1
|
Yin W, Zou S, Sha M, Sun L, Gong H, Xiong C, Huang X, Wang J, Zhang Y, Li X, Liang J, Chang X, Wang S, Su D, Guo W, Zhang Y, Wu T, Chen F. Gain of pancreatic beta cell-specific SCD1 improves glucose homeostasis by maintaining functional beta cell mass under metabolic stress. Diabetologia 2025; 68:629-645. [PMID: 39690249 DOI: 10.1007/s00125-024-06343-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/31/2024] [Indexed: 12/19/2024]
Abstract
AIMS/HYPOTHESIS The key pancreatic beta cell transcription factor v-maf musculoaponeurotic fibrosarcoma oncogene homologue A (MafA) is critical for the maintenance of mature beta cell function and phenotype. The expression levels and/or activities of MafA are reduced when beta cells are chronically exposed to diabetogenic stress, such as hyperglycaemia (i.e. glucotoxicity). Interventional targets and adjuvant therapies to abate MafA loss in beta cells may provide evidence to support the effective treatment of diabetes. In this study, we aimed to investigate the function of stearoyl-CoA desaturase 1 (SCD1) in the stabilisation of MafA expression and activity in order to maintain functional beta cell mass, with a view to suppressing the development of type 2 diabetes. METHODS SCD1 expression levels were analysed in islets obtained from humans with type 2 diabetes, hyperglycaemic db/db mice, and a high-fat diet (HFD)-induced mouse model of diabetes. Pancreatic beta cell-specific Scd1 knockin (βSCD1KI) mice were generated to study the role of SCD1 in beta cell function and identity. The protein-to-protein interactions between SCD1 and MafA were detected in MIN6 and HEK293A cells. We used experiments including chromatin immunoprecipitation, cell-based ubiquitination assay and fatty acid composition analysis to investigate the specific molecular mechanism underlying the effect of SCD1 on the restoration of MafA and beta cell function under glucotoxic conditions. RESULTS SCD1 expression was reduced in beta cells of humans with type 2 diabetes and in HFD-fed and db/db mice compared with healthy controls, which was attributed to glucotoxicity-induced Scd1 promoter histone deacetylation. Gain-of-function of SCD1 in beta cells improved insulin deficiency, glucose intolerance and beta cell dedifferentiation/transdifferentiation in the HFD-induced mouse model of diabetes. Mechanistically, SCD1 directly bound to the E3 ubiquitin ligase HMG-CoA reductase degradation 1 (HRD1) and stabilised nuclear MafA through interrupting MafA-HRD1 interactions in mouse islets and MIN6 cells, which inhibited the ubiquitination-mediated degradation of MafA. Moreover, the products of SCD enzyme reactions (mainly oleic acid) also alleviated glucotoxicity-mediated oxidative stress in MIN6 cells. CONCLUSIONS/INTERPRETATION Our findings indicate that SCD1 stabilises beta cell MafA both in desaturase-dependent and -independent manners, thus improving glucose homeostasis under metabolic stress. This provides a potential novel target for precision medicine for the treatment of diabetes.
Collapse
Affiliation(s)
- Wenyue Yin
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Suyun Zou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Min Sha
- Department of Central Laboratory, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Taizhou, Jiangsu, China
| | - Liangjun Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haoqiang Gong
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Can Xiong
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinyue Huang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianan Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuhan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xirui Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin Liang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wanhua Guo
- Department of Nuclear Medicine, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Tijun Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
Zanfrini E, Bandral M, Jarc L, Ramirez-Torres MA, Pezzolla D, Kufrin V, Rodriguez-Aznar E, Avila AKM, Cohrs C, Speier S, Neumann K, Gavalas A. Generation and application of novel hES cell reporter lines for the differentiation and maturation of hPS cell-derived islet-like clusters. Sci Rep 2024; 14:19863. [PMID: 39191834 DOI: 10.1038/s41598-024-69645-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
The significant advances in the differentiation of human pluripotent stem (hPS) cells into pancreatic endocrine cells, including functional β-cells, have been based on a detailed understanding of the underlying developmental mechanisms. However, the final differentiation steps, leading from endocrine progenitors to mono-hormonal and mature pancreatic endocrine cells, remain to be fully understood and this is reflected in the remaining shortcomings of the hPS cell-derived islet cells (SC-islet cells), which include a lack of β-cell maturation and variability among different cell lines. Additional signals and modifications of the final differentiation steps will have to be assessed in a combinatorial manner to address the remaining issues and appropriate reporter lines would be useful in this undertaking. Here we report the generation and functional validation of hPS cell reporter lines that can monitor the generation of INS+ and GCG+ cells and their resolution into mono-hormonal cells (INSeGFP, INSeGFP/GCGmCHERRY) as well as β-cell maturation (INSeGFP/MAFAmCHERRY) and function (INSGCaMP6). The reporter hPS cell lines maintained strong and widespread expression of pluripotency markers and differentiated efficiently into definitive endoderm and pancreatic progenitor (PP) cells. PP cells from all lines differentiated efficiently into islet cell clusters that robustly expressed the corresponding reporters and contained glucose-responsive, insulin-producing cells. To demonstrate the applicability of these hPS cell reporter lines in a high-content live imaging approach for the identification of optimal differentiation conditions, we adapted our differentiation procedure to generate SC-islet clusters in microwells. This allowed the live confocal imaging of multiple SC-islets for a single condition and, using this approach, we found that the use of the N21 supplement in the last stage of the differentiation increased the number of monohormonal β-cells without affecting the number of α-cells in the SC-islets. The hPS cell reporter lines and the high-content live imaging approach described here will enable the efficient assessment of multiple conditions for the optimal differentiation and maturation of SC-islets.
Collapse
Affiliation(s)
- Elisa Zanfrini
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Manuj Bandral
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Luka Jarc
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Maria Alejandra Ramirez-Torres
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Daniela Pezzolla
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Vida Kufrin
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Eva Rodriguez-Aznar
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Ana Karen Mojica Avila
- Institute of Physiology, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Christian Cohrs
- Institute of Physiology, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Katrin Neumann
- Stem Cell Engineering Facility (SCEF), CRTD, TU Dresden, Dresden, Germany
| | - Anthony Gavalas
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany.
| |
Collapse
|
3
|
Muñoz F, Fex M, Moritz T, Mulder H, Cataldo LR. Unique features of β-cell metabolism are lost in type 2 diabetes. Acta Physiol (Oxf) 2024; 240:e14148. [PMID: 38656044 DOI: 10.1111/apha.14148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/28/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
Pancreatic β cells play an essential role in the control of systemic glucose homeostasis as they sense blood glucose levels and respond by secreting insulin. Upon stimulating glucose uptake in insulin-sensitive tissues post-prandially, this anabolic hormone restores blood glucose levels to pre-prandial levels. Maintaining physiological glucose levels thus relies on proper β-cell function. To fulfill this highly specialized nutrient sensor role, β cells have evolved a unique genetic program that shapes its distinct cellular metabolism. In this review, the unique genetic and metabolic features of β cells will be outlined, including their alterations in type 2 diabetes (T2D). β cells selectively express a set of genes in a cell type-specific manner; for instance, the glucose activating hexokinase IV enzyme or Glucokinase (GCK), whereas other genes are selectively "disallowed", including lactate dehydrogenase A (LDHA) and monocarboxylate transporter 1 (MCT1). This selective gene program equips β cells with a unique metabolic apparatus to ensure that nutrient metabolism is coupled to appropriate insulin secretion, thereby avoiding hyperglycemia, as well as life-threatening hypoglycemia. Unlike most cell types, β cells exhibit specialized bioenergetic features, including supply-driven rather than demand-driven metabolism and a high basal mitochondrial proton leak respiration. The understanding of these unique genetically programmed metabolic features and their alterations that lead to β-cell dysfunction is crucial for a comprehensive understanding of T2D pathophysiology and the development of innovative therapeutic approaches for T2D patients.
Collapse
Affiliation(s)
- Felipe Muñoz
- Clinical Research Center, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund, Sweden
| | - Malin Fex
- Clinical Research Center, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund, Sweden
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hindrik Mulder
- Clinical Research Center, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund, Sweden
| | - Luis Rodrigo Cataldo
- Clinical Research Center, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Hernández-Guadarrama A, Díaz-Román MA, Linzaga-Elizalde I, Domínguez-Mendoza BE, Aguilar-Guadarrama AB. In Silico Analysis: Anti-Inflammatory and α-Glucosidase Inhibitory Activity of New α-Methylene-γ-Lactams. Molecules 2024; 29:1973. [PMID: 38731463 PMCID: PMC11085531 DOI: 10.3390/molecules29091973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
The research about α-methylene-γ-lactams is scarce; however, their synthesis has emerged in recent years mainly because they are isosters of α-methylene-γ-lactones. This last kind of compound is structurally most common in some natural products' nuclei, like sesquiterpene lactones that show biological activity such as anti-inflammatory, anticancer, antibacterial, etc., effects. In this work, seven α-methylene-γ-lactams were evaluated by their inflammation and α-glucosidase inhibition. Thus, compounds 3-methylene-4-phenylpyrrolidin-2-one (1), 3-methylene-4-(p-tolyl)pyrrolidin-2-one (2), 4-(4-chlorophenyl)-3-methylenepyrrolidin-2-one (3), 4-(2-chlorophenyl)-3-methylenepyrrolidin-2-one (4), 5-ethyl-3-methylene-4-phenylpyrrolidin-2-one (5), 5-ethyl-3-methylene-4-(p-tolyl)pyrrolidin-2-one (6) and 4-(4-chlorophenyl)-5-ethyl-3-methylenepyrrolidin-2-one (7) were evaluated via in vitro α-glucosidase assay at 1 mM concentration. From this analysis, 7 exerts the best inhibitory effect on α-glucosidase compared with the vehicle, but it shows a low potency compared with the reference drug at the same dose. On the other side, inflammation edema was induced using TPA (12-O-tetradecanoylphorbol 13-acetate) on mouse ears; compounds 1-7 were tested at 10 µg/ear dose. As a result, 1, 3, and 5 show a better inhibition than indomethacin, at the same doses. This is a preliminary report about the biological activity of these new α-methylene-γ-lactams.
Collapse
Affiliation(s)
| | | | | | | | - A. Berenice Aguilar-Guadarrama
- Centro de Investigaciones Químicas, IICBA, Universidad Autónoma del Estado de Morelos, Avenida Universidad 1001, Col. Chamilpa, Cuernavaca 62209, Mexico; (A.H.-G.); (M.A.D.-R.); (I.L.-E.); (B.E.D.-M.)
| |
Collapse
|
5
|
Osipovich AB, Zhou FY, Chong JJ, Trinh LT, Cottam MA, Shrestha S, Cartailler JP, Magnuson MA. Deletion of Ascl1 in pancreatic β-cells improves insulin secretion, promotes parasympathetic innervation, and attenuates dedifferentiation during metabolic stress. Mol Metab 2023; 78:101811. [PMID: 37769990 PMCID: PMC10570713 DOI: 10.1016/j.molmet.2023.101811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVE ASCL1, a pioneer transcription factor, is essential for neural cell differentiation and function. Previous studies have shown that Ascl1 expression is increased in pancreatic β-cells lacking functional KATP channels or after feeding of a high fat diet (HFD) suggesting that it may contribute to the metabolic stress response of β-cells. METHODS We generated β-cell-specific Ascl1 knockout mice (Ascl1βKO) and assessed their glucose homeostasis, islet morphology and gene expression after feeding either a normal diet or HFD for 12 weeks, or in combination with a genetic disruption of Abcc8, an essential KATP channel component. RESULTS Ascl1 expression is increased in response to both a HFD and membrane depolarization and requires CREB-dependent Ca2+ signaling. No differences in glucose homeostasis or islet morphology were observed in Ascl1βKO mice fed a normal diet or in the absence of KATP channels. However, male Ascl1βKO mice fed a HFD exhibited decreased blood glucose levels, improved glucose tolerance, and increased β-cell proliferation. Bulk RNA-seq analysis of islets from Ascl1βKO mice from three studied conditions showed alterations in genes associated with the secretory function. HFD-fed Ascl1βKO mice showed the most extensive changes with increased expression of genes necessary for glucose sensing, insulin secretion and β-cell proliferation, and a decrease in genes associated with β-cell dysfunction, inflammation and dedifferentiation. HFD-fed Ascl1βKO mice also displayed increased expression of parasympathetic neural markers and cholinergic receptors that was accompanied by increased insulin secretion in response to acetylcholine and an increase in islet innervation. CONCLUSIONS Ascl1 expression is induced by stimuli that cause Ca2+-signaling to the nucleus and contributes in a multifactorial manner to the loss of β-cell function by promoting the expression of genes associated with cellular dedifferentiation, attenuating β-cells proliferation, suppressing acetylcholine sensitivity, and repressing parasympathetic innervation of islets. Thus, the removal of Ascl1 from β-cells improves their function in response to metabolic stress.
Collapse
Affiliation(s)
- Anna B Osipovich
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Frank Y Zhou
- College of Arts and Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Judy J Chong
- College of Arts and Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Linh T Trinh
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Mathew A Cottam
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Shristi Shrestha
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Mark A Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
6
|
Chen Z, Liu XA, Kenny PJ. Central and peripheral actions of nicotine that influence blood glucose homeostasis and the development of diabetes. Pharmacol Res 2023; 194:106860. [PMID: 37482325 DOI: 10.1016/j.phrs.2023.106860] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Cigarette smoking has long been recognized as a risk factor for type 2 diabetes (T2D), although the precise causal mechanisms underlying this relationship remain poorly understood. Recent evidence suggests that nicotine, the primary reinforcing component in tobacco, may play a pivotal role in connecting cigarette smoking and T2D. Extensive research conducted in both humans and animals has demonstrated that nicotine can elevate blood glucose levels, disrupt glucose homeostasis, and induce insulin resistance. The review aims to elucidate the genetic variants of nicotinic acetylcholine receptors associated with diabetes risk and provide a comprehensive overview of the available data on the mechanisms through which nicotine influences blood glucose homeostasis and the development of diabetes. Here we emphasize the central and peripheral actions of nicotine on the release of glucoregulatory hormones, as well as its effects on glucose tolerance and insulin sensitivity. Notably, the central actions of nicotine within the brain, which encompass both insulin-dependent and independent mechanisms, are highlighted as potential targets for intervention strategies in diabetes management.
Collapse
Affiliation(s)
- Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
7
|
Role of the Transcription Factor MAFA in the Maintenance of Pancreatic β-Cells. Int J Mol Sci 2022; 23:ijms23094478. [PMID: 35562869 PMCID: PMC9101179 DOI: 10.3390/ijms23094478] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/16/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
Pancreatic β-cells are specialized to properly regulate blood glucose. Maintenance of the mature β-cell phenotype is critical for glucose metabolism, and β-cell failure results in diabetes mellitus. Recent studies provide strong evidence that the mature phenotype of β-cells is maintained by several transcription factors. These factors are also required for β-cell differentiation from endocrine precursors or maturation from immature β-cells during pancreatic development. Because the reduction or loss of these factors leads to β-cell failure and diabetes, inducing the upregulation or inhibiting downregulation of these transcription factors would be beneficial for studies in both diabetes and stem cell biology. Here, we discuss one such factor, i.e., the transcription factor MAFA. MAFA is a basic leucine zipper family transcription factor that can activate the expression of insulin in β-cells with PDX1 and NEUROD1. MAFA is indeed indispensable for the maintenance of not only insulin expression but also function of adult β-cells. With loss of MAFA in type 2 diabetes, β-cells cannot maintain their mature phenotype and are dedifferentiated. In this review, we first briefly summarize the functional roles of MAFA in β-cells and then mainly focus on the molecular mechanism of cell fate conversion regulated by MAFA.
Collapse
|
8
|
Molecular Mechanism of Pancreatic β-Cell Failure in Type 2 Diabetes Mellitus. Biomedicines 2022; 10:biomedicines10040818. [PMID: 35453568 PMCID: PMC9030375 DOI: 10.3390/biomedicines10040818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023] Open
Abstract
Various important transcription factors in the pancreas are involved in the process of pancreas development, the differentiation of endocrine progenitor cells into mature insulin-producing pancreatic β-cells and the preservation of mature β-cell function. However, when β-cells are continuously exposed to a high glucose concentration for a long period of time, the expression levels of several insulin gene transcription factors are substantially suppressed, which finally leads to pancreatic β-cell failure found in type 2 diabetes mellitus. Here we show the possible underlying pathway for β-cell failure. It is likely that reduced expression levels of MafA and PDX-1 and/or incretin receptor in β-cells are closely associated with β-cell failure in type 2 diabetes mellitus. Additionally, since incretin receptor expression is reduced in the advanced stage of diabetes mellitus, incretin-based medicines show more favorable effects against β-cell failure, especially in the early stage of diabetes mellitus compared to the advanced stage. On the other hand, many subjects have recently suffered from life-threatening coronavirus infection, and coronavirus infection has brought about a new and persistent pandemic. Additionally, the spread of coronavirus infection has led to various limitations on the activities of daily life and has restricted economic development worldwide. It has been reported recently that SARS-CoV-2 directly infects β-cells through neuropilin-1, leading to apoptotic β-cell death and a reduction in insulin secretion. In this review article, we feature a possible molecular mechanism for pancreatic β-cell failure, which is often observed in type 2 diabetes mellitus. Finally, we are hopeful that coronavirus infection will decline and normal daily life will soon resume all over the world.
Collapse
|
9
|
Liang J, Chirikjian M, Pajvani UB, Bartolomé A. MafA Regulation in β-Cells: From Transcriptional to Post-Translational Mechanisms. Biomolecules 2022; 12:535. [PMID: 35454124 PMCID: PMC9033020 DOI: 10.3390/biom12040535] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
β-cells are insulin-producing cells in the pancreas that maintain euglycemic conditions. Pancreatic β-cell maturity and function are regulated by a variety of transcription factors that enable the adequate expression of the cellular machinery involved in nutrient sensing and commensurate insulin secretion. One of the key factors in this regulation is MAF bZIP transcription factor A (MafA). MafA expression is decreased in type 2 diabetes, contributing to β-cell dysfunction and disease progression. The molecular biology underlying MafA is complex, with numerous transcriptional and post-translational regulatory nodes. Understanding these complexities may uncover potential therapeutic targets to ameliorate β-cell dysfunction. This article will summarize the role of MafA in normal β-cell function and disease, with a special focus on known transcriptional and post-translational regulators of MafA expression.
Collapse
Affiliation(s)
- Jiani Liang
- Department of Medicine, Columbia University, New York, NY 10032, USA; (J.L.); (M.C.); (U.B.P.)
| | - Margot Chirikjian
- Department of Medicine, Columbia University, New York, NY 10032, USA; (J.L.); (M.C.); (U.B.P.)
| | - Utpal B. Pajvani
- Department of Medicine, Columbia University, New York, NY 10032, USA; (J.L.); (M.C.); (U.B.P.)
| | - Alberto Bartolomé
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, 28029 Madrid, Spain
| |
Collapse
|
10
|
Wu R, Karagiannopoulos A, Eliasson L, Renström E, Luan C, Zhang E. The Calcium Channel Subunit Gamma-4 as a Novel Regulator of MafA in Pancreatic Beta-Cell Controls Glucose Homeostasis. Biomedicines 2022; 10:770. [PMID: 35453520 PMCID: PMC9030882 DOI: 10.3390/biomedicines10040770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
Impaired fasting glucose (IFG) and impaired glucose tolerance (IGT) are high-risk factors of diabetes development and may be caused by defective insulin secretion in pancreatic beta-cells. Glucose-stimulated insulin secretion is mediated by voltage-gated Ca2+ (CaV) channels in which the gamma-4 subunit (CaVγ4) is required for the beta-cell to maintain its differentiated state. We here aim to explore the involvement of CaVγ4 in controlling glucose homeostasis by employing the CaVγ4-/- mice to study in vivo glucose-metabolism-related phenotypes and glucose-stimulated insulin secretion, and to investigate the underlying mechanisms. We show that CaVγ4-/- mice exhibit perturbed glucose homeostasis, including IFG and IGT. Glucose-stimulated insulin secretion is blunted in CaVγ4-/- mouse islets. Remarkably, CaVγ4 deletion results in reduced expression of the transcription factor essential for beta-cell maturation, MafA, on both mRNA and protein levels in islets from human donors and CaVγ4-/- mice, as well as in INS-1 832/13 cells. Moreover, we prove that CaMKII is responsible for mediating this regulatory pathway linked between CaVγ4 and MafA, which is further confirmed by human islet RNA-seq data. We demonstrate that CaVγ4 is a key player in preserving normal blood glucose homeostasis, which sheds light on CaVγ4 as a novel target for the treatment of prediabetes through correcting the impaired metabolic status.
Collapse
|
11
|
Cataldo LR, Singh T, Achanta K, Bsharat S, Prasad RB, Luan C, Renström E, Eliasson L, Artner I. MAFA and MAFB regulate exocytosis-related genes in human β-cells. Acta Physiol (Oxf) 2022; 234:e13761. [PMID: 34978761 DOI: 10.1111/apha.13761] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/22/2021] [Accepted: 01/01/2022] [Indexed: 12/13/2022]
Abstract
AIMS Reduced expression of exocytotic genes is associated with functional defects in insulin exocytosis contributing to impaired insulin secretion and type 2 diabetes (T2D) development. MAFA and MAFB transcription factors regulate β-cell physiology, and their gene expression is reduced in T2D β cells. We investigate if loss of MAFA and MAFB in human β cells contributes to T2D progression by regulating genes required for insulin exocytosis. METHODS Three approaches were performed: (1) RNAseq analysis with the focus on exocytosis-related genes in MafA-/- mouse islets, (2) correlational analysis between MAFA, MAFB and exocytosis-related genes in human islets and (3) MAFA and MAFB silencing in human islets and EndoC-βH1 cells followed by functional in vitro studies. RESULTS The expression of 30 exocytosis-related genes was significantly downregulated in MafA-/- mouse islets. In human islets, the expression of 29 exocytosis-related genes correlated positively with MAFA and MAFB. Eight exocytosis-related genes were downregulated in MafA-/- mouse islets and positively correlated with MAFA and MAFB in human islets. From this analysis, the expression of RAB3A, STXBP1, UNC13A, VAMP2, NAPA, NSF, STX1A and SYT7 was quantified after acute MAFA or MAFB silencing in EndoC-βH1 cells and human islets. MAFA and MAFB silencing resulted in impaired insulin secretion and reduced STX1A, SYT7 and STXBP1 (EndoC-βH1) and STX1A (human islets) mRNA expression. STX1A and STXBP1 protein expression was also impaired in islets from T2D donors which lack MAFA expression. CONCLUSION Our data indicate that STXBP1 and STX1A are important MAFA/B-regulated exocytosis genes which may contribute to insulin exocytosis defects observed in MAFA-deficient human T2D β cells.
Collapse
Affiliation(s)
- Luis Rodrigo Cataldo
- Endocrine Cell Differentiation and Function Group Stem Cell Centre Lund University Lund Sweden
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
- The Novo Nordisk Foundation Centre for Basic Metabolic Research Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Tania Singh
- Endocrine Cell Differentiation and Function Group Stem Cell Centre Lund University Lund Sweden
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
| | - Kavya Achanta
- Endocrine Cell Differentiation and Function Group Stem Cell Centre Lund University Lund Sweden
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
| | - Sara Bsharat
- Endocrine Cell Differentiation and Function Group Stem Cell Centre Lund University Lund Sweden
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
| | - Rashmi B. Prasad
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
- Department of Clinical Sciences in Malmö Malmo Sweden
| | - Cheng Luan
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
| | - Erik Renström
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
| | - Lena Eliasson
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
- Department of Clinical Sciences in Malmö Malmo Sweden
- Islet Cell Exocytosis Lund University Lund Sweden
| | - Isabella Artner
- Endocrine Cell Differentiation and Function Group Stem Cell Centre Lund University Lund Sweden
- Lund University Diabetes Centre Clinical Research Center Malmo Sweden
| |
Collapse
|
12
|
Oppenländer L, Palit S, Stemmer K, Greisle T, Sterr M, Salinno C, Bastidas-Ponce A, Feuchtinger A, Böttcher A, Ansarullah, Theis FJ, Lickert H. Vertical sleeve gastrectomy triggers fast β-cell recovery upon overt diabetes. Mol Metab 2021; 54:101330. [PMID: 34500108 PMCID: PMC8487975 DOI: 10.1016/j.molmet.2021.101330] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The effectiveness of bariatric surgery in restoring β-cell function has been described in type-2 diabetes (T2D) patients and animal models for years, whereas the mechanistic underpinnings are largely unknown. The possibility of vertical sleeve gastrectomy (VSG) to rescue far-progressed, clinically-relevant T2D and to promote β-cell recovery has not been investigated on a single-cell level. Nevertheless, characterization of the heterogeneity and functional states of β-cells after VSG is a fundamental step to understand mechanisms of glycaemic recovery and to ultimately develop alternative, less-invasive therapies. METHODS We performed VSG in late-stage diabetic db/db mice and analyzed the islet transcriptome using single-cell RNA sequencing (scRNA-seq). Immunohistochemical analyses and quantification of β-cell area and proliferation complement our findings from scRNA-seq. RESULTS We report that VSG was superior to calorie restriction in late-stage T2D and rapidly restored normoglycaemia in morbidly obese and overt diabetic db/db mice. Single-cell profiling of islets of Langerhans showed that VSG induced distinct, intrinsic changes in the β-cell transcriptome, but not in that of α-, δ-, and PP-cells. VSG triggered fast β-cell redifferentiation and functional improvement within only two weeks of intervention, which is not seen upon calorie restriction. Furthermore, VSG expanded β-cell area by means of redifferentiation and by creating a proliferation competent β-cell state. CONCLUSION Collectively, our study reveals the superiority of VSG in the remission of far-progressed T2D and presents paths of β-cell regeneration and molecular pathways underlying the glycaemic benefits of VSG.
Collapse
Affiliation(s)
- Lena Oppenländer
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Subarna Palit
- Institute of Computational Biology, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany
| | - Kerstin Stemmer
- Institute of Diabetes and Obesity, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392, Giessen, Germany
| | - Tobias Greisle
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Ciro Salinno
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology and Tissue Analytics, Helmholtz Center Munich, 85764, Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Ansarullah
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, 85764, Neuherberg, Germany; Department of Mathematics, Technical University of Munich, 85748, Garching, Germany; Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, 81675, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Department of Medicine, Technical University of Munich, 81675, Munich, Germany.
| |
Collapse
|
13
|
Jha MK, Kim JW, Kenny PJ, Chin Fatt C, Minhajuddin A, Salas R, Ely BA, Klein M, Abdallah CG, Xu J, Trivedi MH. Smoking status links habenular volume to glycated hemoglobin: Findings from the Human Connectome Project-Young Adult. Psychoneuroendocrinology 2021; 131:105321. [PMID: 34157587 DOI: 10.1016/j.psyneuen.2021.105321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND The habenula-pancreas axis regulates the stimulatory effects of nicotine on blood glucose levels and may participate in the emergence of type 2 diabetes in human tobacco smokers. This secondary analysis of young adults from the Human Connectome Project (HCP-YA) evaluated whether smoking status links the relationship between habenular volume and glycated hemoglobin (HbA1c), a marker of long-term glycemic control. METHODS Habenula segmentation was performed using a fully-automated myelin content-based approach in HCP-YA participants and the results were inspected visually (n = 693; aged 22-37 years). A linear regression analysis was used with habenular volume as the dependent variable, the smoking-by-HbA1c interaction as the independent variable of interest, and age, gender, race, ethnicity, education, income, employment status, body mass index, and total gray matter volume as covariates. RESULTS Habenula volume and HbA1c were similar in smokers and nonsmokers. There was a significant interaction effect (F(1, 673)= 5.03, p = 0.025) indicating that habenular volume was related to HbA1c in a manner that depended on smoking status. Among participants who were smokers (n = 120), higher HbA1c was associated with apparently larger habenular volume (β = 6.74, standard error=2.36, p = 0.005). No such association between habenular volume and HbA1c was noted among participants who were nonsmokers (n = 573). DISCUSSION Blood glucose levels over an extended time period, reflected by HbA1c, were correlated with habenular volume in smokers, consistent with a relationship between the habenula and blood glucose homeostasis in smokers. Future studies are needed to evaluate how habenular function relates to glycemic control in smokers and nonsmokers.
Collapse
Affiliation(s)
- Manish K Jha
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Center for Depression Research and Clinical Care, UT Southwestern Medical Center, Dallas, TX, United States
| | - Joo-Won Kim
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States; Department of Psychiatry, Baylor College of Medicine, Houston, TX, United States
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cherise Chin Fatt
- Center for Depression Research and Clinical Care, UT Southwestern Medical Center, Dallas, TX, United States
| | - Abu Minhajuddin
- Center for Depression Research and Clinical Care, UT Southwestern Medical Center, Dallas, TX, United States
| | - Ramiro Salas
- Department of Psychiatry, Baylor College of Medicine, Houston, TX, United States; Michael E DeBakey VA Medical Center, Houston, TX, United States; The Menninger Clinic, Houston, TX, United States
| | - Benjamin A Ely
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, The Bronx, NY, United States
| | - Matthew Klein
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Chadi G Abdallah
- Department of Psychiatry, Baylor College of Medicine, Houston, TX, United States; Michael E DeBakey VA Medical Center, Houston, TX, United States
| | - Junqian Xu
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States; Department of Psychiatry, Baylor College of Medicine, Houston, TX, United States
| | - Madhukar H Trivedi
- Center for Depression Research and Clinical Care, UT Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
14
|
Henquin JC. Non-glucose modulators of insulin secretion in healthy humans: (dis)similarities between islet and in vivo studies. Metabolism 2021; 122:154821. [PMID: 34174327 DOI: 10.1016/j.metabol.2021.154821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022]
Abstract
Optimal metabolic homeostasis requires precise temporal and quantitative control of insulin secretion. Both in vivo and in vitro studies have often focused on the regulation by glucose although many additional factors including other nutrients, neurotransmitters, hormones and drugs, modulate the secretory function of pancreatic β-cells. This review is based on the analysis of clinical investigations characterizing the effects of non-glucose modulators of insulin secretion in healthy subjects, and of experimental studies testing the same modulators in islets isolated from normal human donors. The aim was to determine whether the information gathered in vitro can reliably be translated to the in vivo situation. The comparison evidenced both convincing similarities and areas of discordance. The lack of coherence generally stems from the use of exceedingly high concentrations of test agents at too high or too low glucose concentrations in vitro, which casts doubts on the physiological relevance of a number of observations made in isolated islets. Future projects resorting to human islets should avoid extreme experimental conditions, such as oversized stimulations or inhibitions of β-cells, which are unlikely to throw light on normal insulin secretion and contribute to the elucidation of its defects.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
15
|
Cataldo LR, Vishnu N, Singh T, Bertonnier-Brouty L, Bsharat S, Luan C, Renström E, Prasad RB, Fex M, Mulder H, Artner I. The MafA-target gene PPP1R1A regulates GLP1R-mediated amplification of glucose-stimulated insulin secretion in β-cells. Metabolism 2021; 118:154734. [PMID: 33631146 DOI: 10.1016/j.metabol.2021.154734] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/07/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
The amplification of glucose-stimulated insulin secretion (GSIS) through incretin signaling is critical for maintaining physiological glucose levels. Incretins, like glucagon-like peptide 1 (GLP1), are a target of type 2 diabetes drugs aiming to enhance insulin secretion. Here we show that the protein phosphatase 1 inhibitor protein 1A (PPP1R1A), is expressed in β-cells and that its expression is reduced in dysfunctional β-cells lacking MafA and upon acute MafA knock down. MafA is a central regulator of GSIS and β-cell function. We observed a strong correlation of MAFA and PPP1R1A mRNA levels in human islets, moreover, PPP1R1A mRNA levels were reduced in type 2 diabetic islets and positively correlated with GLP1-mediated GSIS amplification. PPP1R1A silencing in INS1 (832/13) β-cells impaired GSIS amplification, PKA-target protein phosphorylation, mitochondrial coupling efficiency and also the expression of critical β-cell marker genes like MafA, Pdx1, NeuroD1 and Pax6. Our results demonstrate that the β-cell transcription factor MafA is required for PPP1R1A expression and that reduced β-cell PPP1R1A levels impaired β-cell function and contributed to β-cell dedifferentiation during type 2 diabetes. Loss of PPP1R1A in type 2 diabetic β-cells may explains the unresponsiveness of type 2 diabetic patients to GLP1R-based treatments.
Collapse
Affiliation(s)
- Luis Rodrigo Cataldo
- Endocrine Cell Differentiation and Function group, Stem Cell Centre, Lund University, Sweden; Lund University Diabetes Centre, Clinical Research Center, Sweden.
| | - Neelanjan Vishnu
- Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Tania Singh
- Endocrine Cell Differentiation and Function group, Stem Cell Centre, Lund University, Sweden; Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Ludivine Bertonnier-Brouty
- Endocrine Cell Differentiation and Function group, Stem Cell Centre, Lund University, Sweden; Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Sara Bsharat
- Endocrine Cell Differentiation and Function group, Stem Cell Centre, Lund University, Sweden; Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Cheng Luan
- Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Erik Renström
- Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Rashmi B Prasad
- Lund University Diabetes Centre, Clinical Research Center, Sweden; Department of Clinical Sciences in Malmö, Sweden
| | - Malin Fex
- Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Hindrik Mulder
- Lund University Diabetes Centre, Clinical Research Center, Sweden
| | - Isabella Artner
- Endocrine Cell Differentiation and Function group, Stem Cell Centre, Lund University, Sweden; Lund University Diabetes Centre, Clinical Research Center, Sweden.
| |
Collapse
|
16
|
Targosova K, Kucera M, Kilianova Z, Slobodova L, Szmicsekova K, Hrabovska A. Cardiac nicotinic receptors show β-subunit-dependent compensatory changes. Am J Physiol Heart Circ Physiol 2021; 320:H1975-H1984. [PMID: 33769917 DOI: 10.1152/ajpheart.00995.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nicotinic receptors (NRs) play an important role in the cholinergic regulation of heart functions, and converging evidence suggests a diverse repertoire of NR subunits in the heart. A recent hypothesis about the plasticity of β NR subunits suggests that β2-subunits and β4-subunits may substitute for each other. In our study, we assessed the hypothetical β-subunit interchangeability in the heart at the level of mRNA. Using two mutant mice strains lacking β2 or β4 NR subunits, we examined the relative expression of NR subunits and other key cholinergic molecules. We investigated the physiology of isolated hearts perfused by Langendorff's method at basal conditions and after cholinergic and/or adrenergic stimulation. Lack of β2 NR subunit was accompanied with decreased relative expression of β4-subunits and α3-subunits. No other cholinergic changes were observed at the level of mRNA, except for increased M3 and decreased M4 muscarinic receptors. Isolated hearts lacking β2 NR subunit showed different dynamics in heart rate response to indirect cholinergic stimulation. In hearts lacking β4 NR subunit, increased levels of β2-subunits were observed together with decreased mRNA for acetylcholine-synthetizing enzyme and M1 and M4 muscarinic receptors. Changes in the expression levels in β4-/- hearts were associated with increased basal heart rate and impaired response to a high dose of acetylcholine upon adrenergic stimulation. In support of the proposed plasticity of cardiac NRs, our results confirmed subunit-dependent compensatory changes to missing cardiac NRs subunits with consequences on isolated heart physiology.NEW & NOTEWORTHY In the present study, we observed an increase in mRNA levels of the β2 NR subunit in β4-/- hearts but not vice versa, thus supporting the hypothesis of β NR subunit plasticity that depends on the specific type of missing β-subunit. This was accompanied with specific cholinergic adaptations. Nevertheless, isolated hearts of β4-/- mice showed increased basal heart rate and a higher sensitivity to a high dose of acetylcholine upon adrenergic stimulation.
Collapse
Affiliation(s)
- Katarina Targosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Matej Kucera
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Zuzana Kilianova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia.,Department of Pharmacology, Slovak Medical University in Bratislava, Bratislava, Slovakia
| | - Lubica Slobodova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia.,Department of Pharmacology, Slovak Medical University in Bratislava, Bratislava, Slovakia
| | - Kristina Szmicsekova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Anna Hrabovska
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia.,Department of Pharmacology, Slovak Medical University in Bratislava, Bratislava, Slovakia.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
17
|
Postnatal maturation of calcium signaling in islets of Langerhans from neonatal mice. Cell Calcium 2020; 94:102339. [PMID: 33422769 DOI: 10.1016/j.ceca.2020.102339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/02/2023]
Abstract
Pancreatic islet cells develop mature physiological responses to glucose and other fuels postnatally. In this study, we used fluorescence imaging techniques to measure changes in intracellular calcium ([Ca2+]i) to compare islets isolated from mice on postnatal days 0, 4, and 12 with islets from adult CD-1 mice. In addition, we used publicly available RNA-sequencing data to compare expression levels of key genes in β-cell physiology with [Ca2+]i data across these ages. We show that islets isolated from mice on postnatal day 0 displayed elevated [Ca2+]i in basal glucose (≤4 mM) but lower [Ca2+]i responses to stimulation by 12-20 mM glucose compared to adult. Neonatal islets displayed more adult-like [Ca2+]i in basal glucose by day 4 but continued to show lower [Ca2+]i responses to 16 and 20 mM glucose stimulation up to at least day 12. A right shift in glucose sensing (EC50) correlated with lower fragment-per-kilobase-of-transcript-per-million-reads-mapped (FPKM) of Slc2a2 (glut2) and Actn3 and increased FPKM for Galk1 and Nupr1. Differences in [Ca2+]i responses to additional stimuli were also observed. Calcium levels in the endoplasmic reticulum were elevated on day 0 but became adult-like by day 4, which corresponded with reduced expression in Atp2a2 (SERCA2) and novel K+-channel Ktd17, increased expression of Pml, Wfs1, Thada, and Herpud1, and basal [Ca2+]i maturing to adult levels. Ion-channel activity also matured rapidly, but RNA sequencing data mining did not yield strong leads. In conclusion, the maturation of islet [Ca2+]i signaling is complex and multifaceted; several possible gene targets were identified that may participate in this process.
Collapse
|
18
|
Marselli L, Piron A, Suleiman M, Colli ML, Yi X, Khamis A, Carrat GR, Rutter GA, Bugliani M, Giusti L, Ronci M, Ibberson M, Turatsinze JV, Boggi U, De Simone P, De Tata V, Lopes M, Nasteska D, De Luca C, Tesi M, Bosi E, Singh P, Campani D, Schulte AM, Solimena M, Hecht P, Rady B, Bakaj I, Pocai A, Norquay L, Thorens B, Canouil M, Froguel P, Eizirik DL, Cnop M, Marchetti P. Persistent or Transient Human β Cell Dysfunction Induced by Metabolic Stress: Specific Signatures and Shared Gene Expression with Type 2 Diabetes. Cell Rep 2020; 33:108466. [PMID: 33264613 DOI: 10.1016/j.celrep.2020.108466] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/06/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic β cell failure is key to type 2 diabetes (T2D) onset and progression. Here, we assess whether human β cell dysfunction induced by metabolic stress is reversible, evaluate the molecular pathways underlying persistent or transient damage, and explore the relationships with T2D islet traits. Twenty-six islet preparations are exposed to several lipotoxic/glucotoxic conditions, some of which impair insulin release, depending on stressor type, concentration, and combination. The reversal of dysfunction occurs after washout for some, although not all, of the lipoglucotoxic insults. Islet transcriptomes assessed by RNA sequencing and expression quantitative trait loci (eQTL) analysis identify specific pathways underlying β cell failure and recovery. Comparison of a large number of human T2D islet transcriptomes with those of persistent or reversible β cell lipoglucotoxicity show shared gene expression signatures. The identification of mechanisms associated with human β cell dysfunction and recovery and their overlap with T2D islet traits provide insights into T2D pathogenesis, fostering the development of improved β cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Lorella Marselli
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy.
| | - Anthony Piron
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Maikel L Colli
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Amna Khamis
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille University Hospital, Lille 59000, France
| | - Gaelle R Carrat
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College, London, UK; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Laura Giusti
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy; School of Pharmacy, University of Camerino, Camerino, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy; Centre for Advanced Studies and Technologies (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Mark Ibberson
- Vital-IT Group, Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | | | - Ugo Boggi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy; Division of General and Transplant Surgery, Cisanello University Hospital, Pisa 56124, Italy
| | - Paolo De Simone
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy; Division of Liver Surgery and Transplantation, Cisanello University Hospital, Pisa 56124, Italy
| | - Vincenzo De Tata
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy
| | - Miguel Lopes
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Daniela Nasteska
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Carmela De Luca
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Marta Tesi
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Emanuele Bosi
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Pratibha Singh
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Daniela Campani
- Department of Surgical, Medical and Molecular Pathology and the Critical Areas, University of Pisa, Pisa 56126, Italy
| | - Anke M Schulte
- Sanofi-Aventis Deutschland GmbH, Diabetes Research, Frankfurt, Germany
| | - Michele Solimena
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden 01307, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg 85764, Germany
| | - Peter Hecht
- Sanofi-Aventis Deutschland GmbH, Diabetes Research, Frankfurt, Germany
| | | | | | | | | | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Mickaël Canouil
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille University Hospital, Lille 59000, France
| | - Philippe Froguel
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium; WELBIO, Université Libre de Bruxelles, Brussels, Belgium; Indiana Biosciences Research Institute, Indianapolis, IN, USA; Division of Endocrinology, ULB Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels 1070, Belgium; Division of Endocrinology, ULB Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy.
| |
Collapse
|
19
|
Xie H, Yepuri N, Meng Q, Dhawan R, Leech CA, Chepurny OG, Holz GG, Cooney RN. Therapeutic potential of α7 nicotinic acetylcholine receptor agonists to combat obesity, diabetes, and inflammation. Rev Endocr Metab Disord 2020; 21:431-447. [PMID: 32851581 PMCID: PMC7572644 DOI: 10.1007/s11154-020-09584-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
The cholinergic anti-inflammatory reflex (CAIR) represents an important homeostatic regulatory mechanism for sensing and controlling the body's response to inflammatory stimuli. Vagovagal reflexes are an integral component of CAIR whose anti-inflammatory effects are mediated by acetylcholine (ACh) acting at α7 nicotinic acetylcholine receptors (α7nAChR) located on cells of the immune system. Recently, it is appreciated that CAIR and α7nAChR also participate in the control of metabolic homeostasis. This has led to the understanding that defective vagovagal reflex circuitry underlying CAIR might explain the coexistence of obesity, diabetes, and inflammation in the metabolic syndrome. Thus, there is renewed interest in the α7nAChR that mediates CAIR, particularly from the standpoint of therapeutics. Of special note is the recent finding that α7nAChR agonist GTS-21 acts at L-cells of the distal intestine to stimulate the release of two glucoregulatory and anorexigenic hormones: glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). Furthermore, α7nAChR agonist PNU 282987 exerts trophic factor-like actions to support pancreatic β-cell survival under conditions of stress resembling diabetes. This review provides an overview of α7nAChR function as it pertains to CAIR, vagovagal reflexes, and metabolic homeostasis. We also consider the possible usefulness of α7nAChR agonists for treatment of obesity, diabetes, and inflammation.
Collapse
Affiliation(s)
- Han Xie
- Departments of Surgery, State University of New York (SUNY), Upstate Medical University, 750 E Adams St., Suite 8141, Syracuse, NY, 13210, USA
| | - Natesh Yepuri
- Departments of Surgery, State University of New York (SUNY), Upstate Medical University, 750 E Adams St., Suite 8141, Syracuse, NY, 13210, USA
| | - Qinghe Meng
- Departments of Surgery, State University of New York (SUNY), Upstate Medical University, 750 E Adams St., Suite 8141, Syracuse, NY, 13210, USA
| | - Ravi Dhawan
- Departments of Surgery, State University of New York (SUNY), Upstate Medical University, 750 E Adams St., Suite 8141, Syracuse, NY, 13210, USA
| | - Colin A Leech
- Departments of Surgery, State University of New York (SUNY), Upstate Medical University, 750 E Adams St., Suite 8141, Syracuse, NY, 13210, USA
| | - Oleg G Chepurny
- Departments of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, USA
| | - George G Holz
- Departments of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, USA
| | - Robert N Cooney
- Departments of Surgery, State University of New York (SUNY), Upstate Medical University, 750 E Adams St., Suite 8141, Syracuse, NY, 13210, USA.
| |
Collapse
|
20
|
Seoane-Collazo P, Diéguez C, Nogueiras R, Rahmouni K, Fernández-Real JM, López M. Nicotine' actions on energy balance: Friend or foe? Pharmacol Ther 2020; 219:107693. [PMID: 32987056 DOI: 10.1016/j.pharmthera.2020.107693] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
Obesity has reached pandemic proportions and is associated with severe comorbidities, such as type 2 diabetes mellitus, hepatic and cardiovascular diseases, and certain cancer types. However, the therapeutic options to treat obesity are limited. Extensive epidemiological studies have shown a strong relationship between smoking and body weight, with non-smokers weighing more than smokers at any age. Increased body weight after smoking cessation is a major factor that interferes with their attempts to quit smoking. Numerous controlled studies in both humans and rodents have reported that nicotine, the main bioactive component of tobacco, exerts a marked anorectic action. Furthermore, nicotine is also known to modulate energy expenditure, by regulating the thermogenic activity of brown adipose tissue (BAT) and the browning of white adipose tissue (WAT), as well as glucose homeostasis. Many of these actions occur at central level, by controlling the activity of hypothalamic neuropeptide systems such as proopiomelanocortin (POMC), or energy sensors such as AMP-activated protein kinase (AMPK). However, direct impact of nicotine on metabolic tissues, such as BAT, WAT, liver and pancreas has also been described. Here, we review the actions of nicotine on energy balance. The relevance of this interaction is interesting, because considering the restricted efficiency of obesity treatments, a possible complementary approach may focus on compounds with known pharmacokinetic profile and pharmacological actions, such as nicotine or nicotinic acetylcholine receptors signaling.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine and Veterans Affairs Health Care System, Iowa City, IA 52242, USA
| | - José Manuel Fernández-Real
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain; Department of Diabetes, Endocrinology and Nutrition (UDEN), Hospital of Girona "Dr Josep Trueta" and Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| |
Collapse
|
21
|
The Constitutive Lack of α7 Nicotinic Receptor Leads to Metabolic Disorders in Mouse. Biomolecules 2020; 10:biom10071057. [PMID: 32708537 PMCID: PMC7408520 DOI: 10.3390/biom10071057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 01/21/2023] Open
Abstract
Objective: Type 2 diabetes (T2D) occurs by deterioration in pancreatic β-cell function and/or progressive loss of pancreatic β-cell mass under the context of insulin resistance. α7 nicotinic acetylcholine receptor (nAChR) may contribute to insulin sensitivity but its role in the pathogenesis of T2D remains undefined. We investigated whether the systemic lack of α7 nAChR was sufficient to impair glucose homeostasis. Methods: We used an α7 nAChR knock-out (α7−/−) mouse model fed a standard chow diet. The effects of the lack of α7 nAChR on islet mass, insulin secretion, glucose and insulin tolerance, body composition, and food behaviour were assessed in vivo and ex vivo experiments. Results: Young α7−/− mice display a chronic mild high glycemia combined with an impaired glucose tolerance and a marked deficit in β-cell mass. In addition to these metabolic disorders, old mice developed adipose tissue inflammation, elevated plasma free fatty acid concentrations and presented glycolytic muscle insulin resistance in old mice. Finally, α7−/− mice, fed a chow diet, exhibited a late-onset excessive gain in body weight through increased fat mass associated with higher food intake. Conclusion: Our work highlights the important role of α7 nAChR in glucose homeostasis. The constitutive lack of α7 nAChR suggests a novel pathway influencing the pathogenesis of T2D.
Collapse
|
22
|
Singh T, Colberg JK, Sarmiento L, Chaves P, Hansen L, Bsharat S, Cataldo LR, Dudenhöffer-Pfeifer M, Fex M, Bryder D, Holmberg D, Sitnicka E, Cilio C, Prasad RB, Artner I. Loss of MafA and MafB expression promotes islet inflammation. Sci Rep 2019; 9:9074. [PMID: 31235823 PMCID: PMC6591483 DOI: 10.1038/s41598-019-45528-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
Maf transcription factors are critical regulators of beta-cell function. We have previously shown that reduced MafA expression in human and mouse islets is associated with a pro-inflammatory gene signature. Here, we investigate if the loss of Maf transcription factors induced autoimmune processes in the pancreas. Transcriptomics analysis showed expression of pro-inflammatory as well as immune cell marker genes. However, clusters of CD4+ T and B220+ B cells were associated primarily with adult MafA−/−MafB+/−, but not MafA−/− islets. MafA expression was detected in the thymus, lymph nodes and bone marrow suggesting a novel role of MafA in regulating immune-cell function. Analysis of pancreatic lymph node cells showed activation of CD4+ T cells, but lack of CD8+ T cell activation which also coincided with an enrichment of naïve CD8+ T cells. Further analysis of T cell marker genes revealed a reduction of T cell receptor signaling gene expression in CD8, but not in CD4+ T cells, which was accompanied with a defect in early T cell receptor signaling in mutant CD8+ T cells. These results suggest that loss of MafA impairs both beta- and T cell function affecting the balance of peripheral immune responses against islet autoantigens, resulting in local inflammation in pancreatic islets.
Collapse
Affiliation(s)
- Tania Singh
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden.,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Jesper K Colberg
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Luis Sarmiento
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Patricia Chaves
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Lisbeth Hansen
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Sara Bsharat
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden.,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Luis R Cataldo
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden.,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | | | - Malin Fex
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - David Bryder
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Dan Holmberg
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Ewa Sitnicka
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Corrado Cilio
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Rashmi B Prasad
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Isabella Artner
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden. .,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden.
| |
Collapse
|
23
|
Fernández-Cabezudo MJ, George JA, Bashir G, Mohamed YA, Al-Mansori A, Qureshi MM, Lorke DE, Petroianu G, Al-Ramadi BK. Involvement of Acetylcholine Receptors in Cholinergic Pathway-Mediated Protection Against Autoimmune Diabetes. Front Immunol 2019; 10:1038. [PMID: 31156627 PMCID: PMC6529936 DOI: 10.3389/fimmu.2019.01038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Type I diabetes (T1D) is a T cell-driven autoimmune disease that results in the killing of pancreatic β-cells and, consequently, loss of insulin production. Using the multiple low-dose streptozotocin (MLD-STZ) model of experimental autoimmune diabetes, we previously reported that pretreatment with a specific acetylcholinesterase inhibitor (AChEI), paraoxon, prevented the development of hyperglycemia in C57BL/6 mice. This correlated with an inhibition of T cell infiltration into the pancreatic islets and a reduction in pro-inflammatory cytokines. The cholinergic anti-inflammatory pathway utilizes nicotinic and muscarinic acetylcholine receptors (nAChRs and mAChRs, respectively) expressed on a variety of cell types. In this study, we carried out a comparative analysis of the effect of specific antagonists of nAChRs or mAChRs on the development of autoimmune diabetes. Co-administration of mecamylamine, a non-selective antagonist of nAChRs maintained the protective effect of AChEI on the development of hyperglycemia. In contrast, co-administration of atropine, a non-selective antagonist of mAChRs, mitigated AChEI-mediated protection. Mice pretreated with mecamylamine had an improved response in glucose tolerance test (GTT) than mice pretreated with atropine. These differential effects of nAChR and mAChR antagonists correlated with the extent of islet cell infiltration and with the structure and functionality of the β-cells. Taken together, our data suggest that mAChRs are essential for the protective effect of cholinergic stimulation in autoimmune diabetes.
Collapse
Affiliation(s)
- Maria J Fernández-Cabezudo
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Junu A George
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Ghada Bashir
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Yassir A Mohamed
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Alreem Al-Mansori
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Mohammed M Qureshi
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Dietrich E Lorke
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Georg Petroianu
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Basel K Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| |
Collapse
|
24
|
Luan C, Ye Y, Singh T, Barghouth M, Eliasson L, Artner I, Zhang E, Renström E. The calcium channel subunit gamma-4 is regulated by MafA and necessary for pancreatic beta-cell specification. Commun Biol 2019; 2:106. [PMID: 30911681 PMCID: PMC6420573 DOI: 10.1038/s42003-019-0351-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 02/08/2019] [Indexed: 12/22/2022] Open
Abstract
Voltage-gated Ca2+ (CaV) channels trigger glucose-induced insulin secretion in pancreatic beta-cell and their dysfunction increases diabetes risk. These heteromeric complexes include the main subunit alpha1, and the accessory ones, including subunit gamma that remains unexplored. Here, we demonstrate that CaV gamma subunit 4 (CaVγ4) is downregulated in islets from human donors with diabetes, diabetic Goto-Kakizaki (GK) rats, as well as under conditions of gluco-/lipotoxic stress. Reduction of CaVγ4 expression results in decreased expression of L-type CaV1.2 and CaV1.3, thereby suppressing voltage-gated Ca2+ entry and glucose stimulated insulin exocytosis. The most important finding is that CaVγ4 expression is controlled by the transcription factor responsible for beta-cell specification, MafA, as verified by chromatin immunoprecipitation and experiments in beta-cell specific MafA knockout mice (MafA Δβcell ). Taken together, these findings suggest that CaVγ4 is necessary for maintaining a functional differentiated beta-cell phenotype. Treatment aiming at restoring CaVγ4 may help to restore beta-cell function in diabetes.
Collapse
Affiliation(s)
- Cheng Luan
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, 202 13 Malmö, Sweden
| | - Yingying Ye
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, 202 13 Malmö, Sweden
| | - Tania Singh
- Stem Cell Center, Department of Laboratory Medicine, Lund University, 221 85 Lund, Sweden
| | - Mohammad Barghouth
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, 202 13 Malmö, Sweden
| | - Lena Eliasson
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, 202 13 Malmö, Sweden
| | - Isabella Artner
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, 202 13 Malmö, Sweden
- Stem Cell Center, Department of Laboratory Medicine, Lund University, 221 85 Lund, Sweden
| | - Enming Zhang
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, 202 13 Malmö, Sweden
| | - Erik Renström
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, 202 13 Malmö, Sweden
| |
Collapse
|
25
|
Cataldo Bascuñan LR, Lyons C, Bennet H, Artner I, Fex M. Serotonergic regulation of insulin secretion. Acta Physiol (Oxf) 2019; 225:e13101. [PMID: 29791774 DOI: 10.1111/apha.13101] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 05/14/2018] [Accepted: 05/17/2018] [Indexed: 12/13/2022]
Abstract
The exact physiological role for the monoamine serotonin (5-HT) in modulation of insulin secretion is yet to be fully understood. Although the presence of this monoamine in islets of Langerhans is well established, it is only with recent advances that the complex signalling network in islets involving 5-HT is being unravelled. With more than fourteen different 5-HT receptors expressed in human islets and receptor-independent mechanisms in insulin-producing β-cells, our understanding of 5-HT's regulation of insulin secretion is increasing. It is now widely accepted that failure of the pancreatic β-cell to release sufficient amounts of insulin is the main cause of type 2 diabetes (T2D), an ongoing global epidemic. In this context, 5-HT signalling may be of importance. In fact, 5-HT may serve an essential role in regulating the release of insulin and glucagon, the two main hormones that control glucose and lipid homoeostasis. In this review, we will discuss past and current understanding of 5-HT's role in the endocrine pancreas.
Collapse
Affiliation(s)
- L. R. Cataldo Bascuñan
- Endocrine Cell Differentiation and Function Group; Stem Cell Centre; Lund University; Lund Sweden
| | - C. Lyons
- Department of Clinical Sciences in Malmö; Unit of Molecular Metabolism; Lund University Diabetes Centre; Lund University; Malmö Sweden
- Clinical Research Center; Lund University; Malmö Sweden
- Malmö University Hospital; Lund University; Malmö Sweden
| | - H. Bennet
- Department of Clinical Sciences in Malmö; Unit of Molecular Metabolism; Lund University Diabetes Centre; Lund University; Malmö Sweden
- Clinical Research Center; Lund University; Malmö Sweden
- Malmö University Hospital; Lund University; Malmö Sweden
| | - I. Artner
- Endocrine Cell Differentiation and Function Group; Stem Cell Centre; Lund University; Lund Sweden
| | - M. Fex
- Department of Clinical Sciences in Malmö; Unit of Molecular Metabolism; Lund University Diabetes Centre; Lund University; Malmö Sweden
- Clinical Research Center; Lund University; Malmö Sweden
- Malmö University Hospital; Lund University; Malmö Sweden
| |
Collapse
|
26
|
Singh T, Sarmiento L, Luan C, Prasad RB, Johansson J, Cataldo LR, Renström E, Soneji S, Cilio C, Artner I. MafA Expression Preserves Immune Homeostasis in Human and Mouse Islets. Genes (Basel) 2018; 9:genes9120644. [PMID: 30567413 PMCID: PMC6315686 DOI: 10.3390/genes9120644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023] Open
Abstract
Type 1 (T1D) and type 2 (T2D) diabetes are triggered by a combination of environmental and/or genetic factors. Maf transcription factors regulate pancreatic beta (β)-cell function, and have also been implicated in the regulation of immunomodulatory cytokines like interferon-β (IFNβ1). In this study, we assessed MAFA and MAFB co-expression with pro-inflammatory cytokine signaling genes in RNA-seq data from human pancreatic islets. Interestingly, MAFA expression was strongly negatively correlated with cytokine-induced signaling (such as IFNAR1, DDX58) and T1D susceptibility genes (IFIH1), whereas correlation of these genes with MAFB was weaker. In order to evaluate if the loss of MafA altered the immune status of islets, MafA deficient mouse islets (MafA−/−) were assessed for inherent anti-viral response and susceptibility to enterovirus infection. MafA deficient mouse islets had elevated basal levels of Ifnβ1, Rig1 (DDX58 in humans), and Mda5 (IFIH1) which resulted in reduced virus propagation in response to coxsackievirus B3 (CVB3) infection. Moreover, an acute knockdown of MafA in β-cell lines also enhanced Rig1 and Mda5 protein levels. Our results suggest that precise regulation of MAFA levels is critical for islet cell-specific cytokine production, which is a critical parameter for the inflammatory status of pancreatic islets.
Collapse
Affiliation(s)
- Tania Singh
- Stem Cell Center, Lund University, 22184, Lund, Sweden.
| | | | - Cheng Luan
- Lund University Diabetes Center, 22184, Lund, Sweden.
| | | | | | | | - Erik Renström
- Lund University Diabetes Center, 22184, Lund, Sweden.
| | - Shamit Soneji
- Stem Cell Center, Lund University, 22184, Lund, Sweden.
| | - Corrado Cilio
- Lund University Diabetes Center, 22184, Lund, Sweden.
| | - Isabella Artner
- Stem Cell Center, Lund University, 22184, Lund, Sweden.
- Lund University Diabetes Center, 22184, Lund, Sweden.
| |
Collapse
|
27
|
Gupta D, Lacayo AA, Greene SM, Leahy JL, Jetton TL. β-Cell mass restoration by α7 nicotinic acetylcholine receptor activation. J Biol Chem 2018; 293:20295-20306. [PMID: 30397183 DOI: 10.1074/jbc.ra118.004617] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Although it is well-established how nutrients, growth factors, and hormones impact functional β-cell mass (BCM), the influence of the central nervous system in this regard, and especially in the context of islet immune modulation, has been understudied. Here we investigated the expression and activity of pancreatic islet α7 nicotinic acetylcholine receptor (α7nAChR) in islet anti-inflammatory and prosurvival signaling. Systemic administration of α7nAChR agonists in mice improved glucose tolerance and curtailed streptozotocin-induced hyperglycemia by retaining BCM, in part through maintaining Pdx1 and MafA expression and reducing apoptosis. α7nAChR activation of mouse islets ex vivo led to reduced inflammatory drive through a JAK2-STAT3 pathway that couples with CREB/Irs2/Akt survival signaling. Because the vagus nerve conveys anti-inflammatory signals to immune cells of the spleen and other nonneural tissues in the viscera by activating α7nAChR agonists, our study suggests a novel role for β-cell α7nAChR that functions to maintain β-cell survival and mass homeostasis through modulating islet cytokine and phosphatidylinositol 3-kinase-dependent signaling pathways. Exploiting these pathways may have therapeutic potential for the treatment of autoimmune diabetes.
Collapse
Affiliation(s)
- Dhananjay Gupta
- From the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05446
| | - Adam A Lacayo
- From the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05446
| | - Shane M Greene
- From the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05446
| | - John L Leahy
- From the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05446
| | - Thomas L Jetton
- From the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05446.
| |
Collapse
|
28
|
Rorsman P, Ashcroft FM. Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev 2018; 98:117-214. [PMID: 29212789 PMCID: PMC5866358 DOI: 10.1152/physrev.00008.2017] [Citation(s) in RCA: 517] [Impact Index Per Article: 73.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
The pancreatic β-cell plays a key role in glucose homeostasis by secreting insulin, the only hormone capable of lowering the blood glucose concentration. Impaired insulin secretion results in the chronic hyperglycemia that characterizes type 2 diabetes (T2DM), which currently afflicts >450 million people worldwide. The healthy β-cell acts as a glucose sensor matching its output to the circulating glucose concentration. It does so via metabolically induced changes in electrical activity, which culminate in an increase in the cytoplasmic Ca2+ concentration and initiation of Ca2+-dependent exocytosis of insulin-containing secretory granules. Here, we review recent advances in our understanding of the β-cell transcriptome, electrical activity, and insulin exocytosis. We highlight salient differences between mouse and human β-cells, provide models of how the different ion channels contribute to their electrical activity and insulin secretion, and conclude by discussing how these processes become perturbed in T2DM.
Collapse
Affiliation(s)
- Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Frances M Ashcroft
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Eliasson L, Esguerra JLS, Wendt A. Lessons from basic pancreatic beta cell research in type-2 diabetes and vascular complications. Diabetol Int 2017; 8:139-152. [PMID: 30603317 DOI: 10.1007/s13340-017-0304-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/08/2017] [Indexed: 12/14/2022]
Abstract
The changes in life-style with increased access of food and reduced physical activity have resulted in the global epidemic of obesity. Consequently, individuals with type 2 diabetes and cardiovascular disease have also escalated. A central organ in the development of diabetes is the pancreas, and more specifically the pancreatic beta cells within the islets of Langerhans. Beta cells have been assigned the important task of secreting insulin when blood glucose is increased to lower the glucose level. An early sign of diabetes pathogenesis is lack of first phase insulin response and reduced second phase secretion. In this review, which is based on the foreign investigator award lecture given at the JSDC meeting in Sendai in October 2016, we discuss a possible cellular explanation for the reduced first phase insulin response and how this can be influenced by lipids. Moreover, since patients with cardiovascular disease and high levels of cholesterol are often treated with statins, we summarize recent data regarding effects on statins on glucose homeostasis and insulin secretion. Finally, we suggest microRNAs (miRNAs) as central players in the adjustment of beta cell function during the development of diabetes. We specifically discuss miRNAs regarding their involvement in insulin secretion regulation, differential expression in type 2 diabetes, and potential as biomarkers for prediction of diabetes and cardiovascular complications.
Collapse
Affiliation(s)
- Lena Eliasson
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| |
Collapse
|
30
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|