1
|
Wu WB, Gao F, Tang YH, Wang HZ, Dong H, Lu FE, Yuan F. Huanglian-Renshen-Decoction Maintains Islet β-Cell Identity in T2DM Mice through Regulating GLP-1 and GLP-1R in Both Islet and Intestine. Chin J Integr Med 2025; 31:39-48. [PMID: 39551849 DOI: 10.1007/s11655-024-3915-1] [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] [Accepted: 08/08/2023] [Indexed: 11/19/2024]
Abstract
OBJECTIVE To elucidate the effect of Huanglian-Renshen-Decoction (HRD) on ameliorating type 2 diabetes mellitus by maintaining islet β -cell identity through regulating paracrine and endocrine glucagon-like peptide-1 (GLP-1)/GLP-1 receptor (GLP-1R) in both islet and intestine. METHODS The db/db mice were divided into the model (distilled water), low-dose HRD (LHRD, 3 g/kg), high-dose HRD (HHRD, 6 g/kg), and liraglutide (400 µ g/kg) groups using a random number table, 8 mice in each group. The db/m mice were used as the control group (n=8, distilled water). The entire treatment of mice lasted for 6 weeks. Blood insulin, glucose, and GLP-1 levels were quantified using enzyme-linked immunosorbent assay kits. The proliferation and apoptosis factors of islet cells were determined by immunohistochemistry (IHC) and immunofluorescence (IF) staining. Then, GLP-1, GLP-1R, prohormone convertase 1/3 (PC1/3), PC2, v-maf musculoaponeurotic fibrosarcoma oncogene homologue A (MafA), and pancreatic and duodenal homeobox 1 (PDX1) were detected by Western blot, IHC, IF, and real-time quantitative polymerase chain reaction, respectively. RESULTS HRD reduced the weight and blood glucose of the db/db mice, and improved insulin sensitivity at the same time (P<0.05 or P<0.01). HRD also promoted mice to secrete more insulin and less glucagon (P<0.05 or P<0.01). Moreover, it also increased the number of islet β cell and decreased islet α cell mass (P<0.01). After HRD treatment, the levels of GLP-1, GLP-1R, PC1/3, PC2, MafA, and PDX1 in the pancreas and intestine significantly increased (P<0.05 or P<0.01). CONCLUSION HRD can maintain the normal function and identity of islet β cell, and the underlying mechanism is related to promoting the paracrine and endocrine activation of GLP-1 in pancreas and intestine.
Collapse
Affiliation(s)
- Wen-Bin Wu
- Institution of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fan Gao
- Institution of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue-Heng Tang
- Institution of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong-Zhan Wang
- Institution of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fu-Er Lu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fen Yuan
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Rutter GA, Gresch A, Delgadillo Silva L, Benninger RKP. Exploring pancreatic beta-cell subgroups and their connectivity. Nat Metab 2024; 6:2039-2053. [PMID: 39117960 DOI: 10.1038/s42255-024-01097-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024]
Abstract
Functional pancreatic islet beta cells are essential to ensure glucose homeostasis across species from zebrafish to humans. These cells show significant heterogeneity, and emerging studies have revealed that connectivity across a hierarchical network is required for normal insulin release. Here, we discuss current thinking and areas of debate around intra-islet connectivity, cellular hierarchies and potential "controlling" beta-cell populations. We focus on methodologies, including comparisons of different cell preparations as well as in vitro and in vivo approaches to imaging and controlling the activity of human and rodent islet preparations. We also discuss the analytical approaches that can be applied to live-cell data to identify and study critical subgroups of cells with a disproportionate role in control Ca2+ dynamics and thus insulin secretion (such as "first responders", "leaders" and "hubs", as defined by Ca2+ responses to glucose stimulation). Possible mechanisms by which this hierarchy is achieved, its physiological relevance and how its loss may contribute to islet failure in diabetes mellitus are also considered. A glossary of terms and links to computational resources are provided.
Collapse
Affiliation(s)
- Guy A Rutter
- CHUM Research Center and Faculty of Medicine, University of Montréal, Montréal, QC, Canada.
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Anne Gresch
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luis Delgadillo Silva
- CHUM Research Center and Faculty of Medicine, University of Montréal, Montréal, QC, Canada
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
3
|
Yu V, Yong F, Marta A, Khadayate S, Osakwe A, Bhattacharya S, Varghese SS, Chabosseau P, Tabibi SM, Chen K, Georgiadou E, Parveen N, Suleiman M, Stamoulis Z, Marselli L, De Luca C, Tesi M, Ostinelli G, Delgadillo-Silva L, Wu X, Hatanaka Y, Montoya A, Elliott J, Patel B, Demchenko N, Whilding C, Hajkova P, Shliaha P, Kramer H, Ali Y, Marchetti P, Sladek R, Dhawan S, Withers DJ, Rutter GA, Millership SJ. Differential CpG methylation at Nnat in the early establishment of beta cell heterogeneity. Diabetologia 2024; 67:1079-1094. [PMID: 38512414 PMCID: PMC11058053 DOI: 10.1007/s00125-024-06123-6] [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: 11/15/2023] [Accepted: 01/09/2024] [Indexed: 03/23/2024]
Abstract
AIMS/HYPOTHESIS Beta cells within the pancreatic islet represent a heterogenous population wherein individual sub-groups of cells make distinct contributions to the overall control of insulin secretion. These include a subpopulation of highly connected 'hub' cells, important for the propagation of intercellular Ca2+ waves. Functional subpopulations have also been demonstrated in human beta cells, with an altered subtype distribution apparent in type 2 diabetes. At present, the molecular mechanisms through which beta cell hierarchy is established are poorly understood. Changes at the level of the epigenome provide one such possibility, which we explore here by focusing on the imprinted gene Nnat (encoding neuronatin [NNAT]), which is required for normal insulin synthesis and secretion. METHODS Single-cell RNA-seq datasets were examined using Seurat 4.0 and ClusterProfiler running under R. Transgenic mice expressing enhanced GFP under the control of the Nnat enhancer/promoter regions were generated for FACS of beta cells and downstream analysis of CpG methylation by bisulphite sequencing and RNA-seq, respectively. Animals deleted for the de novo methyltransferase DNA methyltransferase 3 alpha (DNMT3A) from the pancreatic progenitor stage were used to explore control of promoter methylation. Proteomics was performed using affinity purification mass spectrometry and Ca2+ dynamics explored by rapid confocal imaging of Cal-520 AM and Cal-590 AM. Insulin secretion was measured using homogeneous time-resolved fluorescence imaging. RESULTS Nnat mRNA was differentially expressed in a discrete beta cell population in a developmental stage- and DNA methylation (DNMT3A)-dependent manner. Thus, pseudo-time analysis of embryonic datasets demonstrated the early establishment of Nnat-positive and -negative subpopulations during embryogenesis. NNAT expression is also restricted to a subset of beta cells across the human islet that is maintained throughout adult life. NNAT+ beta cells also displayed a discrete transcriptome at adult stages, representing a subpopulation specialised for insulin production, and were diminished in db/db mice. 'Hub' cells were less abundant in the NNAT+ population, consistent with epigenetic control of this functional specialisation. CONCLUSIONS/INTERPRETATION These findings demonstrate that differential DNA methylation at Nnat represents a novel means through which beta cell heterogeneity is established during development. We therefore hypothesise that changes in methylation at this locus may contribute to a loss of beta cell hierarchy and connectivity, potentially contributing to defective insulin secretion in some forms of diabetes. DATA AVAILABILITY The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD048465.
Collapse
Affiliation(s)
- Vanessa Yu
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Fiona Yong
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore
| | - Angellica Marta
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | | | - Adrien Osakwe
- Quantitative Life Sciences Program, McGill University, Montréal, QC, Canada
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Sneha S Varghese
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Pauline Chabosseau
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Sayed M Tabibi
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Keran Chen
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Eleni Georgiadou
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Nazia Parveen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa, Italy
| | - Zoe Stamoulis
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Medical Sciences Division, University of Oxford, Oxford, UK
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa, Italy
| | - Carmela De Luca
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa, Italy
| | - Marta Tesi
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa, Italy
| | - Giada Ostinelli
- CHUM Research Center and Faculty of Medicine, University of Montréal, Montréal, QC, Canada
| | - Luis Delgadillo-Silva
- CHUM Research Center and Faculty of Medicine, University of Montréal, Montréal, QC, Canada
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Yuki Hatanaka
- MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | | | | | | | - Nikita Demchenko
- MRC Laboratory of Medical Sciences, London, UK
- Imaging Resource Facility, Research Operations, St George's, University of London, London, UK
| | | | - Petra Hajkova
- MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | | | | | - Yusuf Ali
- Nutrition, Metabolism and Health Programme & Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Republic of Singapore
- Singapore Eye Research Institute (SERI), Singapore General Hospital, Singapore, Republic of Singapore
- Clinical Research Unit, Khoo Teck Puat Hospital, National Healthcare Group, Singapore, Republic of Singapore
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa, Italy
| | - Robert Sladek
- Quantitative Life Sciences Program, McGill University, Montréal, QC, Canada
- Departments of Medicine and Human Genetics, McGill University, Montréal, QC, Canada
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Dominic J Withers
- MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Guy A Rutter
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore.
- CHUM Research Center and Faculty of Medicine, University of Montréal, Montréal, QC, Canada.
| | - Steven J Millership
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
4
|
Cuozzo F, Viloria K, Shilleh AH, Nasteska D, Frazer-Morris C, Tong J, Jiao Z, Boufersaoui A, Marzullo B, Rosoff DB, Smith HR, Bonner C, Kerr-Conte J, Pattou F, Nano R, Piemonti L, Johnson PRV, Spiers R, Roberts J, Lavery GG, Clark A, Ceresa CDL, Ray DW, Hodson L, Davies AP, Rutter GA, Oshima M, Scharfmann R, Merrins MJ, Akerman I, Tennant DA, Ludwig C, Hodson DJ. LDHB contributes to the regulation of lactate levels and basal insulin secretion in human pancreatic β cells. Cell Rep 2024; 43:114047. [PMID: 38607916 PMCID: PMC11164428 DOI: 10.1016/j.celrep.2024.114047] [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: 03/16/2023] [Revised: 02/19/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Using 13C6 glucose labeling coupled to gas chromatography-mass spectrometry and 2D 1H-13C heteronuclear single quantum coherence NMR spectroscopy, we have obtained a comparative high-resolution map of glucose fate underpinning β cell function. In both mouse and human islets, the contribution of glucose to the tricarboxylic acid (TCA) cycle is similar. Pyruvate fueling of the TCA cycle is primarily mediated by the activity of pyruvate dehydrogenase, with lower flux through pyruvate carboxylase. While the conversion of pyruvate to lactate by lactate dehydrogenase (LDH) can be detected in islets of both species, lactate accumulation is 6-fold higher in human islets. Human islets express LDH, with low-moderate LDHA expression and β cell-specific LDHB expression. LDHB inhibition amplifies LDHA-dependent lactate generation in mouse and human β cells and increases basal insulin release. Lastly, cis-instrument Mendelian randomization shows that low LDHB expression levels correlate with elevated fasting insulin in humans. Thus, LDHB limits lactate generation in β cells to maintain appropriate insulin release.
Collapse
Affiliation(s)
- Federica Cuozzo
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Katrina Viloria
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ali H Shilleh
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Charlotte Frazer-Morris
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jason Tong
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Zicong Jiao
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Geneplus-Beijing, Changping District, Beijing 102206, China
| | - Adam Boufersaoui
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Bryan Marzullo
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Daniel B Rosoff
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Hannah R Smith
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Caroline Bonner
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire de Lille (CHU Lille), Institute Pasteur Lille, U1190 -European Genomic Institute for Diabetes (EGID), F59000 Lille, France
| | - Julie Kerr-Conte
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire de Lille (CHU Lille), Institute Pasteur Lille, U1190 -European Genomic Institute for Diabetes (EGID), F59000 Lille, France
| | - Francois Pattou
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire de Lille (CHU Lille), Institute Pasteur Lille, U1190 -European Genomic Institute for Diabetes (EGID), F59000 Lille, France
| | - Rita Nano
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Paul R V Johnson
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Rebecca Spiers
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jennie Roberts
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Centre for Systems Health and Integrated Metabolic Research (SHiMR), Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Carlo D L Ceresa
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - David W Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Amy P Davies
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; CHUM Research Centre and Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Masaya Oshima
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Raphaël Scharfmann
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Matthew J Merrins
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Ildem Akerman
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK.
| | - Christian Ludwig
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK.
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
Qin Y, Chen J, Qian D, Li Z, Zhang L, Ma Q. Excessive Tryptophan and Phenylalanine Induced Pancreatic Injury and Glycometabolism Disorder in Grower-finisher Pigs. J Nutr 2024; 154:1333-1346. [PMID: 38582698 DOI: 10.1016/j.tjnut.2024.01.019] [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: 11/10/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND The increase in circulating insulin levels is associated with the onset of type 2 diabetes (T2D), and the levels of branched-chain amino acids and aromatic amino acids (AAAs) are altered in T2D, but whether AAAs play a role in insulin secretion and signaling remains unclear. OBJECTIVES This study aimed to investigate the effects of different AAAs on pancreatic function and on the use of insulin in finishing pigs. METHODS A total of 18 healthy finishing pigs (Large White) with average body weight of 100 ± 1.15 kg were randomly allocated to 3 dietary treatments: Con, a normal diet supplemented with 0.68% alanine; Phe, a normal diet supplemented with 1.26% phenylalanine; and Trp, a normal diet supplemented with 0.78% tryptophan. The 3 diets were isonitrogenous. There were 6 replicates in each group. RESULTS Herein, we investigated the effects of tryptophan and phenylalanine on pancreatic function and the use of insulin in finishing pigs and found that the addition of tryptophan and phenylalanine aggravated pancreatic fat deposition, increased the relative content of saturated fatty acids, especially palmitate (C16:0) and stearate (C18:0), and the resulting lipid toxicity disrupted pancreatic secretory function. We also found that tryptophan and phenylalanine inhibited the growth and secretion of β-cells, downregulated the gene expression of the PI3K/Akt pathway in the pancreas and liver, and reduced glucose utilization in the liver. CONCLUSIONS Using fattening pigs as a model, multiorgan combined analysis of the insulin-secreting organ pancreas and the main insulin-acting organ liver, excessive intake of tryptophan and phenylalanine will aggravate pancreatic damage leading to glucose metabolism disorders, providing new evidence for the occurrence and development of T2D.
Collapse
Affiliation(s)
- Yingjie Qin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Jiayi Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Dali Qian
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Zhongyu Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
6
|
Peercy BE, Hodson DJ. Synchronizing beta cells in the pancreas. eLife 2024; 13:e95103. [PMID: 38270512 PMCID: PMC10810605 DOI: 10.7554/elife.95103] [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] [Indexed: 01/26/2024] Open
Abstract
The secretion of insulin from the pancreas relies on both gap junctions and subpopulations of beta cells with specific intrinsic properties.
Collapse
Affiliation(s)
- Bradford E Peercy
- Department of Mathematics and Statistics, University of Maryland Baltimore County (UMBC)BaltimoreUnited States
| | - David J Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
7
|
Yöntem FD, Ayaz S, Bulut Ş, Aldoğan EH, Ahbab MA. Endoplasmic reticulum stress and pro-inflammatory responses induced by phthalate metabolites monoethylhexyl phthalate and monobutyl phthalate in 1.1B4 pancreatic beta cells. Toxicology 2024; 501:153695. [PMID: 38048874 DOI: 10.1016/j.tox.2023.153695] [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: 10/06/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023]
Abstract
In recent years, phthalates and their metabolites have been associated with metabolic diseases such as diabetes mellitus. To investigate the effects of phthalate metabolites exposure on insulin production and release, 1.1B4 pancreatic beta cells were treated with different concentrations (0.001-1000 µM) of monoethylhexyl phthalate (MEHP) and monobutyl phthalate (MBP). For such purpose, the 1.1B4 cells were evaluated for their viability, apoptosis rate, lysosomal membrane permeabilization (LMP), mitochondrial membrane potential (ΔΨm), oxidative stress, ER stress status, in addition to their secretory functions. MEHP, not MBP, exhibited a notable reduction in metabolic viability, particularly at higher concentrations (500 and 1000 µM) following 24-hour exposure. Similarly, both MEHP and MBP induced decreased metabolic viability at high concentrations after 48- and 72-hour exposure. Notably, neither MEHP nor MBP demonstrated a significant impact on apoptosis rates after 24-hour exposure, and MBP induced mild necrosis at 1000 µM concentration. Cell proliferation rates, indicated by PCNA expression, decreased with 10 and 1000 µM MEHP and 0.1 and 10 µM MBP exposures. LMP analysis revealed an increase in 1000 µM MBP group. Exposure to 0.001 µM of both MEHP and MBP significantly reduced cellular glutathione (GSH) levels. No significant change in intracellular reactive oxygen species (ROS) levels and ΔΨm was observed, but MBP-exposed cells exhibited elevated levels of lipid peroxidation. Functional assessments of pancreatic beta cells unveiled reduced insulin secretion at low glucose concentrations following exposure to both MEHP and MBP, with concurrent alterations in the expression levels of key proteins associated with beta cell function, including GLUT1, GCK, PDX1, and MafA. Moreover, MEHP and MBP exposures were associated with alterations in ER stress-related pathways, including JNK, GADD153, and NF-κB expression, as well as PPARα and PPARγ levels. In conclusion, this study provides comprehensive insights into the diverse impacts of MEHP and MBP on 1.1B4 pancreatic beta cells, emphasizing their potential role in modulating cell survival, metabolic function, and stress response pathways.
Collapse
Affiliation(s)
- Fulya Dal Yöntem
- Koç University, Faculty of Medicine, Department of Biophysics, Istanbul, Turkey; Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Sinem Ayaz
- Istanbul University, Cerrahpasa, Institute of Graduate Studies, Department of Clinical Microbiology, Istanbul, Turkey; Haliç University, Faculty of Medicine, Department of Clinical Microbiology, Istanbul, Turkey
| | - Şeyma Bulut
- Bezmialem Vakif University, Faculty of Medicine, Department of Medical Biology, Istanbul, Turkey; Bezmialem Vakıf University, Institute of Health Sciences, Department of Biotechnology, Istanbul, Turkey
| | | | - Müfide Aydoğan Ahbab
- University of Health Sciences Türkiye, Hamidiye Vocational School of Health Services, Istanbul, Turkey.
| |
Collapse
|
8
|
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
|
9
|
Yu V, Yong F, Marta A, Khadayate S, Osakwe A, Bhattacharya S, Varghese SS, Chabosseau P, Tabibi SM, Chen K, Georgiadou E, Parveen N, Suleiman M, Stamoulis Z, Marselli L, De Luca C, Tesi M, Ostinelli G, Delgadillo-Silva L, Wu X, Hatanaka Y, Montoya A, Elliott J, Patel B, Demchenko N, Whilding C, Hajkova P, Shliaha P, Kramer H, Ali Y, Marchetti P, Sladek R, Dhawan S, Withers DJ, Rutter GA, Millership SJ. Differential CpG methylation at Nnat in the early establishment of beta cell heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.04.527050. [PMID: 38076935 PMCID: PMC10705251 DOI: 10.1101/2023.02.04.527050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Aims/hypothesis Beta cells within the pancreatic islet represent a heterogenous population wherein individual sub-groups of cells make distinct contributions to the overall control of insulin secretion. These include a subpopulation of highly-connected 'hub' cells, important for the propagation of intercellular Ca2+ waves. Functional subpopulations have also been demonstrated in human beta cells, with an altered subtype distribution apparent in type 2 diabetes. At present, the molecular mechanisms through which beta cell hierarchy is established are poorly understood. Changes at the level of the epigenome provide one such possibility which we explore here by focussing on the imprinted gene neuronatin (Nnat), which is required for normal insulin synthesis and secretion. Methods Single cell RNA-seq datasets were examined using Seurat 4.0 and ClusterProfiler running under R. Transgenic mice expressing eGFP under the control of the Nnat enhancer/promoter regions were generated for fluorescence-activated cell (FAC) sorting of beta cells and downstream analysis of CpG methylation by bisulphite and RNA sequencing, respectively. Animals deleted for the de novo methyltransferase, DNMT3A from the pancreatic progenitor stage were used to explore control of promoter methylation. Proteomics was performed using affinity purification mass spectrometry and Ca2+ dynamics explored by rapid confocal imaging of Cal-520 and Cal-590. Insulin secretion was measured using Homogeneous Time Resolved Fluorescence Imaging. Results Nnat mRNA was differentially expressed in a discrete beta cell population in a developmental stage- and DNA methylation (DNMT3A)-dependent manner. Thus, pseudo-time analysis of embryonic data sets demonstrated the early establishment of Nnat-positive and negative subpopulations during embryogenesis. NNAT expression is also restricted to a subset of beta cells across the human islet that is maintained throughout adult life. NNAT+ beta cells also displayed a discrete transcriptome at adult stages, representing a sub-population specialised for insulin production, reminiscent of recently-described "βHI" cells and were diminished in db/db mice. 'Hub' cells were less abundant in the NNAT+ population, consistent with epigenetic control of this functional specialization. Conclusions/interpretation These findings demonstrate that differential DNA methylation at Nnat represents a novel means through which beta cell heterogeneity is established during development. We therefore hypothesise that changes in methylation at this locus may thus contribute to a loss of beta cell hierarchy and connectivity, potentially contributing to defective insulin secretion in some forms of diabetes.
Collapse
Affiliation(s)
- Vanessa Yu
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Fiona Yong
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637553, Singapore
| | - Angellica Marta
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Sanjay Khadayate
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Adrien Osakwe
- Departments of Medicine, Human Genetics and Quantitative Life Sciences, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Sneha S. Varghese
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Pauline Chabosseau
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Sayed M. Tabibi
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Keran Chen
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Eleni Georgiadou
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Nazia Parveen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Zoe Stamoulis
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - 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
| | - Giada Ostinelli
- CHUM Research Center and Faculty of Medicine, University of Montréal, 900 Rue St Denis, Montréal, H2X OA9, QC, Canada
| | - Luis Delgadillo-Silva
- CHUM Research Center and Faculty of Medicine, University of Montréal, 900 Rue St Denis, Montréal, H2X OA9, QC, Canada
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Yuki Hatanaka
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Alex Montoya
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - James Elliott
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Bhavik Patel
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Nikita Demchenko
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Chad Whilding
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Petra Hajkova
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Pavel Shliaha
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Holger Kramer
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Yusuf Ali
- Nutrition, Metabolism and Health Programme & Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, 308232
- Singapore Eye Research Institute (SERI), Singapore General Hospital, Singapore, 168751
- Clinical Research Unit, Khoo Teck Puat Hospital, National Healthcare Group, Singapore, 768828
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Robert Sladek
- Departments of Medicine, Human Genetics and Quantitative Life Sciences, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Dominic J. Withers
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Guy A. Rutter
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637553, Singapore
- CHUM Research Center and Faculty of Medicine, University of Montréal, 900 Rue St Denis, Montréal, H2X OA9, QC, Canada
| | - Steven J. Millership
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
10
|
Abidov M, Sokolova K, Danilova I, Baykenova M, Gette I, Mychlynina E, Aydin Ozgur B, Gurol AO, Yilmaz MT. Hepatic insulin synthesis increases in rat models of diabetes mellitus type 1 and 2 differently. PLoS One 2023; 18:e0294432. [PMID: 38019818 PMCID: PMC10686419 DOI: 10.1371/journal.pone.0294432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
Insulin-positive (+) cells (IPCs), detected in multiple organs, are of great interest as a probable alternative to ameliorate pancreatic beta-cells dysfunction and insulin deficiency in diabetes. Liver is a potential source of IPCs due to it common embryological origin with pancreas. We previously demonstrated the presence of IPCs in the liver of healthy and diabetic rats, but detailed description and analysis of the factors, which potentially can induced ectopic hepatic expression of insulin in type 1 (T1D) and type 2 diabetes (T2D), were not performed. In present study we evaluate mass of hepatic IPCs in the rat models of T1D and T2D and discuss factors, which may stimulate it generation: glycaemia, organ injury, involving of hepatic stem/progenitor cell compartment, expression of transcription factors and inflammation. Quantity of IPCs in the liver was up by 1.7-fold in rats with T1D and 10-fold in T2D compared to non-diabetic (ND) rats. We concluded that ectopic hepatic expression of insulin gene is activated by combined action of a number of factors, with inflammation playing a decision role.
Collapse
Affiliation(s)
- Musa Abidov
- Institute of Immunopathology and Preventive Medicine, Ljubljana, Slovenia
| | - Ksenia Sokolova
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Irina Danilova
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Madina Baykenova
- Kostanay Oblast Tuberculosis Dispensary, Kostanay, Republic of Kazakhstan
| | - Irina Gette
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Elena Mychlynina
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Burcin Aydin Ozgur
- Department of Medical Biology and Genetics, Faculty of Medicine, Demiroglu Bilim University, Istanbul, Turkey
- Diabetes Application and Research Center, Demiroglu Bilim University, Istanbul, Turkey
| | - Ali Osman Gurol
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
- Diabetes Application and Research Center, Istanbul University, Istanbul, Turkey
| | - M. Temel Yilmaz
- International Diabetes Center, Acibadem University, Istanbul, Turkey
| |
Collapse
|
11
|
Šterk M, Barać U, Stožer A, Gosak M. Both electrical and metabolic coupling shape the collective multimodal activity and functional connectivity patterns in beta cell collectives: A computational model perspective. Phys Rev E 2023; 108:054409. [PMID: 38115462 DOI: 10.1103/physreve.108.054409] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/20/2023] [Indexed: 12/21/2023]
Abstract
Pancreatic beta cells are coupled excitable oscillators that synchronize their activity via different communication pathways. Their oscillatory activity manifests itself on multiple timescales and consists of bursting electrical activity, subsequent oscillations in the intracellular Ca^{2+}, as well as oscillations in metabolism and exocytosis. The coordination of the intricate activity on the multicellular level plays a key role in the regulation of physiological pulsatile insulin secretion and is incompletely understood. In this paper, we investigate theoretically the principles that give rise to the synchronized activity of beta cell populations by building up a phenomenological multicellular model that incorporates the basic features of beta cell dynamics. Specifically, the model is composed of coupled slow and fast oscillatory units that reflect metabolic processes and electrical activity, respectively. Using a realistic description of the intercellular interactions, we study how the combination of electrical and metabolic coupling generates collective rhythmicity and shapes functional beta cell networks. It turns out that while electrical coupling solely can synchronize the responses, the addition of metabolic interactions further enhances coordination, the spatial range of interactions increases the number of connections in the functional beta cell networks, and ensures a better consistency with experimental findings. Moreover, our computational results provide additional insights into the relationship between beta cell heterogeneity, their activity profiles, and functional connectivity, supplementing thereby recent experimental results on endocrine networks.
Collapse
Affiliation(s)
- Marko Šterk
- Department of Physics, Faculty of Natural Sciences and Mathematics, Koroška cesta 160, University of Maribor, 2000 Maribor, Slovenia
- Institute of Physiology, Faculty of Medicine, Taborska ulica 8, University of Maribor, 2000 Maribor, Slovenia
- Alma Mater Europaea, Slovenska ulica 17, 2000 Maribor, Slovenia
| | - Uroš Barać
- Department of Physics, Faculty of Natural Sciences and Mathematics, Koroška cesta 160, University of Maribor, 2000 Maribor, Slovenia
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, Taborska ulica 8, University of Maribor, 2000 Maribor, Slovenia
| | - Marko Gosak
- Department of Physics, Faculty of Natural Sciences and Mathematics, Koroška cesta 160, University of Maribor, 2000 Maribor, Slovenia
- Institute of Physiology, Faculty of Medicine, Taborska ulica 8, University of Maribor, 2000 Maribor, Slovenia
- Alma Mater Europaea, Slovenska ulica 17, 2000 Maribor, Slovenia
| |
Collapse
|
12
|
Li Y, Li R, Luo X, Xu F, Yang M, Zheng L, Wu Q, Jiang W, Li Y. Vascular endothelial growth factor B regulates insulin secretion in β cells of type 2 diabetes mellitus mice via PLCγ and the IP3R‑evoked Ca2 +/CaMK2 signaling pathway. Mol Med Rep 2023; 28:197. [PMID: 37681454 PMCID: PMC10510031 DOI: 10.3892/mmr.2023.13084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Vascular endothelial growth factor B (VEGFB) plays a crucial role in glucolipid metabolism and is highly associated with type 2 diabetes mellitus (T2DM). The role of VEGFB in the insulin secretion of β cells remains unverified. Thus, the present study aimed to discuss the effect of VEGFB on regulating insulin secretion in T2DM development, and its underlying mechanism. A high‑fat diet and streptozocin (STZ) were used for inducing T2DM in mice model, and VEGFB gene in islet cells of T2DM mice was knocked out by CRISPR Cas9 and overexpressed by adeno‑Associated Virus (AAV) injection. The effect of VEGFB and its underlying mechanism was assessed by light microscopy, electron microscopy and fluorescence confocal microscopy, enzyme‑linked immunosorbent assay, mass spectrometer and western blot analysis. The decrement of insulin secretion in islet β cell of T2DM mice were aggravated and blood glucose remained at a high level after VEGFB knockout (KO). However, glucose tolerance and insulin sensitivity of T2DM mice were improved after the AAV‑VEGFB186 injection. VEGFB KO or overexpression can inhibit or activate PLCγ/IP3R in a VEGFR1‑dependent manner. Then, the change of PLCγ/IP3R caused by VEGFB/VEGFR1 will alter the expression of key factors on the Ca2+/CaMK2 signaling pathway such as PPP3CA. Moreover, VEGFB can cause altered insulin secretion by changing the calcium concentration in β cells of T2DM mice. These findings indicated that VEGFB activated the Ca2+/CaMK2 pathway via VEGFR1‑PLCγ and IP3R pathway to regulate insulin secretion, which provides new insight into the regulatory mechanism of abnormal insulin secretion in T2DM.
Collapse
Affiliation(s)
- Yuqi Li
- Department of Pathophysiology, School of Basic Medicine of Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Rongrong Li
- Department of Pathophysiology, School of Basic Medicine of Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Xu Luo
- Department of Pathophysiology, School of Basic Medicine of Binzhou Medical University, Yantai, Shandong 264000, P.R. China
- Department of Laboratory, Guiyang Centers for Disease Control and Prevention, Guiyang, Guizhou 550000, P.R. China
| | - Fang Xu
- Department of Pathophysiology, School of Basic Medicine of Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Meizi Yang
- Department of Pharmacology, School of Basic Medicine of Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Lanhui Zheng
- The First School of Clinical Medicine, Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Qihao Wu
- The First School of Clinical Medicine, Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Wenguo Jiang
- Department of Pharmacy, Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Yana Li
- Department of Pathophysiology, School of Basic Medicine of Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
13
|
Kaneto H, Obata A, Shimoda M, Kimura T, Obata Y, Ikeda T, Moriuchi S, Nakanishi S, Mune T, Kaku K. Comprehensive Search for GPCR Compounds which Can Enhance MafA and/or PDX-1 Expression Levels Using a Small Molecule Compound Library. J Diabetes Res 2023; 2023:8803172. [PMID: 37720599 PMCID: PMC10504048 DOI: 10.1155/2023/8803172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/24/2023] [Accepted: 08/31/2023] [Indexed: 09/19/2023] Open
Abstract
It has been shown that chronic hyperglycemia gradually decreases insulin biosynthesis and secretion which is accompanied by reduced expression of very important insulin gene transcription factors MafA and PDX-1. Such phenomena are well known as β-cell glucose toxicity. It has been shown that the downregulation of MafA and/or PDX-1 expression considerably explains the molecular mechanism for glucose toxicity. However, it remained unknown which molecules can enhance MafA and/or PDX-1 expression levels. In this study, we comprehensively searched for G protein-coupled receptor (GPCR) compounds which can enhance MafA and/or PDX-1 expression levels using a small molecule compound library in pancreatic β-cell line MIN6 cells and islets isolated from nondiabetic C57BL/6 J mice and obese type 2 diabetic C57BL/KsJ-db/db mice. We found that fulvestrant and dexmedetomidine hydrochloride increased MafA, PDX-1, or insulin expression levels in MIN6 cells. We confirmed that fulvestrant and dexmedetomidine hydrochloride increased MafA, PDX-1, or insulin expression levels in islets from nondiabetic mice as well. Furthermore, these reagents more clearly enhanced MafA, PDX-1, or insulin expression levels in islets from obese type 2 diabetic db/db mice in which MafA and PDX-1 expression levels are reduced due to glucose toxicity. In conclusion, fulvestrant and dexmedetomidine hydrochloride increased MafA, PDX-1, or insulin expression levels in MIN6 cells and islets from nondiabetic mice and obese type 2 diabetic db/db mice. To the best of our knowledge, this is the first report showing some molecule which can enhance MafA and/or PDX-1 expression levels. Therefore, although further extensive study is necessary, we think that the information in this study could be, at least in part, useful at some point such as in the development of new antidiabetes medicine based on the molecular mechanism of β-cell glucose toxicity in the future.
Collapse
Affiliation(s)
- Hideaki Kaneto
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Atsushi Obata
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Masashi Shimoda
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Tomohiko Kimura
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Yoshiyuki Obata
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Tomoko Ikeda
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Saeko Moriuchi
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Shuhei Nakanishi
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Tomoatsu Mune
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, Japan
| | - Kohei Kaku
- Kawasaki Medical School General Medical Center, Japan
| |
Collapse
|
14
|
Liu Z, Hu W, Qin Y, Sun L, Jing L, Lu M, Li Y, Qu J, Yang Z. Isl1 promotes gene transcription through physical interaction with Set1/Mll complexes. Eur J Cell Biol 2023; 102:151295. [PMID: 36758343 DOI: 10.1016/j.ejcb.2023.151295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Histone H3 lysine 4 (H3K4) methylation is generally recognized as a prominent marker of gene activation. While Set1/Mll complexes are major methyltransferases that are responsible for H3K4 methylation, the mechanism of how these complexes are recruited into the target gene promotor is still unclear. Here, starting with an affinity purification-mass spectrometry approach, we have found that Isl1, a highly tissue-specific expressed LIM/homeodomain transcription factor, is physically associated with Set1/Mll complexes. We then show that Wdr5 directly binds to Isl1. And this binding is likely mediated by the homeodomain of Isl1. Functionally, using mouse β-cell and human neuroblastoma tumor cell lines, we show that both Wdr5 binding and H3K4 methylation level at promoters of some Isl1 target genes are significantly reduced upon depletion of Isl1, suggesting Isl1 is required for efficient locus-specific H3K4 methylation. Taken together, our results establish a critical role of Set1/Mll complexes in regulating the target gene expression of Isl1.
Collapse
Affiliation(s)
- Zhe Liu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weijing Hu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yali Qin
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Sun
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lingyun Jing
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Manman Lu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Li
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Qu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhenhua Yang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
15
|
Wu D, Jiang Y, Wang Z, Ni Y, Ma A, Zhou Y, Liu R, Lou YR, Wang Q. Metabolomics analysis of islet regeneration in partial pancreatectomy mice reveals increased levels of long-chain fatty acids and activated cAMP signaling pathway. Biochem Biophys Res Commun 2023; 667:34-42. [PMID: 37207562 DOI: 10.1016/j.bbrc.2023.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/03/2023] [Indexed: 05/21/2023]
Abstract
Islet regeneration is a complex process involving multiple metabolic adaptions, but the specific characterization of the islet metabolome in relation to cell proliferation has not been established. This study aimed to investigate the metabolomic changes of regenerative islets from partial pancreatectomy (Ppx) mice and speculate underlying mechanisms. Islet samples were collected from C57/BL6 mice undergoing 70-80% Ppx or sham surgery, followed by analyses of glucose homeostasis, islet morphology, and untargeted metabolomics profiles using liquid chromatography-tandem mass spectrometry (LC-MS/MS). There is no difference in blood glucose and body weight between sham and Ppx mice. After surgery, the Ppx mice showed impaired glucose tolerance, increased Ki67 positive beta cells, and elevated beta-cell mass. LC-MS/MS analysis identified fourteen differentially changed metabolites in islets of Ppx mice, including long-chain fatty acids (e.g., docosahexaenoic acid) and amino acid derivatives (e.g., creatine). Pathway analysis based on the KEGG database revealed five significantly enriched signaling pathways including cAMP signaling pathway. Further immunostaining assay on pancreatic tissue sections showed the levels of p-CREB, a transcription factor downstream of cAMP, elevated in islets from Ppx mice. In conclusion, our results demonstrate that islet regeneration involves metabolic alterations in long-chain fatty acids and amino acid derivatives, as well as the activation of the cAMP signaling pathway.
Collapse
Affiliation(s)
- Di Wu
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China
| | - Yaojing Jiang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China
| | - Zhihong Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China
| | - Yunzhi Ni
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China
| | - Anran Ma
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China
| | - Yue Zhou
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China
| | - Rui Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China
| | - Yan-Ru Lou
- School of Pharmacy, Fudan University, Shanghai, China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetes, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Dror E, Fagnocchi L, Wegert V, Apostle S, Grimaldi B, Gruber T, Panzeri I, Heyne S, Höffler KD, Kreiner V, Ching R, Tsai-Hsiu Lu T, Semwal A, Johnson B, Senapati P, Lempradl A, Schones D, Imhof A, Shen H, Pospisilik JA. Epigenetic dosage identifies two major and functionally distinct β cell subtypes. Cell Metab 2023; 35:821-836.e7. [PMID: 36948185 PMCID: PMC10160009 DOI: 10.1016/j.cmet.2023.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/17/2023] [Accepted: 03/08/2023] [Indexed: 03/24/2023]
Abstract
The mechanisms that specify and stabilize cell subtypes remain poorly understood. Here, we identify two major subtypes of pancreatic β cells based on histone mark heterogeneity (βHI and βLO). βHI cells exhibit ∼4-fold higher levels of H3K27me3, distinct chromatin organization and compaction, and a specific transcriptional pattern. βHI and βLO cells also differ in size, morphology, cytosolic and nuclear ultrastructure, epigenomes, cell surface marker expression, and function, and can be FACS separated into CD24+ and CD24- fractions. Functionally, βHI cells have increased mitochondrial mass, activity, and insulin secretion in vivo and ex vivo. Partial loss of function indicates that H3K27me3 dosage regulates βHI/βLO ratio in vivo, suggesting that control of β cell subtype identity and ratio is at least partially uncoupled. Both subtypes are conserved in humans, with βHI cells enriched in humans with type 2 diabetes. Thus, epigenetic dosage is a novel regulator of cell subtype specification and identifies two functionally distinct β cell subtypes.
Collapse
Affiliation(s)
- Erez Dror
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany.
| | - Luca Fagnocchi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Vanessa Wegert
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Stefanos Apostle
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Brooke Grimaldi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Tim Gruber
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ilaria Panzeri
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Steffen Heyne
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Kira Daniela Höffler
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Victor Kreiner
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Reagan Ching
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Tess Tsai-Hsiu Lu
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Ayush Semwal
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ben Johnson
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Parijat Senapati
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Adelheid Lempradl
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Dustin Schones
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Axel Imhof
- Biomedical Center Munich, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Hui Shen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - John Andrew Pospisilik
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
17
|
Fu Q, Jiang H, Qian Y, Lv H, Dai H, Zhou Y, Chen Y, He Y, Gao R, Zheng S, Liang Y, Li S, Xu X, Xu K, Yang T. Single-cell RNA sequencing combined with single-cell proteomics identifies the metabolic adaptation of islet cell subpopulations to high-fat diet in mice. Diabetologia 2023; 66:724-740. [PMID: 36538064 PMCID: PMC9765371 DOI: 10.1007/s00125-022-05849-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022]
Abstract
AIMS/HYPOTHESIS Islets have complex heterogeneity and subpopulations. Cell surface markers representing alpha, beta and delta cell subpopulations are urgently needed for investigations to explore the compositional changes of each subpopulation in obesity progress and diabetes onset, and the adaptation mechanism of islet metabolism induced by a high-fat diet (HFD). METHODS Single-cell RNA sequencing (scRNA-seq) was applied to identify alpha, beta and delta cell subpopulation markers in an HFD-induced mouse model of glucose intolerance. Flow cytometry and immunostaining were used to sort and assess the proportion of each subpopulation. Single-cell proteomics was performed on sorted cells, and the functional status of each alpha, beta and delta cell subpopulation in glucose intolerance was deeply elucidated based on protein expression. RESULTS A total of 33,999 cells were analysed by scRNA-seq and clustered into eight populations, including alpha, beta and delta cells. For alpha cells, scRNA-seq revealed that the Ace2low subpopulation had downregulated expression of genes related to alpha cell function and upregulated expression of genes associated with beta cell characteristics in comparison with the Ace2high subpopulation. The impaired function and increased fragility of ACE2low alpha cells exposure to HFD was further suggested by single-cell proteomics. As for beta cells, the CD81high subpopulation may indicate an immature signature of beta cells compared with the CD81low subpopulation, which had robust function. We also found differential expression of Slc2a2 in delta cells and a potentially stronger cellular function and metabolism in GLUT2low delta cells than GLUT2high delta cells. Moreover, an increased proportion of ACE2low alpha cells and CD81low beta cells, with a constant proportion of GLUT2low delta cells, were observed in HFD-induced glucose intolerance. CONCLUSIONS/INTERPRETATION We identified ACE2, CD81 and GLUT2 as surface markers to distinguish, respectively, alpha, beta and delta cell subpopulations with heterogeneous maturation and function. The changes in the proportion and functional status of islet endocrine subpopulations reflect the metabolic adaptation of islets to high-fat stress, which weakened the function of alpha cells and enhanced the function of beta and delta cells to bring about glycaemic homeostasis. Our findings provide a fundamental resource for exploring the mechanisms maintaining each islet endocrine subpopulation's fate and function in health and disease. DATA AVAILABILITY The scRNA-seq analysis datasets from the current study are available in the Gene Expression Omnibus (GEO) repository under the accession number GSE203376.
Collapse
Affiliation(s)
- Qi Fu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hemin Jiang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Qian
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Lv
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Dai
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuncai Zhou
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Chen
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunqiang He
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Gao
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuai Zheng
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yucheng Liang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Siqi Li
- BGI-Shenzhen, Shenzhen, China
- BGI-Wuhan Clinical Laboratories, BGI-Shenzhen, Wuhan, China
| | - Xinyu Xu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuanfeng Xu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Tao Yang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Galvis D, Hodson DJ, Wedgwood KC. Spatial distribution of heterogeneity as a modulator of collective dynamics in pancreatic beta-cell networks and beyond. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:fnetp.2023.1170930. [PMID: 36987428 PMCID: PMC7614376 DOI: 10.3389/fnetp.2023.1170930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
We study the impact of spatial distribution of heterogeneity on collective dynamics in gap-junction coupled beta-cell networks comprised on cells from two populations that differ in their intrinsic excitability. Initially, these populations are uniformly and randomly distributed throughout the networks. We develop and apply an iterative algorithm for perturbing the arrangement of the network such that cells from the same population are increasingly likely to be adjacent to one another. We find that the global input strength, or network drive, necessary to transition the network from a state of quiescence to a state of synchronised and oscillatory activity decreases as network sortedness increases. Moreover, for weak coupling, we find that regimes of partial synchronisation and wave propagation arise, which depend both on network drive and network sortedness. We then demonstrate the utility of this algorithm for studying the distribution of heterogeneity in general networks, for which we use Watts-Strogatz networks as a case study. This work highlights the importance of heterogeneity in node dynamics in establishing collective rhythms in complex, excitable networks and has implications for a wide range of real-world systems that exhibit such heterogeneity.
Collapse
Affiliation(s)
- Daniel Galvis
- Centre for Systems Modelling and Quantitative Biomedicine, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Correspondence: Daniel Galvis,
| | - David J. Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
- Oxford Centre for Diabetes, Endocrinology, and Metabolism (OCDEM), Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Kyle C.A. Wedgwood
- Living Systems Institute, University of Exeter, Exeter, UK
- EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
19
|
Sosa-Larios TC, Ortega-Márquez AL, Rodríguez-Aguilera JR, Vázquez-Martínez ER, Domínguez-López A, Morimoto S. A low-protein maternal diet during gestation affects the expression of key pancreatic β-cell genes and the methylation status of the regulatory region of the MafA gene in the offspring of Wistar rats. Front Vet Sci 2023; 10:1138564. [PMID: 36992977 PMCID: PMC10040775 DOI: 10.3389/fvets.2023.1138564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
Maternal nutrition during gestation has important effects on gene expression-mediated metabolic programming in offspring. To evaluate the effect of a protein-restricted maternal diet during gestation, pancreatic islets from male progeny of Wistar rats were studied at postnatal days (PND) 36 (juveniles) and 90 (young adults). The expression of key genes involved in β-cell function and the DNA methylation pattern of the regulatory regions of two such genes, Pdx1 (pancreatic and duodenal homeobox 1) and MafA (musculoaponeurotic fibrosarcoma oncogene family, protein A), were investigated. Gene expression analysis in the pancreatic islets of restricted offspring showed significant differences compared with the control group at PND 36 (P < 0.05). The insulin 1 and 2 (Ins1 and Ins2), Glut2 (glucose transporter 2), Pdx1, MafA, and Atf2 (activating transcription factor 2), genes were upregulated, while glucokinase (Gck) and NeuroD1 (neuronal differentiation 1) were downregulated. Additionally, we studied whether the gene expression differences in Pdx1 and MafA between control and restricted offspring were associated with differential DNA methylation status in their regulatory regions. A decrease in the DNA methylation levels was found in the 5' flanking region between nucleotides −8118 to −7750 of the MafA regulatory region in restricted offspring compared with control pancreatic islets. In conclusion, low protein availability during gestation causes the upregulation of MafA gene expression in pancreatic β-cells in the male juvenile offspring at least in part through DNA hypomethylation. This process may contribute to developmental dysregulation of β-cell function and influence the long-term health of the offspring.
Collapse
Affiliation(s)
- Tonantzin C. Sosa-Larios
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Ana L. Ortega-Márquez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Jesús R. Rodríguez-Aguilera
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edgar R. Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aaron Domínguez-López
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Sumiko Morimoto
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
- *Correspondence: Sumiko Morimoto
| |
Collapse
|
20
|
Chabosseau P, Yong F, Delgadillo-Silva LF, Lee EY, Melhem R, Li S, Gandhi N, Wastin J, Noriega LL, Leclerc I, Ali Y, Hughes JW, Sladek R, Martinez-Sanchez A, Rutter GA. Molecular phenotyping of single pancreatic islet leader beta cells by "Flash-Seq". Life Sci 2023; 316:121436. [PMID: 36706832 DOI: 10.1016/j.lfs.2023.121436] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
AIMS Spatially-organized increases in cytosolic Ca2+ within pancreatic beta cells in the pancreatic islet underlie the stimulation of insulin secretion by high glucose. Recent data have revealed the existence of subpopulations of beta cells including "leaders" which initiate Ca2+ waves. Whether leader cells possess unique molecular features, or localisation, is unknown. MAIN METHODS High speed confocal Ca2+ imaging was used to identify leader cells and connectivity analysis, running under MATLAB and Python, to identify highly connected "hub" cells. To explore transcriptomic differences between beta cell sub-groups, individual leaders or followers were labelled by photo-activation of the cryptic fluorescent protein PA-mCherry and subjected to single cell RNA sequencing ("Flash-Seq"). KEY FINDINGS Distinct Ca2+ wave types were identified in individual islets, with leader cells present in 73 % (28 of 38 islets imaged). Scale-free, power law-adherent behaviour was also observed in 29 % of islets, though "hub" cells in these islets did not overlap with leaders. Transcripts differentially expressed (295; padj < 0.05) between leader and follower cells included genes involved in cilium biogenesis and transcriptional regulation. Providing some support for these findings, ADCY6 immunoreactivity tended to be higher in leader than follower cells, whereas cilia number and length tended to be lower in the former. Finally, leader cells were located significantly closer to delta, but not alpha, cells in Euclidian space than were follower cells. SIGNIFICANCE The existence of both a discrete transcriptome and unique localisation implies a role for these features in defining the specialized function of leaders. These data also raise the possibility that localised signalling between delta and leader cells contributes to the initiation and propagation of islet Ca2+ waves.
Collapse
Affiliation(s)
- Pauline Chabosseau
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Fiona Yong
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom; Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore
| | - Luis F Delgadillo-Silva
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Eun Young Lee
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States; Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Rana Melhem
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Shiying Li
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Nidhi Gandhi
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Jules Wastin
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Livia Lopez Noriega
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Isabelle Leclerc
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Yusuf Ali
- Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore
| | - Jing W Hughes
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Robert Sladek
- Departments of Medicine and Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Guy A Rutter
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada; Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom; Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore.
| |
Collapse
|
21
|
Brownrigg GP, Xia YH, Chu CMJ, Wang S, Chao C, Zhang JA, Skovsø S, Panzhinskiy E, Hu X, Johnson JD, Rideout EJ. Sex differences in islet stress responses support female β cell resilience. Mol Metab 2023; 69:101678. [PMID: 36690328 PMCID: PMC9971554 DOI: 10.1016/j.molmet.2023.101678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/07/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Pancreatic β cells play a key role in maintaining glucose homeostasis; dysfunction of this critical cell type causes type 2 diabetes (T2D). Emerging evidence points to sex differences in β cells, but few studies have examined male-female differences in β cell stress responses and resilience across multiple contexts, including diabetes. Here, we address the need for high-quality information on sex differences in β cell and islet gene expression and function using both human and rodent samples. METHODS In humans, we compared β cell gene expression and insulin secretion in donors with T2D to non-diabetic donors in both males and females. In mice, we generated a well-powered islet RNAseq dataset from 20-week-old male and female siblings with similar insulin sensitivity. Our unbiased gene expression analysis pointed to a sex difference in the endoplasmic reticulum (ER) stress response. Based on this analysis, we hypothesized female islets would be more resilient to ER stress than male islets. To test this, we subjected islets isolated from age-matched male and female mice to thapsigargin treatment and monitored protein synthesis, cell death, and β cell insulin production and secretion. Transcriptomic and proteomic analyses were used to characterize sex differences in islet responses to ER stress. RESULTS Our single-cell analysis of human β cells revealed sex-specific changes to gene expression and function in T2D, correlating with more robust insulin secretion in human islets isolated from female donors with T2D compared to male donors with T2D. In mice, RNA sequencing revealed differential enrichment of unfolded protein response pathway-associated genes, where female islets showed higher expression of genes linked with protein synthesis, folding, and processing. This differential expression was physiologically significant, as islets isolated from female mice were more resilient to ER stress induction with thapsigargin. Specifically, female islets showed a greater ability to maintain glucose-stimulated insulin production and secretion during ER stress compared with males. CONCLUSIONS Our data demonstrate sex differences in β cell gene expression in both humans and mice, and that female β cells show a greater ability to maintain glucose-stimulated insulin secretion across multiple physiological and pathological contexts.
Collapse
Affiliation(s)
- George P Brownrigg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yi Han Xia
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Chieh Min Jamie Chu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Su Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Charlotte Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jiashuo Aaron Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Søs Skovsø
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Evgeniy Panzhinskiy
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Xiaoke Hu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
22
|
Abstract
The islets of Langerhans are highly organized structures that have species-specific, three-dimensional tissue architecture. Islet architecture is critical for proper hormone secretion in response to nutritional stimuli. Islet architecture is disrupted in all types of diabetes mellitus and in cadaveric islets for transplantation during isolation, culture, and perfusion, limiting patient outcomes. Moreover, recapitulating native islet architecture remains a key challenge for in vitro generation of islets from stem cells. In this review, we discuss work that has led to the current understanding of determinants of pancreatic islet architecture, and how this architecture is maintained or disrupted during tissue remodeling in response to normal and pathological metabolic changes. We further discuss both empirical and modeling data that highlight the importance of islet architecture for islet function.
Collapse
Affiliation(s)
- Melissa T. Adams
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barak Blum
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
- CONTACT Barak Blum Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705, USA
| |
Collapse
|
23
|
Ebrahim N, Shakirova K, Dashinimaev E. PDX1 is the cornerstone of pancreatic β-cell functions and identity. Front Mol Biosci 2022; 9:1091757. [PMID: 36589234 PMCID: PMC9798421 DOI: 10.3389/fmolb.2022.1091757] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Diabetes has been a worldwide healthcare problem for many years. Current methods of treating diabetes are still largely directed at symptoms, aiming to control the manifestations of the pathology. This creates an overall need to find alternative measures that can impact on the causes of the disease, reverse diabetes, or make it more manageable. Understanding the role of key players in the pathogenesis of diabetes and the related β-cell functions is of great importance in combating diabetes. PDX1 is a master regulator in pancreas organogenesis, the maturation and identity preservation of β-cells, and of their role in normal insulin function. Mutations in the PDX1 gene are correlated with many pancreatic dysfunctions, including pancreatic agenesis (homozygous mutation) and MODY4 (heterozygous mutation), while in other types of diabetes, PDX1 expression is reduced. Therefore, alternative approaches to treat diabetes largely depend on knowledge of PDX1 regulation, its interaction with other transcription factors, and its role in obtaining β-cells through differentiation and transdifferentiation protocols. In this article, we review the basic functions of PDX1 and its regulation by genetic and epigenetic factors. Lastly, we summarize different variations of the differentiation protocols used to obtain β-cells from alternative cell sources, using PDX1 alone or in combination with various transcription factors and modified culture conditions. This review shows the unique position of PDX1 as a potential target in the genetic and cellular treatment of diabetes.
Collapse
Affiliation(s)
- Nour Ebrahim
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia,Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | - Ksenia Shakirova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Erdem Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia,Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia,*Correspondence: Erdem Dashinimaev,
| |
Collapse
|
24
|
Chu CMJ, Modi H, Ellis C, Krentz NAJ, Skovsø S, Zhao YB, Cen H, Noursadeghi N, Panzhinskiy E, Hu X, Dionne DA, Xia YH, Xuan S, Huising MO, Kieffer TJ, Lynn FC, Johnson JD. Dynamic Ins2 Gene Activity Defines β-Cell Maturity States. Diabetes 2022; 71:2612-2631. [PMID: 36170671 DOI: 10.2337/db21-1065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 09/20/2022] [Indexed: 01/11/2023]
Abstract
Transcriptional and functional cellular specialization has been described for insulin-secreting β-cells of the endocrine pancreas. However, it is not clear whether β-cell heterogeneity is stable or reflects dynamic cellular states. We investigated the temporal kinetics of endogenous insulin gene activity using live cell imaging, with complementary experiments using FACS and single-cell RNA sequencing, in β-cells from Ins2GFP knockin mice. In vivo staining and FACS analysis of islets from Ins2GFP mice confirmed that at a given moment, ∼25% of β-cells exhibited significantly higher activity at the evolutionarily conserved insulin gene, Ins2. Live cell imaging over days captured Ins2 gene activity dynamics in single β-cells. Autocorrelation analysis revealed a subset of oscillating cells, with mean oscillation periods of 17 h. Increased glucose concentrations stimulated more cells to oscillate and resulted in higher average Ins2 gene activity per cell. Single-cell RNA sequencing showed that Ins2(GFP)HIGH β-cells were enriched for markers of β-cell maturity. Ins2(GFP)HIGH β-cells were also significantly less viable at all glucose concentrations and in the context of endoplasmic reticulum stress. Collectively, our results demonstrate that the heterogeneity of insulin production, observed in mouse and human β-cells, can be accounted for by dynamic states of insulin gene activity.
Collapse
Affiliation(s)
- Chieh Min Jamie Chu
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Honey Modi
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Cara Ellis
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Nicole A J Krentz
- BC Children's Hospital Research Institute, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - Søs Skovsø
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Yiwei Bernie Zhao
- Biomedical Research Centre, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Haoning Cen
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Nilou Noursadeghi
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Evgeniy Panzhinskiy
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Xiaoke Hu
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Derek A Dionne
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Yi Han Xia
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Shouhong Xuan
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Timothy J Kieffer
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| | - Francis C Lynn
- BC Children's Hospital Research Institute, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - James D Johnson
- Diabetes Focus Team, Life Sciences Institute, Departments of Cellular and Physiological Sciences and Surgery, University of British Columbia, Vancouver, Canada
| |
Collapse
|
25
|
Usher ET, Showalter SA. Biophysical insights into glucose-dependent transcriptional regulation by PDX1. J Biol Chem 2022; 298:102623. [PMID: 36272648 PMCID: PMC9691942 DOI: 10.1016/j.jbc.2022.102623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/22/2022] Open
Abstract
The pancreatic and duodenal homeobox 1 (PDX1) is a central regulator of glucose-dependent transcription of insulin in pancreatic β cells. PDX1 transcription factor activity is integral to the development and sustained health of the pancreas; accordingly, deciphering the complex network of cellular cues that lead to PDX1 activation or inactivation is an important step toward understanding the etiopathologies of pancreatic diseases and the development of novel therapeutics. Despite nearly 3 decades of research into PDX1 control of Insulin expression, the molecular mechanisms that dictate the function of PDX1 in response to glucose are still elusive. The transcriptional activation functions of PDX1 are regulated, in part, by its two intrinsically disordered regions, which pose a barrier to its structural and biophysical characterization. Indeed, many studies of PDX1 interactions, clinical mutations, and posttranslational modifications lack molecular level detail. Emerging methods for the quantitative study of intrinsically disordered regions and refined models for transactivation now enable us to validate and interrogate the biochemical and biophysical features of PDX1 that dictate its function. The goal of this review is to summarize existing PDX1 studies and, further, to generate a comprehensive resource for future studies of transcriptional control via PDX1.
Collapse
Affiliation(s)
- Emery T Usher
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Scott A Showalter
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania, USA.
| |
Collapse
|
26
|
Shrestha S, Erikson G, Lyon J, Spigelman AF, Bautista A, Manning Fox JE, dos Santos C, Shokhirev M, Cartailler JP, Hetzer MW, MacDonald PE, Arrojo e Drigo R. Aging compromises human islet beta cell function and identity by decreasing transcription factor activity and inducing ER stress. SCIENCE ADVANCES 2022; 8:eabo3932. [PMID: 36197983 PMCID: PMC9534504 DOI: 10.1126/sciadv.abo3932] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/17/2022] [Indexed: 05/02/2023]
Abstract
Pancreatic islet beta cells are essential for maintaining glucose homeostasis. To understand the impact of aging on beta cells, we performed meta-analysis of single-cell RNA sequencing datasets, transcription factor (TF) regulon analysis, high-resolution confocal microscopy, and measured insulin secretion from nondiabetic donors spanning most of the human life span. This revealed the range of molecular and functional changes that occur during beta cell aging, including the transcriptional deregulation that associates with cellular immaturity and reorganization of beta cell TF networks, increased gene transcription rates, and reduced glucose-stimulated insulin release. These alterations associate with activation of endoplasmic reticulum (ER) stress and autophagy pathways. We propose that a chronic state of ER stress undermines old beta cell structure function to increase the risk of beta cell failure and type 2 diabetes onset as humans age.
Collapse
Affiliation(s)
- Shristi Shrestha
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Nashville, TN 37232, USA
| | - Galina Erikson
- Integrative Genomics and Bioinformatics Core, Salk Institute of Biological Studies, La Jolla, CA 92037, USA
| | - James Lyon
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta T6G2E1, Canada
| | - Aliya F. Spigelman
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta T6G2E1, Canada
| | - Austin Bautista
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta T6G2E1, Canada
| | - Jocelyn E. Manning Fox
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta T6G2E1, Canada
| | - Cristiane dos Santos
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Maxim Shokhirev
- Integrative Genomics and Bioinformatics Core, Salk Institute of Biological Studies, La Jolla, CA 92037, USA
| | | | - Martin W. Hetzer
- Molecular and Cell Biology Laboratory, Salk Institute of Biological Studies, La Jolla, CA 92037, USA
| | - Patrick E. MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta T6G2E1, Canada
| | - Rafael Arrojo e Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
27
|
Expression Profiling of Pdx1, Ngn3, and MafA in the Liver and Pancreas of Recovering Streptozotocin-Induced Diabetic Rats. Genes (Basel) 2022; 13:genes13091625. [PMID: 36140793 PMCID: PMC9498460 DOI: 10.3390/genes13091625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Studies in animal diabetic models have demonstrated the possibility of islet regeneration through treatment with natural extracts, such as Allium sativum (garlic). This study aimed to investigate the effect of garlic extract (GE) on the expression of three genes (Ngn3, Pdx1, and MafA) in the pancreas and liver of diabetic rats. Thirty-two rats were divided into two groups, streptozotocin (STZ)-induced diabetic rats (n = 16) and healthy rats (n = 16). Both groups were subdivided into GE-treated (n = 8), and those administered 0.9% normal saline (NS) (n = 8) for 1 week (n = 4) and 8 weeks (n = 4). In the pancreas of diabetic rats treated with GE for 1 week, all three genes, Ngn3, Pdx1, and MafA, were significantly upregulated (p ≤ 0.01, p ≤ 0.05, and p ≤ 0.001, respectively) when compared to diabetic rats treated with NS only. However, after eight weeks of GE treatment, the expression of all three genes decreased as blood insulin increased. In the liver, only Pdx1 expression significantly (p ≤ 0.05) increased after 8 weeks. The significant expression of Ngn3, Pdx1, and MafA in the pancreas by week 1 may have induced the maturation of juvenile β-cells, which escaped the effects of STZ and caused an increase in serum insulin.
Collapse
|
28
|
Chen CW, Guan BJ, Alzahrani MR, Gao Z, Gao L, Bracey S, Wu J, Mbow CA, Jobava R, Haataja L, Zalavadia AH, Schaffer AE, Lee H, LaFramboise T, Bederman I, Arvan P, Mathews CE, Gerling IC, Kaestner KH, Tirosh B, Engin F, Hatzoglou M. Adaptation to chronic ER stress enforces pancreatic β-cell plasticity. Nat Commun 2022; 13:4621. [PMID: 35941159 PMCID: PMC9360004 DOI: 10.1038/s41467-022-32425-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic β-cells are prone to endoplasmic reticulum (ER) stress due to their role in insulin secretion. They require sustainable and efficient adaptive stress responses to cope with this stress. Whether episodes of chronic stress directly compromise β-cell identity is unknown. We show here under reversible, chronic stress conditions β-cells undergo transcriptional and translational reprogramming associated with impaired expression of regulators of β-cell function and identity. Upon recovery from stress, β-cells regain their identity and function, indicating a high degree of adaptive plasticity. Remarkably, while β-cells show resilience to episodic ER stress, when episodes exceed a threshold, β-cell identity is gradually lost. Single cell RNA-sequencing analysis of islets from type 1 diabetes patients indicates severe deregulation of the chronic stress-adaptation program and reveals novel biomarkers of diabetes progression. Our results suggest β-cell adaptive exhaustion contributes to diabetes pathogenesis.
Collapse
Affiliation(s)
- Chien-Wen Chen
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Bo-Jhih Guan
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mohammed R Alzahrani
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Zhaofeng Gao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Long Gao
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Syrena Bracey
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jing Wu
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Cheikh A Mbow
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Raul Jobava
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Leena Haataja
- The Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI, 48105, USA
| | - Ajay H Zalavadia
- Lerner Research Institute, Cleveland Clinic, 9620 Carnegie Ave N Bldg, Cleveland, OH, 44106, US
| | - Ashleigh E Schaffer
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Thomas LaFramboise
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Peter Arvan
- The Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI, 48105, USA
| | - Clayton E Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, US
| | - Ivan C Gerling
- Department of Medicine, University of Tennessee, Memphis, TN, US
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Boaz Tirosh
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
- The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA.
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA.
| | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
29
|
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: 14] [Impact Index Per Article: 4.7] [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
|
30
|
Simoni A, Huber HA, Georgia SK, Finley SD. Phosphatases are predicted to govern prolactin-mediated JAK–STAT signaling in pancreatic beta cells. Integr Biol (Camb) 2022; 14:37-48. [DOI: 10.1093/intbio/zyac004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Patients with diabetes are unable to produce a sufficient amount of insulin to properly regulate their blood glucose levels. One potential method of treating diabetes is to increase the number of insulin-secreting beta cells in the pancreas to enhance insulin secretion. It is known that during pregnancy, pancreatic beta cells proliferate in response to the pregnancy hormone, prolactin (PRL). Leveraging this proliferative response to PRL may be a strategy to restore endogenous insulin production for patients with diabetes. To investigate this potential treatment, we previously developed a computational model to represent the PRL-mediated JAK–STAT signaling pathway in pancreatic beta cells. Here, we applied the model to identify the importance of particular signaling proteins in shaping the response of a population of beta cells. We simulated a population of 10 000 heterogeneous cells with varying initial protein concentrations responding to PRL stimulation. We used partial least squares regression to analyze the significance and role of each of the varied protein concentrations in producing the response of the cell. Our regression models predict that the concentrations of the cytosolic and nuclear phosphatases strongly influence the response of the cell. The model also predicts that increasing PRL receptor strengthens negative feedback mediated by the inhibitor suppressor of cytokine signaling. These findings reveal biological targets that can potentially be used to modulate the proliferation of pancreatic beta cells to enhance insulin secretion and beta cell regeneration in the context of diabetes.
Collapse
Affiliation(s)
- Ariella Simoni
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Holly A Huber
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Senta K Georgia
- Departments of Pediatrics and Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
31
|
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
|
32
|
Liang J, Chirikjian M, Pajvani UB, Bartolomé A. MafA Regulation in β-Cells: From Transcriptional to Post-Translational Mechanisms. Biomolecules 2022; 12:biom12040535. [PMID: 35454124 PMCID: PMC9033020 DOI: 10.3390/biom12040535] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [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
- Correspondence:
| |
Collapse
|
33
|
Guérineau NC, Campos P, Le Tissier PR, Hodson DJ, Mollard P. Cell Networks in Endocrine/Neuroendocrine Gland Function. Compr Physiol 2022; 12:3371-3415. [PMID: 35578964 DOI: 10.1002/cphy.c210031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reproduction, growth, stress, and metabolism are determined by endocrine/neuroendocrine systems that regulate circulating hormone concentrations. All these systems generate rhythms and changes in hormone pulsatility observed in a variety of pathophysiological states. Thus, the output of endocrine/neuroendocrine systems must be regulated within a narrow window of effective hormone concentrations but must also maintain a capacity for plasticity to respond to changing physiological demands. Remarkably most endocrinologists still have a "textbook" view of endocrine gland organization which has emanated from 20th century histological studies on thin 2D tissue sections. However, 21st -century technological advances, including in-depth 3D imaging of specific cell types have vastly changed our knowledge. We now know that various levels of multicellular organization can be found across different glands, that organizational motifs can vary between species and can be modified to enhance or decrease hormonal release. This article focuses on how the organization of cells regulates hormone output using three endocrine/neuroendocrine glands that present different levels of organization and complexity: the adrenal medulla, with a single neuroendocrine cell type; the anterior pituitary, with multiple intermingled cell types; and the pancreas with multiple intermingled cell types organized into distinct functional units. We give an overview of recent methodologies that allow the study of the different components within endocrine systems, particularly their temporal and spatial relationships. We believe the emerging findings about network organization, and its impact on hormone secretion, are crucial to understanding how homeostatic regulation of endocrine axes is carried out within endocrine organs themselves. © 2022 American Physiological Society. Compr Physiol 12:3371-3415, 2022.
Collapse
Affiliation(s)
| | - Pauline Campos
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Paul R Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.,COMPARE University of Birmingham and University of Nottingham Midlands, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Patrice Mollard
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
34
|
San Martín A, Arce-Molina R, Aburto C, Baeza-Lehnert F, Barros LF, Contreras-Baeza Y, Pinilla A, Ruminot I, Rauseo D, Sandoval PY. Visualizing physiological parameters in cells and tissues using genetically encoded indicators for metabolites. Free Radic Biol Med 2022; 182:34-58. [PMID: 35183660 DOI: 10.1016/j.freeradbiomed.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
The study of metabolism is undergoing a renaissance. Since the year 2002, over 50 genetically-encoded fluorescent indicators (GEFIs) have been introduced, capable of monitoring metabolites with high spatial/temporal resolution using fluorescence microscopy. Indicators are fusion proteins that change their fluorescence upon binding a specific metabolite. There are indicators for sugars, monocarboxylates, Krebs cycle intermediates, amino acids, cofactors, and energy nucleotides. They permit monitoring relative levels, concentrations, and fluxes in living systems. At a minimum they report relative levels and, in some cases, absolute concentrations may be obtained by performing ad hoc calibration protocols. Proper data collection, processing, and interpretation are critical to take full advantage of these new tools. This review offers a survey of the metabolic indicators that have been validated in mammalian systems. Minimally invasive, these indicators have been instrumental for the purposes of confirmation, rebuttal and discovery. We envision that this powerful technology will foster metabolic physiology.
Collapse
Affiliation(s)
- A San Martín
- Centro de Estudios Científicos (CECs), Valdivia, Chile.
| | - R Arce-Molina
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - C Aburto
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | | | - L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - Y Contreras-Baeza
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - A Pinilla
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - D Rauseo
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - P Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| |
Collapse
|
35
|
Jiang Z, Li H, Schroer SA, Voisin V, Ju Y, Pacal M, Erdmann N, Shi W, Chung PED, Deng T, Chen NJ, Ciavarra G, Datti A, Mak TW, Harrington L, Dick FA, Bader GD, Bremner R, Woo M, Zacksenhaus E. Hypophosphorylated pRb knock-in mice exhibit hallmarks of aging and vitamin C-preventable diabetes. EMBO J 2022; 41:e106825. [PMID: 35023164 PMCID: PMC8844977 DOI: 10.15252/embj.2020106825] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/29/2021] [Accepted: 12/08/2021] [Indexed: 12/25/2022] Open
Abstract
Despite extensive analysis of pRB phosphorylation in vitro, how this modification influences development and homeostasis in vivo is unclear. Here, we show that homozygous Rb∆K4 and Rb∆K7 knock‐in mice, in which either four or all seven phosphorylation sites in the C‐terminal region of pRb, respectively, have been abolished by Ser/Thr‐to‐Ala substitutions, undergo normal embryogenesis and early development, notwithstanding suppressed phosphorylation of additional upstream sites. Whereas Rb∆K4 mice exhibit telomere attrition but no other abnormalities, Rb∆K7 mice are smaller and display additional hallmarks of premature aging including infertility, kyphosis, and diabetes, indicating an accumulative effect of blocking pRb phosphorylation. Diabetes in Rb∆K7 mice is insulin‐sensitive and associated with failure of quiescent pancreatic β‐cells to re‐enter the cell cycle in response to mitogens, resulting in induction of DNA damage response (DDR), senescence‐associated secretory phenotype (SASP), and reduced pancreatic islet mass and circulating insulin level. Pre‐treatment with the epigenetic regulator vitamin C reduces DDR, increases cell cycle re‐entry, improves islet morphology, and attenuates diabetes. These results have direct implications for cell cycle regulation, CDK‐inhibitor therapeutics, diabetes, and longevity.
Collapse
Affiliation(s)
- Zhe Jiang
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Huiqin Li
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Stephanie A Schroer
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Veronique Voisin
- The Donnelly Centre, Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - YoungJun Ju
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Marek Pacal
- Lunenfeld Tanenbaum Research Institute - Sinai Health System, Mount Sinai Hospital, Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Natalie Erdmann
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Hospital, Toronto, ON, Canada
| | - Wei Shi
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Philip E D Chung
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tao Deng
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Nien-Jung Chen
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Hospital, Toronto, ON, Canada
| | - Giovanni Ciavarra
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Alessandro Datti
- Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy.,Network Biology Collaborative Centre, SMART Laboratory for High-Throughput Screening Programs, Mount Sinai Hospital, Toronto, ON, Canada
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Hospital, Toronto, ON, Canada
| | - Lea Harrington
- Department of Medicine, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Frederick A Dick
- Department of Biochemistry, Western University, London, ON, Canada
| | - Gary D Bader
- The Donnelly Centre, Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Rod Bremner
- Lunenfeld Tanenbaum Research Institute - Sinai Health System, Mount Sinai Hospital, Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Minna Woo
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Eldad Zacksenhaus
- Max Bell Research Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
36
|
Sun Y, Yu Z, Zhang Y, Wang H, Chi Z, Chen X, Xu D. Downregulation of microRNA?342-3p eases insulin resistance and liver gluconeogenesis via regulating Rfx3 in gestational diabetes mellitus. Crit Rev Eukaryot Gene Expr 2022; 32:83-95. [DOI: 10.1615/critreveukaryotgeneexpr.2022043275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Benninger RKP, Kravets V. The physiological role of β-cell heterogeneity in pancreatic islet function. Nat Rev Endocrinol 2022; 18:9-22. [PMID: 34667280 PMCID: PMC8915749 DOI: 10.1038/s41574-021-00568-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 01/03/2023]
Abstract
Endocrine cells within the pancreatic islets of Langerhans are heterogeneous in terms of transcriptional profile, protein expression and the regulation of hormone release. Even though this heterogeneity has long been appreciated, only within the past 5 years have detailed molecular analyses led to an improved understanding of its basis. Although we are beginning to recognize why some subpopulations of endocrine cells are phenotypically different to others, arguably the most important consideration is how this heterogeneity affects the regulation of hormone release to control the homeostasis of glucose and other energy-rich nutrients. The focus of this Review is the description of how endocrine cell heterogeneity (and principally that of insulin-secreting β-cells) affects the regulation of hormone secretion within the islets of Langerhans. This discussion includes an overview of the functional characteristics of the different islet cell subpopulations and describes how they can communicate to influence islet function under basal and glucose-stimulated conditions. We further discuss how changes to the specific islet cell subpopulations or their numbers might underlie islet dysfunction in type 2 diabetes mellitus. We conclude with a discussion of several key open questions regarding the physiological role of islet cell heterogeneity.
Collapse
Affiliation(s)
- Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Vira Kravets
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
38
|
Böni-Schnetzler M, Méreau H, Rachid L, Wiedemann SJ, Schulze F, Trimigliozzi K, Meier DT, Donath MY. IL-1beta promotes the age-associated decline of beta cell function. iScience 2021; 24:103250. [PMID: 34746709 PMCID: PMC8554531 DOI: 10.1016/j.isci.2021.103250] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/03/2021] [Accepted: 10/07/2021] [Indexed: 11/08/2022] Open
Abstract
Aging is the prime risk factor for the development of type 2 diabetes. We investigated the role of the interleukin-1 (IL-1) system on insulin secretion in aged mice. During aging, expression of the protective IL-1 receptor antagonist decreased in islets, whereas IL-1beta gene expression increased specifically in the CD45 + islet immune cell fraction. One-year-old mice with a whole-body knockout of IL-1beta had higher insulin secretion in vivo and in isolated islets, along with enhanced proliferation marker Ki67 and elevated size and number of islets. Myeloid cell-specific IL-1beta knockout preserved glucose-stimulated insulin secretion during aging, whereas it declined in control mice. Isolated islets from aged myeloIL-1beta ko mice secreted more insulin along with increased expression of Ins2, Kir6.2, and of the cell-cycle gene E2f1. IL-1beta treatment of isolated islets reduced E2f1, Ins2, and Kir6.2 expression in beta cells. We conclude that IL-1beta contributes the age-associated decline of beta cell function. Islets from aged mice have increased IL-1beta and decreased IL-1Ra expression Islet immune cells are the source of increased IL-1beta expression during aging Myeloid-cell-specific IL-1beta knockout preserves insulin secretion in aged mice IL-1beta targets genes regulating insulin secretion and proliferation during aging
Collapse
Affiliation(s)
- Marianne Böni-Schnetzler
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| | - Hélène Méreau
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| | - Leila Rachid
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| | - Sophia J Wiedemann
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| | - Friederike Schulze
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| | - Kelly Trimigliozzi
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| | - Daniel T Meier
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| | - Marc Y Donath
- Endocrinology, Diabetes, and Metabolism, University Hospital of Basel, 4031 Basel, Switzerland.,Department of Biomedicine, Diabetes Research, University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
39
|
Abstract
β-Cells in the islet of Langerhans have a central role in maintaining energy homeostasis. Understanding the physiology of β-cells and other islet cells requires a deep understanding of their structural and functional organization, their interaction with vessels and nerves, the layout of paracrine interactions, and the relationship between subcellular compartments and protein complexes inside each cell. These elements are not static; they are dynamic and exert their biological actions at different scales of time. Therefore, scientists must be able to investigate (and visualize) short- and long-lived events within the pancreas and β-cells. Current technological advances in microscopy are able to bridge multiple spatiotemporal scales in biology to reveal the complexity and heterogeneity of β-cell biology. Here, I briefly discuss the historical discoveries that leveraged microscopes to establish the basis of β-cell anatomy and structure, the current imaging platforms that allow the study of islet and β-cell biology at multiple scales of resolution, and their challenges and implications. Lastly, I outline how the remarkable longevity of structural elements at different scales in biology, from molecules to cells to multicellular structures, could represent a previously unrecognized organizational pattern in developing and adult β-cells and pancreas biology.
Collapse
Affiliation(s)
- Rafael Arrojo E Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| |
Collapse
|
40
|
Shrestha S, Saunders DC, Walker JT, Camunas-Soler J, Dai XQ, Haliyur R, Aramandla R, Poffenberger G, Prasad N, Bottino R, Stein R, Cartailler JP, Parker SC, MacDonald PE, Levy SE, Powers AC, Brissova M. Combinatorial transcription factor profiles predict mature and functional human islet α and β cells. JCI Insight 2021; 6:e151621. [PMID: 34428183 PMCID: PMC8492318 DOI: 10.1172/jci.insight.151621] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Islet-enriched transcription factors (TFs) exert broad control over cellular processes in pancreatic α and β cells, and changes in their expression are associated with developmental state and diabetes. However, the implications of heterogeneity in TF expression across islet cell populations are not well understood. To define this TF heterogeneity and its consequences for cellular function, we profiled more than 40,000 cells from normal human islets by single-cell RNA-Seq and stratified α and β cells based on combinatorial TF expression. Subpopulations of islet cells coexpressing ARX/MAFB (α cells) and MAFA/MAFB (β cells) exhibited greater expression of key genes related to glucose sensing and hormone secretion relative to subpopulations expressing only one or neither TF. Moreover, all subpopulations were identified in native pancreatic tissue from multiple donors. By Patch-Seq, MAFA/MAFB-coexpressing β cells showed enhanced electrophysiological activity. Thus, these results indicate that combinatorial TF expression in islet α and β cells predicts highly functional, mature subpopulations.
Collapse
Affiliation(s)
- Shristi Shrestha
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee, USA
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John T. Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Joan Camunas-Soler
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Xiao-Qing Dai
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Rachana Haliyur
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Radhika Aramandla
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Greg Poffenberger
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nripesh Prasad
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Rita Bottino
- Imagine Pharma, Devon, Pennsylvania, USA
- Institute of Cellular Therapeutics, Allegheny-Singer Research Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | - Stephen C.J. Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick E. MacDonald
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Shawn E. Levy
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Marcela Brissova
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
41
|
Nasteska D, Cuozzo F, Viloria K, Johnson EM, Thakker A, Bany Bakar R, Westbrook RL, Barlow JP, Hoang M, Joseph JW, Lavery GG, Akerman I, Cantley J, Hodson L, Tennant DA, Hodson DJ. Prolyl-4-hydroxylase 3 maintains β cell glucose metabolism during fatty acid excess in mice. JCI Insight 2021; 6:e140288. [PMID: 34264866 PMCID: PMC8409982 DOI: 10.1172/jci.insight.140288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
The α-ketoglutarate–dependent dioxygenase, prolyl-4-hydroxylase 3 (PHD3), is an HIF target that uses molecular oxygen to hydroxylate peptidyl prolyl residues. Although PHD3 has been reported to influence cancer cell metabolism and liver insulin sensitivity, relatively little is known about the effects of this highly conserved enzyme in insulin-secreting β cells in vivo. Here, we show that the deletion of PHD3 specifically in β cells (βPHD3KO) was associated with impaired glucose homeostasis in mice fed a high-fat diet. In the early stages of dietary fat excess, βPHD3KO islets energetically rewired, leading to defects in the management of pyruvate fate and a shift from glycolysis to increased fatty acid oxidation (FAO). However, under more prolonged metabolic stress, this switch to preferential FAO in βPHD3KO islets was associated with impaired glucose-stimulated ATP/ADP rises, Ca2+ fluxes, and insulin secretion. Thus, PHD3 might be a pivotal component of the β cell glucose metabolism machinery in mice by suppressing the use of fatty acids as a primary fuel source during the early phases of metabolic stress.
Collapse
Affiliation(s)
- Daniela Nasteska
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Federica Cuozzo
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Katrina Viloria
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Elspeth M Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Alpesh Thakker
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Rula Bany Bakar
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Rebecca L Westbrook
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Jonathan P Barlow
- Mitochondrial Profiling Centre, School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Monica Hoang
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Jamie W Joseph
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Ildem Akerman
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - James Cantley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - David J Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
42
|
Lee D, Kim KH, Jang TS, Kang KS. Identification of bioactive compounds from mulberry enhancing glucose-stimulated insulin secretion. Bioorg Med Chem Lett 2021; 43:128096. [PMID: 33984475 DOI: 10.1016/j.bmcl.2021.128096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 10/21/2022]
Abstract
Previously, we isolated six heterocyclic compounds (1-6) from the fruits of mulberry trees (Morus alba L.) and determined that loliolide affords rat pancreatic islet β-cell (INS-1) protection against streptozotocin‑induced cytotoxicity. In the present study, we further investigated the effect of the six heterocyclic compounds (1-6) on glucose-stimulated insulin secretion (GSIS) in INS-1 cells. Among them, (R)‑5‑hydroxypyrrolidin‑2‑one(1) and indole (6) increased GSIS without inducing cytotoxicity. Additionally, compounds 1 and 6 enhanced the phosphorylation of total insulin receptor substrate-2, phosphatidylinositol 3-kinase, and Akt, and activated pancreatic and duodenal homeobox-1, which play a crucial role in β-cell functions related to insulin secretion. Collectively, these findings indicate that (R)‑5‑hydroxypyrrolidin‑2‑one(1) and indole (6), isolated from M. alba fruits, may be beneficial in managing type 2 diabetes.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Tae Su Jang
- Department of Medicine, Dankook University, Cheonan, Chungnam 31116, Republic of Korea.
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea.
| |
Collapse
|
43
|
Small subpopulations of β-cells do not drive islet oscillatory [Ca2+] dynamics via gap junction communication. PLoS Comput Biol 2021; 17:e1008948. [PMID: 33939712 PMCID: PMC8118513 DOI: 10.1371/journal.pcbi.1008948] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 05/13/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
The islets of Langerhans exist as multicellular networks that regulate blood glucose levels. The majority of cells in the islet are excitable, insulin-producing β-cells that are electrically coupled via gap junction channels. β-cells are known to display heterogeneous functionality. However, due to gap junction coupling, β-cells show coordinated [Ca2+] oscillations when stimulated with glucose, and global quiescence when unstimulated. Small subpopulations of highly functional β-cells have been suggested to control [Ca2+] dynamics across the islet. When these populations were targeted by optogenetic silencing or photoablation, [Ca2+] dynamics across the islet were largely disrupted. In this study, we investigated the theoretical basis of these experiments and how small populations can disproportionality control islet [Ca2+] dynamics. Using a multicellular islet model, we generated normal, skewed or bimodal distributions of β-cell heterogeneity. We examined how islet [Ca2+] dynamics were disrupted when cells were targeted via hyperpolarization or populations were removed; to mimic optogenetic silencing or photoablation, respectively. Targeted cell populations were chosen based on characteristics linked to functional subpopulation, including metabolic rate of glucose oxidation or [Ca2+] oscillation frequency. Islets were susceptible to marked suppression of [Ca2+] when ~10% of cells with high metabolic activity were hyperpolarized; where hyperpolarizing cells with normal metabolic activity had little effect. However, when highly metabolic cells were removed from the model, [Ca2+] oscillations remained. Similarly, when ~10% of cells with either the highest frequency or earliest elevations in [Ca2+] were removed from the islet, the [Ca2+] oscillation frequency remained largely unchanged. Overall, these results indicate small populations of β-cells with either increased metabolic activity or increased frequency are unable to disproportionately control islet-wide [Ca2+] via gap junction coupling. Therefore, we need to reconsider the physiological basis for such small β-cell populations or the mechanism by which they may be acting to control normal islet function. Many biological systems can be studied using network theory. How heterogeneous cell subpopulations come together to create complex multicellular behavior is of great value in understanding function and dysfunction in tissues. The pancreatic islet of Langerhans is a highly coupled structure that is important for maintaining blood glucose homeostasis. β-cell electrical activity is coordinated via gap junction communication. The function of the insulin-producing β-cell within the islet is disrupted in diabetes. As such, to understand the causes of islet dysfunction we need to understand how different cells within the islet contribute to its overall function via gap junction coupling. Using a computational model of β-cell electrophysiology, we investigated how small highly functional β-cell populations within the islet contribute to its function. We found that when small populations with greater functionality were introduced into the islet, they displayed signatures of this enhanced functionality. However, when these cells were removed, the islet, retained near-normal function. Thus, in a highly coupled system, such as an islet, the heterogeneity of cells allows small subpopulations to be dispensable, and thus their absence is unable to disrupt the larger cellular network. These findings can be applied to other electrical systems that have heterogeneous cell populations.
Collapse
|
44
|
Lorenzo PI, Cobo-Vuilleumier N, Martín-Vázquez E, López-Noriega L, Gauthier BR. Harnessing the Endogenous Plasticity of Pancreatic Islets: A Feasible Regenerative Medicine Therapy for Diabetes? Int J Mol Sci 2021; 22:4239. [PMID: 33921851 PMCID: PMC8073058 DOI: 10.3390/ijms22084239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes is a chronic metabolic disease caused by an absolute or relative deficiency in functional pancreatic β-cells that leads to defective control of blood glucose. Current treatments for diabetes, despite their great beneficial effects on clinical symptoms, are not curative treatments, leading to a chronic dependence on insulin throughout life that does not prevent the secondary complications associated with diabetes. The overwhelming increase in DM incidence has led to a search for novel antidiabetic therapies aiming at the regeneration of the lost functional β-cells to allow the re-establishment of the endogenous glucose homeostasis. Here we review several aspects that must be considered for the development of novel and successful regenerative therapies for diabetes: first, the need to maintain the heterogeneity of islet β-cells with several subpopulations of β-cells characterized by different transcriptomic profiles correlating with differences in functionality and in resistance/behavior under stress conditions; second, the existence of an intrinsic islet plasticity that allows stimulus-mediated transcriptome alterations that trigger the transdifferentiation of islet non-β-cells into β-cells; and finally, the possibility of using agents that promote a fully functional/mature β-cell phenotype to reduce and reverse the process of dedifferentiation of β-cells during diabetes.
Collapse
Affiliation(s)
- Petra I. Lorenzo
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Nadia Cobo-Vuilleumier
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Eugenia Martín-Vázquez
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Livia López-Noriega
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Benoit R. Gauthier
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 028029 Madrid, Spain
| |
Collapse
|
45
|
Abstract
Glucagon-Like Peptide-1 (GLP-1) is an important peptide hormone secreted by L-cells in the gastrointestinal tract in response to nutrients. It is produced by the differential cleavage of the proglucagon peptide. GLP-1 elicits a wide variety of physiological responses in many tissues that contribute to metabolic homeostasis. For these reasons, therapies designed to either increase endogenous GLP-1 levels or introduce exogenous peptide mimetics are now widely used in the management of diabetes. In addition to GLP-1 production from L-cells, recent reports suggest that pancreatic islet alpha cells may also synthesize and secrete GLP-1. Intra-islet GLP-1 may therefore play an unappreciated role in islet health and glucose regulation, suggesting a potential functional paracrine role for islet-derived GLP-1. In this review, we assess the current literature from an islet-centric point-of-view to better understand the production, degradation, and actions of GLP-1 within the endocrine pancreas in rodents and humans. The relevance of intra-islet GLP-1 in human physiology is discussed regarding the potential role of intra-islet GLP-1 in islet health and dysfunction.
Collapse
Affiliation(s)
- Scott A. Campbell
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal Diabetes Research Centre CRCHUM, Montréal, Canada
| | - Janyne Johnson
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Peter E. Light
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- CONTACT Peter E. Light Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AlbertaT6G 2E1, Canada
| |
Collapse
|