Bhattacharya, Supriyo, Stein, Roland, and Thurmond, Debbie C.
- Published
- 2024
- Full Text
- View/download PDF
38. The islet tissue plasminogen activator/plasmin system is upregulated with human islet amyloid polypeptide aggregation and protects beta cells from aggregation-induced toxicity
- Author
-
Esser, Nathalie, Hogan, Meghan F., Templin, Andrew T., Akter, Rehana, Fountaine, Brendy S., Castillo, Joseph J., El-Osta, Assam, Manathunga, Lakshan, Zhyvoloup, Alexander, Raleigh, Daniel P., Zraika, Sakeneh, Hull, Rebecca L., and Kahn, Steven E.
- Published
- 2024
- Full Text
- View/download PDF
39. Relationship between pancreatic vesicular monoamine transporter 2 (VMAT2) and insulin expression in human pancreas
- Author
-
Paul L. Harris, Yoshifumi Saisho, Peter C. Butler, Tatyana Gurlo, Ryan Galasso, Alexandra E. Butler, and Robert A. Rizza
- Subjects
Male ,type 1 diabetes ,Physiology ,medicine.medical_treatment ,Cardiorespiratory Medicine and Haematology ,Vesicular monoamine transporter 2 ,Insulin-Secreting Cells ,80 and over ,Pancreatic polypeptide ,Insulin ,2.1 Biological and endogenous factors ,vesicular monoamine transporter ,Aetiology ,Cancer ,Aged, 80 and over ,Diabetes ,Type 2 diabetes ,General Medicine ,Middle Aged ,medicine.anatomical_structure ,Type 1 diabetes ,Female ,type 2 diabetes ,Beta cell ,Type 2 ,Type 1 ,Adult ,medicine.medical_specialty ,insulin ,Biochemistry & Molecular Biology ,Histology ,Tail of pancreas ,Clinical Sciences ,and over ,Biology ,Autoimmune Disease ,Pancreatic Cancer ,Rare Diseases ,Internal medicine ,medicine ,Diabetes Mellitus ,Humans ,Beta cell mass ,Pancreatic hormone ,Metabolic and endocrine ,Aged ,Original Paper ,Vesicular monoamine transporter ,beta cell mass ,Cell Biology ,medicine.disease ,Endocrinology ,Diabetes Mellitus, Type 1 ,pancreatic polypeptide ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,Vesicular Monoamine Transport Proteins ,biology.protein ,Biochemistry and Cell Biology ,Digestive Diseases - Abstract
Vesicular monoamine transporter 2 (VMAT2) is expressed in pancreatic beta cells and has recently been proposed as a target for measurement of beta cell mass in vivo. We questioned, (1) What proportion of beta cells express VMAT2? (2) Is VMAT2 expressed by other pancreatic endocrine or non-endocrine cells? (3) Is the relationship between VMAT2 and insulin expression disturbed in type 1 (T1DM) or type 2 diabetes (T2DM)? Human pancreas (7 non-diabetics, 5 T2DM, 10 T1DM) was immunostained for insulin, VMAT2 and other pancreatic hormones. Most beta cells expressed VMAT2. VMAT2 expression was not changed by the presence of diabetes. In tail of pancreas VMAT2 immunostaining closely correlated with insulin staining. However, VMAT2 was also expressed in some pancreatic polypeptide (PP) cells. Although VMAT2 was not excluded as a target for beta cell mass measurement, expression of VMAT2 in PP cells predicts residual VMAT2 expression in human pancreas even in the absence of beta cells. Electronic supplementary material The online version of this article (doi:10.1007/s10735-008-9195-9) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
40. Effect of transcription factor MEOX on insulin gene expression in glucagon-like peptide 1-secreting cells
- Author
-
Ryu, Gyeong Ryul, Bae, Dongryeoul, Uddin, Shahab, Meah, Mohammed Sohel, Ahmad, Waqas, Silvano, Kris John, Ahn, Gyeongik, Cha, Joon-Yung, Lee, Esder, Song, Ki-Ho, Kim, Woe-Yeon, and Kim, Min Gab
- Published
- 2024
- Full Text
- View/download PDF
41. Capillary contact points determine beta cell polarity, control secretion and are disrupted in the db/db mouse model of diabetes
- Author
-
Jevon, Dillon, Cottle, Louise, Hallahan, Nicole, Harwood, Richard, Samra, Jaswinder S., Gill, Anthony J., Loudovaris, Thomas, Thomas, Helen E., and Thorn, Peter
- Published
- 2024
- Full Text
- View/download PDF
42. Silencing the FABP3 gene in insulin-secreting cells reduces fatty acid uptake and protects against lipotoxicity
- Author
-
Hyder, Ayman, Sheta, Basma, Eissa, Manar, and Schrezenmeir, Jürgen
- Published
- 2024
- Full Text
- View/download PDF
43. Diabetes in remission. Insulin secretory dynamics
- Author
-
R E Gleason, Byung N Park, and J S Soeldner
- Subjects
Adult ,Blood Glucose ,Male ,medicine.medical_specialty ,Adolescent ,Chromatography, Paper ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Insulin Antibodies ,Remission, Spontaneous ,Radioimmunoassay ,Fatty Acids, Nonesterified ,Insulin resistance ,In vivo ,Internal medicine ,Diabetes mellitus ,Insulin Secretion ,Internal Medicine ,medicine ,Diabetes Mellitus ,Dietary Carbohydrates ,Animals ,Humans ,Insulin ,Child ,business.industry ,Insulin sensitivity ,Glucose Tolerance Test ,medicine.disease ,Functional recovery ,Dietary Fats ,Peripheral ,Endocrinology ,Diabetes Mellitus, Type 1 ,Injections, Intravenous ,Cattle ,Female ,Dietary Proteins ,Beta cell ,business - Abstract
Insulin secretory capacity and sensitivity to exogenous insulin were examined in eight nonobese, ketosis-prone young diabetics at the time of a clinical remission during which they could be managed without any insulin therapy. The insulin secretory capacity of these patients was assessed with oral glucose, intravenous glucose and an oral mixed meal test. The insulin responses were variable and relatively subnormal compared to those in normal controls, but definite and substantial responses were found in the majority of the patients to all the stimuli. When small amounts of crystalline insulin were administered intravenously to produce peripheral venous insulin levels in the physiologic range, these patients responded with a normal decrease in blood glucose and plasma free fatty acid levels suggesting maintenance of in vivo insulin sensitivity. Since ketosis-prone, insulin-dependent diabetics are generally characterized by nearly complete failure of beta-cell function, the present finding supports the concept that clinical remission of diabetes is dependent on a functional recovery of the beta cell. Furthermore, it seems likely that no significant “insulin resistance” that often accompanies severe diabetes is present in this remission state.
- Published
- 1974
44. Insulin Secretion in Malignant Hyperpyrexia
- Author
-
F. I. R. Martin, R. F. W. Moulds, M. A. Denborough, G. L. Warne, and P. Tse
- Subjects
Adult ,Blood Glucose ,medicine.medical_specialty ,medicine.medical_treatment ,Insulin resistance ,Internal medicine ,Insulin response ,Insulin Secretion ,Medicine ,Myocyte ,Humans ,Insulin ,In patient ,Insulin secretion ,Pancreas ,General Environmental Science ,Glucose tolerance test ,medicine.diagnostic_test ,business.industry ,Muscles ,Cell Membrane ,General Engineering ,Malignant hyperthermia ,General Medicine ,Papers and Originals ,Glucose Tolerance Test ,medicine.disease ,Insulin oscillation ,Membrane ,medicine.anatomical_structure ,Endocrinology ,General Earth and Planetary Sciences ,Calcium ,Beta cell ,Insulin Resistance ,business ,Malignant Hyperthermia - Abstract
An increased glucose-induced insulin response has been observed in patients susceptible to malignant hyperpyrexia. This raises the possibility that the membrane abnormality present in the calcium-storing membranes in the muscle cell in malignant hyperpyrexia may be present also in the beta cell of the pancreas.
- Published
- 1974
45. Insulin, Nobel laureates and The Journal of Physiology.
- Subjects
INSULIN ,NOBEL Prize winners ,NOBEL Prize in Physiology or Medicine - Published
- 2022
- Full Text
- View/download PDF
46. In vivo models of gestational and type 2 diabetes mellitus characterized by endocrine pancreas cell impairments.
- Author
-
Szlapinski, Sandra K. and Hill, David J.
- Subjects
TYPE 2 diabetes ,ENDOCRINE cells ,ISLANDS of Langerhans ,GESTATIONAL diabetes ,GLUCOSE intolerance ,HYPERGLYCEMIA - Abstract
Insulin resistance contributes to the development of various diseases, including type 2 diabetes and gestational diabetes. Even though gestational diabetes is specific to pregnancy, it can result in long-term glucose intolerance and type 2 diabetes after delivery. Given the substantial health and economic burdens associated with diabetes, it is imperative to better understand the mechanisms leading to insulin resistance and type 2 diabetes so that treatments targeted at reversing symptoms can be developed. Considering that the endocrine cells of the pancreas (islets of Langerhans) largely contribute to the pathogenesis of diabetes (beta-cell insufficiency and dysfunction), the elucidation of the various mechanisms of endocrine cell plasticity is important to understand. By better defining these mechanisms, targeted therapeutics can be developed to reverse symptoms of beta-cell deficiency and insulin resistance in diabetes. Animal models play an important role in better understanding these mechanisms, as techniques for in vivo imaging of endocrine cells in the pancreas are limited. Therefore, this review article will discuss the available rodent models of gestational and type 2 diabetes that are characterized by endocrine cell impairments in the pancreas, discuss the models with a comparison to human diabetes, and explore the potential mechanisms of endocrine cell plasticity that contribute to these phenotypes, as these mechanisms could ultimately be used to reverse blood glucose dysregulation in diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. The impact of apelin-13 on cisplatin-induced endocrine pancreas damage in rats: an in vivo study
- Author
-
Ciftel, Serpil, Tumkaya, Levent, Saral, Sinan, Mercantepe, Tolga, Akyildiz, Kerimali, Yilmaz, Adnan, and Mercantepe, Filiz
- Published
- 2024
- Full Text
- View/download PDF
48. Keeping pace: the primary cilium as the conducting baton of the islet
- Author
-
Idevall-Hagren, Olof, Incedal Nilsson, Ceren, and Sanchez, Gonzalo
- Published
- 2024
- Full Text
- View/download PDF
49. Pancreatic β-cell identity, glucose sensing and the control of insulin secretion.
- Author
-
Rutter, Guy A., Pullen, Timothy J., Hodson, David J., and Martinez-Sanchez, Aida
- Subjects
TREATMENT of diabetes ,PANCREATIC beta cells ,TYPE 1 diabetes ,GLUCOSE analysis ,GENE expression ,GENETIC transcription ,INSULIN resistance ,ELECTROPHYSIOLOGY ,GENETICS - Abstract
Insulin release from pancreatic β-cells is required to maintain normal glucose homoeostasis in man and many other animals. Defective insulin secretion underlies all forms of diabetes mellitus, a disease currently reaching epidemic proportions worldwide. Although the destruction of β-cells is responsible for Type 1 diabetes (T1D), both lowered β-cell mass and loss of secretory function are implicated in Type 2 diabetes (T2D). Emerging results suggest that a functional deficiency, involving de-differentiation of the mature β-cell towards a more progenitor-like state, may be an important driver for impaired secretion in T2D. Conversely, at least in rodents, reprogramming of islet non-β to β-cells appears to occur spontaneously in models of T1D, and may occur in man. In the present paper, we summarize the biochemical properties which define the 'identity' of the mature β-cell as a glucose sensor par excellence. In particular, we discuss the importance of suppressing a group of 11 'disallowed' housekeeping genes, including Ldha and the monocarboxylate transporter Mct1 (Slc16a1), for normal nutrient sensing. We then survey the changes in the expression and/or activity of β-cell-enriched transcription factors, including FOXO1, PDX1, NKX6.1, MAFA and RFX6, as well as non-coding RNAs, which may contribute to β-cell de-differentiation and functional impairment in T2D. The relevance of these observations for the development of new approaches to treat T1D and T2D is considered. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
50. Structure-functional changes in eNAMPT at high concentrations mediate mouse and human beta cell dysfunction in type 2 diabetes
- Author
-
Sayers, Sophie R., Beavil, Rebecca L., Fine, Nicholas H. F., Huang, Guo C., Choudhary, Pratik, Pacholarz, Kamila J., Barran, Perdita E., Butterworth, Sam, Mills, Charlotte E., Cruickshank, J. Kennedy, Silvestre, Marta P., Poppitt, Sally D., McGill, Anne-Thea, Lavery, Gareth G., Hodson, David J., and Caton, Paul W.
- Published
- 2020
- Full Text
- View/download PDF
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.