1
|
Arias CF, Acosta FJ, Bertocchini F, Fernández-Arias C. A functional approach to homeostatic regulation. Biol Direct 2024; 19:134. [PMID: 39709473 DOI: 10.1186/s13062-024-00577-9] [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: 08/09/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024] Open
Abstract
In this work, we present a novel modeling framework for understanding the dynamics of homeostatic regulation. Inspired by engineering control theory, this framework incorporates unique features of biological systems. First, biological variables often play physiological roles, and taking this functional context into consideration is essential to fully understand the goals and constraints of homeostatic regulation. Second, biological signals are not abstract variables, but rather material molecules that may undergo complex turnover processes of synthesis and degradation. We suggest that the particular nature of biological signals may condition the type of information they can convey, and their potential role in shaping the dynamics and the ultimate purpose of homeostatic systems. We show that the dynamic interplay between regulated variables and control signals is a key determinant of biological homeostasis, challenging the necessity and the convenience of strictly extrapolating concepts from engineering control theory in modeling the dynamics of homeostatic systems. This work provides a simple, unified framework for studying biological regulation and identifies general principles that transcend molecular details of particular homeostatic mechanisms. We show how this approach can be naturally applied to apparently different regulatory systems, contributing to a deeper understanding of homeostasis as a fundamental process in living systems.
Collapse
Affiliation(s)
- Clemente F Arias
- Grupo Interdisciplinar de Sistemas Complejos de Madrid (GISC), 28040, Madrid, Spain.
| | - Francisco J Acosta
- Departamento de Ecología, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | | | - Cristina Fernández-Arias
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| |
Collapse
|
2
|
Kajani S, Laker RC, Ratkova E, Will S, Rhodes CJ. Hepatic glucagon action: beyond glucose mobilization. Physiol Rev 2024; 104:1021-1060. [PMID: 38300523 DOI: 10.1152/physrev.00028.2023] [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: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Glucagon's ability to promote hepatic glucose production has been known for over a century, with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism [when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions] is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a "catabolic hormone." Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive "omics" technologies, has implicated glucagon as more than just a "glucose liberator." Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that "catabolic hormone."
Collapse
Affiliation(s)
- Sarina Kajani
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Rhianna C Laker
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Ekaterina Ratkova
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sarah Will
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Christopher J Rhodes
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| |
Collapse
|
3
|
Reed J, Bain SC, Kanamarlapudi V. The Regulation of Metabolic Homeostasis by Incretins and the Metabolic Hormones Produced by Pancreatic Islets. Diabetes Metab Syndr Obes 2024; 17:2419-2456. [PMID: 38894706 PMCID: PMC11184168 DOI: 10.2147/dmso.s415934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/07/2024] [Indexed: 06/21/2024] Open
Abstract
In healthy humans, the complex biochemical interplay between organs maintains metabolic homeostasis and pathological alterations in this process result in impaired metabolic homeostasis, causing metabolic diseases such as diabetes and obesity, which are major global healthcare burdens. The great advancements made during the last century in understanding both metabolic disease phenotypes and the regulation of metabolic homeostasis in healthy individuals have yielded new therapeutic options for diseases like type 2 diabetes (T2D). However, it is unlikely that highly desirable more efficacious treatments will be developed for metabolic disorders until the complex systemic regulation of metabolic homeostasis becomes more intricately understood. Hormones produced by pancreatic islet beta-cells (insulin) and alpha-cells (glucagon) are pivotal for maintaining metabolic homeostasis; the activity of insulin and glucagon are reciprocally correlated to achieve strict control of glucose levels (normoglycaemia). Metabolic hormones produced by other pancreatic islet cells and incretins produced by the gut are also crucial for maintaining metabolic homeostasis. Recent studies highlighted the incomplete understanding of metabolic hormonal synergism and, therefore, further elucidation of this will likely lead to more efficacious treatments for diseases such as T2D. The objective of this review is to summarise the systemic actions of the incretins and the metabolic hormones produced by the pancreatic islets and their interactions with their respective receptors.
Collapse
Affiliation(s)
- Joshua Reed
- Institute of Life Science, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Stephen C Bain
- Institute of Life Science, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | | |
Collapse
|
4
|
Grandl G, Collden G, Feng J, Bhattacharya S, Klingelhuber F, Schomann L, Bilekova S, Ansarullah, Xu W, Far FF, Tost M, Gruber T, Bastidas-Ponce A, Zhang Q, Novikoff A, Liskiewicz A, Liskiewicz D, Garcia-Caceres C, Feuchtinger A, Tschöp MH, Krahmer N, Lickert H, Müller TD. Global, neuronal or β cell-specific deletion of inceptor improves glucose homeostasis in male mice with diet-induced obesity. Nat Metab 2024; 6:448-457. [PMID: 38418586 DOI: 10.1038/s42255-024-00991-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 01/19/2024] [Indexed: 03/01/2024]
Abstract
Insulin resistance is an early complication of diet-induced obesity (DIO)1, potentially leading to hyperglycaemia and hyperinsulinaemia, accompanied by adaptive β cell hypertrophy and development of type 2 diabetes2. Insulin not only signals via the insulin receptor (INSR), but also promotes β cell survival, growth and function via the insulin-like growth factor 1 receptor (IGF1R)3-6. We recently identified the insulin inhibitory receptor (inceptor) as the key mediator of IGF1R and INSR desensitization7. But, although β cell-specific loss of inceptor improves β cell function in lean mice7, it warrants clarification whether inceptor signal inhibition also improves glycaemia under conditions of obesity. We assessed the glucometabolic effects of targeted inceptor deletion in either the brain or the pancreatic β cells under conditions of DIO in male mice. In the present study, we show that global and neuronal deletion of inceptor, as well as its adult-onset deletion in the β cells, improves glucose homeostasis by enhancing β cell health and function. Moreover, we demonstrate that inceptor-mediated improvement in glucose control does not depend on inceptor function in agouti-related protein-expressing or pro-opiomelanocortin neurons. Our data demonstrate that inceptor inhibition improves glucose homeostasis in mice with DIO, hence corroborating that inceptor is a crucial regulator of INSR and IGF1R signalling.
Collapse
Affiliation(s)
- Gerald Grandl
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Gustav Collden
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Jin Feng
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
- School of Medicine, Technische Universität München, Munich, Germany
| | - Sreya Bhattacharya
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
- School of Medicine, Technische Universität München, Munich, Germany
| | - Felix Klingelhuber
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Leopold Schomann
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Sara Bilekova
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Ansarullah
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Weiwei Xu
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Fataneh Fathi Far
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Monica Tost
- Core Facility Pathology & Tissue Analytics, Helmholtz Center Munich, Munich, Germany
| | - Tim Gruber
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- German Center for Diabetes Research, Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Qian Zhang
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Aaron Novikoff
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Arkadiusz Liskiewicz
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Daniela Liskiewicz
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Cristina Garcia-Caceres
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Annette Feuchtinger
- Core Facility Pathology & Tissue Analytics, Helmholtz Center Munich, Munich, Germany
| | - Matthias H Tschöp
- German Center for Diabetes Research, Neuherberg, Germany
- School of Medicine, Technische Universität München, Munich, Germany
- Helmholtz Zentrum München, Neuherberg, Germany
| | - Natalie Krahmer
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Heiko Lickert
- German Center for Diabetes Research, Neuherberg, Germany.
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany.
- School of Medicine, Technische Universität München, Munich, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| |
Collapse
|
5
|
Tu X, Jain A, Parra Bueno P, Decker H, Liu X, Yasuda R. Local autocrine plasticity signaling in single dendritic spines by insulin-like growth factors. SCIENCE ADVANCES 2023; 9:eadg0666. [PMID: 37531435 PMCID: PMC10396292 DOI: 10.1126/sciadv.adg0666] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
The insulin superfamily of peptides is essential for homeostasis as well as neuronal plasticity, learning, and memory. Here, we show that insulin-like growth factors 1 and 2 (IGF1 and IGF2) are differentially expressed in hippocampal neurons and released in an activity-dependent manner. Using a new fluorescence resonance energy transfer sensor for IGF1 receptor (IGF1R) with two-photon fluorescence lifetime imaging, we find that the release of IGF1 triggers rapid local autocrine IGF1R activation on the same spine and more than several micrometers along the stimulated dendrite, regulating the plasticity of the activated spine in CA1 pyramidal neurons. In CA3 neurons, IGF2, instead of IGF1, is responsible for IGF1R autocrine activation and synaptic plasticity. Thus, our study demonstrates the cell type-specific roles of IGF1 and IGF2 in hippocampal plasticity and a plasticity mechanism mediated by the synthesis and autocrine signaling of IGF peptides in pyramidal neurons.
Collapse
Affiliation(s)
- Xun Tu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- International Max Planck Research School for Brain and Behavior, Jupiter, FL, USA
- FAU/Max Planck Florida Institute Joint Graduate Program in Integrative Biology and Neuroscience, Florida Atlantic University, Boca Raton, FL, USA
| | - Anant Jain
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Paula Parra Bueno
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Helena Decker
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Xiaodan Liu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| |
Collapse
|
6
|
Aoyama S, Nishida Y, Uzawa H, Himuro M, Kanai A, Ueki K, Ito M, Iida H, Tanida I, Miyatsuka T, Fujitani Y, Matsumoto M, Watada H. Monitoring autophagic flux in vivo revealed its physiological response and significance of heterogeneity in pancreatic beta cells. Cell Chem Biol 2023; 30:658-671.e4. [PMID: 36944338 DOI: 10.1016/j.chembiol.2023.03.001] [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: 08/20/2022] [Revised: 01/12/2023] [Accepted: 02/28/2023] [Indexed: 03/23/2023]
Abstract
Autophagy plays an essential role in preserving cellular homeostasis in pancreatic beta cells. However, the extent of autophagic flux in pancreatic islets induced in various physiological settings remains unclear. In this study, we generate transgenic mice expressing pHluorin-LC3-mCherry reporter for monitoring systemic autophagic flux by measuring the pHluorin/mCherry ratio, validating them in the starvation and insulin-deficient model. Our findings reveal that autophagic flux in pancreatic islets enhances after starvation, and suppression of the flux after short-term refeeding needs more prolonged re-starvation in islets than in the other insulin-targeted organs. Furthermore, heterogeneity of autophagic flux in pancreatic beta cells manifests under insulin resistance, and intracellular calcium influx by glucose stimulation increases more in high- than low-autophagic flux beta cells, with differential gene expression, including lipoprotein lipase. Our pHluorin-LC3-mCherry mice enable us to reveal biological insight into heterogeneity in autophagic flux in pancreatic beta cells.
Collapse
Affiliation(s)
- Shuhei Aoyama
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yuya Nishida
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Hirotsugu Uzawa
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Miwa Himuro
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Akiko Kanai
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kyosei Ueki
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Minami Ito
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hitoshi Iida
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Isei Tanida
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takeshi Miyatsuka
- Department of Endocrinology, Diabetes and Metabolism, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi 371-8512, Japan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, 757 Ichibancho, Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
7
|
Basu L, Bhagat V, Ching MEA, Di Giandomenico A, Dostie S, Greenberg D, Greenberg M, Hahm J, Hilton NZ, Lamb K, Jentz EM, Larsen M, Locatelli CAA, Maloney M, MacGibbon C, Mersali F, Mulchandani CM, Najam A, Singh I, Weisz T, Wong J, Senior PA, Estall JL, Mulvihill EE, Screaton RA. Recent Developments in Islet Biology: A Review With Patient Perspectives. Can J Diabetes 2023; 47:207-221. [PMID: 36481263 PMCID: PMC9640377 DOI: 10.1016/j.jcjd.2022.11.003] [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: 09/01/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
Navigating the coronavirus disease-2019 (COVID-19, now COVID) pandemic has required resilience and creativity worldwide. Despite early challenges to productivity, more than 2,000 peer-reviewed articles on islet biology were published in 2021. Herein, we highlight noteworthy advances in islet research between January 2021 and April 2022, focussing on 5 areas. First, we discuss new insights into the role of glucokinase, mitogen-activated protein kinase-kinase/extracellular signal-regulated kinase and mitochondrial function on insulin secretion from the pancreatic β cell, provided by new genetically modified mouse models and live imaging. We then discuss a new connection between lipid handling and improved insulin secretion in the context of glucotoxicity, focussing on fatty acid-binding protein 4 and fetuin-A. Advances in high-throughput "omic" analysis evolved to where one can generate more finely tuned genetic and molecular profiles within broad classifications of type 1 diabetes and type 2 diabetes. Next, we highlight breakthroughs in diabetes treatment using stem cell-derived β cells and innovative strategies to improve islet survival posttransplantation. Last, we update our understanding of the impact of severe acute respiratory syndrome-coronavirus-2 infection on pancreatic islet function and discuss current evidence regarding proposed links between COVID and new-onset diabetes. We address these breakthroughs in 2 settings: one for a scientific audience and the other for the public, particularly those living with or affected by diabetes. Bridging biomedical research in diabetes to the community living with or affected by diabetes, our partners living with type 1 diabetes or type 2 diabetes also provide their perspectives on these latest advances in islet biology.
Collapse
Affiliation(s)
- Lahari Basu
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Vriti Bhagat
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Ma Enrica Angela Ching
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | | | - Sylvie Dostie
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Dana Greenberg
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Marley Greenberg
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Jiwon Hahm
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - N Zoe Hilton
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Krista Lamb
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Emelien M Jentz
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Matt Larsen
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Cassandra A A Locatelli
- University of Ottawa Heart Institute, Energy Substrate Laboratory, Ottawa, Ontario, Canada; Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, Ontario, Canada
| | - MaryAnn Maloney
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | | | - Farida Mersali
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | | | - Adhiyat Najam
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Ishnoor Singh
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Tom Weisz
- Diabetes Action Canada, Toronto General Hospital, Toronto, Ontario, Canada
| | - Jordan Wong
- Alberta Diabetes Institute and Department of Pharmacology, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada; Alberta Diabetes Institute and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Peter A Senior
- Alberta Diabetes Institute and Department of Medicine, Edmonton, Alberta, Canada
| | - Jennifer L Estall
- Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada; Institut de recherches cliniques de Montréal, Center for Cardiometabolic Health, Montréal, Québec, Canada
| | - Erin E Mulvihill
- University of Ottawa Heart Institute, Energy Substrate Laboratory, Ottawa, Ontario, Canada; Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Robert A Screaton
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Shedding light on non-alcoholic fatty liver disease: Pathogenesis, molecular mechanisms, models, and emerging therapeutics. Life Sci 2022; 312:121185. [PMID: 36375569 DOI: 10.1016/j.lfs.2022.121185] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder globally impacting an estimated 25% of the population associated with severe consequences such as cirrhosis, hepatocellular carcinoma (HCC), and overall mortality. Fatty liver disease is triggered through multiple pathways, but the most prominent cause is either diabetes or obesity, or a combination of both. Therefore, hepatic glucose, insulin and fatty acid signaling becomes a dire need to understand which is well elaborated in this review. This review summarizes the popular two-hit pathogenesis of NAFLD, the molecular mechanisms underlying hepatic insulin resistance. As fatty liver disease gets advanced, it requires in-vitro as well as in-vivo models closer to disease progression in humans for better understanding the pathological state and identifying a novel therapeutic target. This review summarizes in-vitro (2D cell-culture/co-culture, 3D spheroid/organoid/liver-on-a-chip) models as well as in-vivo (genetically/dietary/chemically induced fatty liver disease) research models. Fatty liver disease research has gathered lots of attention recently since there is no FDA approved therapy available so far. However, there have been numerous promising targets to treat fatty liver disease including potential therapeutic targets under clinical trials are listed in this review.
Collapse
|
9
|
Upregulation of Klotho Aggravates Insulin Resistance in Gestational Diabetes Mellitus Trophoblast Cells. Genet Res (Camb) 2022; 2022:1500768. [DOI: 10.1155/2022/1500768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/07/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Objective. Insulin resistance (IR) plays a key role in gestational diabetes mellitus (GDM) pathogenesis. The antiaging protein klotho has been proven to be closely related to IR. The purpose of this study was to investigate the effect of klotho on IR in GDM trophoblast cells. Methods. The GDM cell model of HTR-8/SVneo cells was induced by high glucose (HG). Plasmid transfection was used to mediate the overexpression or silencing of klotho. The effects of klotho on cell viability, IR, and the IGF-1/PI3K pathways were observed by RT-qPCR, western blot, Cell Counting Kit-8 detection, glucose uptake assay, and immunofluorescence detection. Results. Klotho expression was up-regulated in HG-induced cells. Overexpression of klotho could reduce the cell viability, insulin signaling molecules (INSR-α, INSR-β, IRS1, IRS2, and GLUT4), and glucose uptake in HTR-8/SVneo cells of the HG group. In addition, the overexpression of klotho inhibited the levels of IGF-1, IGF-1R/p-IGF-1R, and the phosphorylation and activation of the signal transduction molecules PI3K/Akt/mTOR. On the contrary, klotho deletions could reverse these changes of HTR-8/SVneo cells induced by HG. Conclusion. In a word, the results of this study showed that the regulation of klotho played an important role in the IR of trophoblast cells induced by HG, which was mediated at least in part by the IGF-1/PI3K/Akt/mTOR pathway.
Collapse
|
10
|
Biondi G, Marrano N, Dipaola L, Borrelli A, Rella M, D'Oria R, Genchi VA, Caccioppoli C, Porreca I, Cignarelli A, Perrini S, Marchetti P, Vincenti L, Laviola L, Giorgino F, Natalicchio A. The p66Shc Protein Mediates Insulin Resistance and Secretory Dysfunction in Pancreatic β-Cells Under Lipotoxic Conditions. Diabetes 2022; 71:1763-1771. [PMID: 35612429 DOI: 10.2337/db21-1066] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022]
Abstract
We evaluated the role of the p66Shc redox adaptor protein in pancreatic β-cell insulin resistance that develops under lipotoxic conditions and with excess body fat. Prolonged exposure to palmitate in vitro or the presence of overweight/obesity augmented p66Shc expression levels and caused an impaired ability of exogenous insulin to increase cellular insulin content and secreted C-peptide levels in INS-1E cells and human and murine islets. In INS-1E cells, p66Shc knockdown resulted in enhanced insulin-induced augmentation of insulin content and C-peptide secretion and prevented the ability of palmitate to impair these effects of insulin. Conversely, p66Shc overexpression impaired insulin-induced augmentation of insulin content and C-peptide secretion in both the absence and presence of palmitate. Under lipotoxic condition, the effects of p66Shc are mediated by a p53-induced increase in p66Shc protein levels and JNK-induced p66Shc phosphorylation at Ser36 and appear to involve the phosphorylation of the ribosomal protein S6 kinase at Thr389 and of insulin receptor substrate 1 at Ser307, resulting in the inhibition of insulin-stimulated protein kinase B phosphorylation at Ser473. Thus, the p66Shc protein mediates the impaired β-cell function and insulin resistance induced by saturated fatty acids and excess body fat.
Collapse
Affiliation(s)
- Giuseppina Biondi
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Nicola Marrano
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Lucia Dipaola
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Anna Borrelli
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Martina Rella
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Rossella D'Oria
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Valentina A Genchi
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Cristina Caccioppoli
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Immacolata Porreca
- Genetic Research Centre "Gaetano Salvatore" BioGeM, Ariano Irpino, Italy
| | - Angelo Cignarelli
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Sebastio Perrini
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Leonardo Vincenti
- Division of General Surgery, University Hospital Polyclinic, Bari, Italy
| | - Luigi Laviola
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Francesco Giorgino
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| |
Collapse
|
11
|
Dos Santos T, Galipeau M, Schukarucha Gomes A, Greenberg M, Larsen M, Lee D, Maghera J, Mulchandani CM, Patton M, Perera I, Polishevska K, Ramdass S, Shayeganpour K, Vafaeian K, Van Allen K, Wang Y, Weisz T, Estall JL, Mulvihill EE, Screaton RA. Islet Biology During COVID-19: Progress and Perspectives. Can J Diabetes 2022; 46:419-427. [PMID: 35589534 PMCID: PMC8608413 DOI: 10.1016/j.jcjd.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/28/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023]
Abstract
The coronavirus-2019 (COVID-19) pandemic has had significant impact on research directions and productivity in the past 2 years. Despite these challenges, since 2020, more than 2,500 peer-reviewed articles have been published on pancreatic islet biology. These include updates on the roles of isocitrate dehydrogenase, pyruvate kinase and incretin hormones in insulin secretion, as well as the discovery of inceptor and signalling by circulating RNAs. The year 2020 also brought advancements in in vivo and in vitro models, including a new transgenic mouse for assessing beta-cell proliferation, a "pancreas-on-a-chip" to study glucose-stimulated insulin secretion and successful genetic editing of primary human islet cells. Islet biologists evaluated the functionality of stem-cell-derived islet-like cells coated with semipermeable biomaterials to prevent autoimmune attack, revealing the importance of cell maturation after transplantation. Prompted by observations that COVID-19 symptoms can worsen for people with obesity or diabetes, researchers examined how islets are directly affected by severe acute respiratory syndrome coronavirus 2. Herein, we highlight novel functional insights, technologies and therapeutic approaches that emerged between March 2020 and July 2021, written for both scientific and lay audiences. We also include a response to these advancements from patient stakeholders, to help lend a broader perspective to developments and challenges in islet research.
Collapse
Affiliation(s)
- Theodore Dos Santos
- Alberta Diabetes Institute & Department of Pharmacology, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | - Maria Galipeau
- Department of Molecular Biology, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada; Institut de recherches cliniques de Montréal, Cardiometabolic Disease Centre, Montréal, Québec, Canada
| | - Amanda Schukarucha Gomes
- Alberta Diabetes Institute & Department of Pharmacology, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Daniel Lee
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Jasmine Maghera
- Alberta Diabetes Institute & Department of Pharmacology, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | | | - Megan Patton
- Toronto General Hospital, Toronto, Ontario, Canada
| | - Ineli Perera
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Kateryna Polishevska
- Alberta Diabetes Institute and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | - Kasra Shayeganpour
- Alberta Diabetes Institute and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | - Kyle Van Allen
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Yufeng Wang
- University Health Network, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Tom Weisz
- Toronto General Hospital, Toronto, Ontario, Canada
| | - Jennifer L Estall
- Department of Molecular Biology, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada; Institut de recherches cliniques de Montréal, Cardiometabolic Disease Centre, Montréal, Québec, Canada
| | - Erin E Mulvihill
- Energy Substrate Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada; Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Robert A Screaton
- Sunnybrook Research Institute, Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Khan D, Moffett RC, Flatt PR, Tarasov AI. Classical and non-classical islet peptides in the control of β-cell function. Peptides 2022; 150:170715. [PMID: 34958851 DOI: 10.1016/j.peptides.2021.170715] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/25/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022]
Abstract
The dual role of the pancreas as both an endocrine and exocrine gland is vital for food digestion and control of nutrient metabolism. The exocrine pancreas secretes enzymes into the small intestine aiding digestion of sugars and fats, whereas the endocrine pancreas secretes a cocktail of hormones into the blood, which is responsible for blood glucose control and regulation of carbohydrate, protein and fat metabolism. Classical islet hormones, insulin, glucagon, pancreatic polypeptide and somatostatin, interact in an autocrine and paracrine manner, to fine-tube the islet function and insulin secretion to the needs of the body. Recently pancreatic islets have been reported to express a number of non-classical peptide hormones involved in metabolic signalling, whose major production site was believed to reside outside pancreas, e.g. in the small intestine. We highlight the key non-classical islet peptides, and consider their involvement, together with established islet hormones, in regulation of stimulus-secretion coupling as well as proliferation, survival and transdifferentiation of β-cells. We furthermore focus on the paracrine interaction between classical and non-classical islet hormones in the maintenance of β-cell function. Understanding the functional relationships between these islet peptides might help to develop novel, more efficient treatments for diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| | - R Charlotte Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Andrei I Tarasov
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
13
|
Serra-Navarro B, Fernandez-Ruiz R, García-Alamán A, Pradas-Juni M, Fernandez-Rebollo E, Esteban Y, Mir-Coll J, Mathieu J, Dalle S, Hahn M, Ahlgren U, Weinstein LS, Vidal J, Gomis R, Gasa R. Gsα-dependent signaling is required for postnatal establishment of a functional β-cell mass. Mol Metab 2021; 53:101264. [PMID: 34091063 PMCID: PMC8239471 DOI: 10.1016/j.molmet.2021.101264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/17/2021] [Accepted: 05/30/2021] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Early postnatal life is a critical period for the establishment of the functional β-cell mass that will sustain whole-body glucose homeostasis during the lifetime. β cells are formed from progenitors during embryonic development but undergo significant expansion in quantity and attain functional maturity after birth. The signals and pathways involved in these processes are not fully elucidated. Cyclic adenosine monophosphate (cAMP) is an intracellular signaling molecule that is known to regulate insulin secretion, gene expression, proliferation, and survival of adult β cells. The heterotrimeric G protein Gs stimulates the cAMP-dependent pathway by activating adenylyl cyclase. In this study, we sought to explore the role of Gs-dependent signaling in postnatal β-cell development. METHODS To study Gs-dependent signaling, we generated conditional knockout mice in which the α subunit of the Gs protein (Gsα) was ablated from β-cells using the Cre deleter line Ins1Cre. Mice were characterized in terms of glucose homeostasis, including in vivo glucose tolerance, glucose-induced insulin secretion, and insulin sensitivity. β-cell mass was studied using histomorphometric analysis and optical projection tomography. β-cell proliferation was studied by ki67 and phospho-histone H3 immunostatining, and apoptosis was assessed by TUNEL assay. Gene expression was determined in isolated islets and sorted β cells by qPCR. Intracellular cAMP was studied in isolated islets using HTRF-based technology. The activation status of the cAMP and insulin-signaling pathways was determined by immunoblot analysis of the relevant components of these pathways in isolated islets. In vitro proliferation of dissociated islet cells was assessed by BrdU incorporation. RESULTS Elimination of Gsα in β cells led to reduced β-cell mass, deficient insulin secretion, and severe glucose intolerance. These defects were evident by weaning and were associated with decreased proliferation and inadequate expression of key β-cell identity and maturation genes in postnatal β-cells. Additionally, loss of Gsα caused a broad multilevel disruption of the insulin transduction pathway that resulted in the specific abrogation of the islet proliferative response to insulin. CONCLUSION We conclude that Gsα is required for β-cell growth and maturation in the early postnatal stage and propose that this is partly mediated via its crosstalk with insulin signaling. Our findings disclose a tight connection between these two pathways in postnatal β cells, which may have implications for using cAMP-raising agents to promote β-cell regeneration and maturation in diabetes.
Collapse
Affiliation(s)
- Berta Serra-Navarro
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Rebeca Fernandez-Ruiz
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Ainhoa García-Alamán
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Marta Pradas-Juni
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Eduardo Fernandez-Rebollo
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Yaiza Esteban
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Joan Mir-Coll
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Julia Mathieu
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, Montpellier, France
| | - Stephane Dalle
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, Montpellier, France
| | - Max Hahn
- Umeå Centre for Molecular Medicine (UCMM), Umeå, Sweden
| | - Ulf Ahlgren
- Umeå Centre for Molecular Medicine (UCMM), Umeå, Sweden
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Josep Vidal
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain; Department of Endocrinology and Nutrition, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain; Universitat Oberta de Catalunya (UOC), Barcelona, Spain
| | - Rosa Gasa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain.
| |
Collapse
|
14
|
Henquin JC. Non-glucose modulators of insulin secretion in healthy humans: (dis)similarities between islet and in vivo studies. Metabolism 2021; 122:154821. [PMID: 34174327 DOI: 10.1016/j.metabol.2021.154821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022]
Abstract
Optimal metabolic homeostasis requires precise temporal and quantitative control of insulin secretion. Both in vivo and in vitro studies have often focused on the regulation by glucose although many additional factors including other nutrients, neurotransmitters, hormones and drugs, modulate the secretory function of pancreatic β-cells. This review is based on the analysis of clinical investigations characterizing the effects of non-glucose modulators of insulin secretion in healthy subjects, and of experimental studies testing the same modulators in islets isolated from normal human donors. The aim was to determine whether the information gathered in vitro can reliably be translated to the in vivo situation. The comparison evidenced both convincing similarities and areas of discordance. The lack of coherence generally stems from the use of exceedingly high concentrations of test agents at too high or too low glucose concentrations in vitro, which casts doubts on the physiological relevance of a number of observations made in isolated islets. Future projects resorting to human islets should avoid extreme experimental conditions, such as oversized stimulations or inhibitions of β-cells, which are unlikely to throw light on normal insulin secretion and contribute to the elucidation of its defects.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
15
|
White MF, Kahn CR. Insulin action at a molecular level - 100 years of progress. Mol Metab 2021; 52:101304. [PMID: 34274528 PMCID: PMC8551477 DOI: 10.1016/j.molmet.2021.101304] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022] Open
Abstract
The discovery of insulin 100 years ago and its application to the treatment of human disease in the years since have marked a major turning point in the history of medicine. The availability of purified insulin allowed for the establishment of its physiological role in the regulation of blood glucose and ketones, the determination of its amino acid sequence, and the solving of its structure. Over the last 50 years, the function of insulin has been applied into the discovery of the insulin receptor and its signaling cascade to reveal the role of impaired insulin signaling-or resistance-in the progression of type 2 diabetes. It has also become clear that insulin signaling can impact not only classical insulin-sensitive tissues, but all tissues of the body, and that in many of these tissues the insulin signaling cascade regulates unexpected physiological functions. Despite these remarkable advances, much remains to be learned about both insulin signaling and how to use this molecular knowledge to advance the treatment of type 2 diabetes and other insulin-resistant states.
Collapse
Affiliation(s)
- Morris F White
- Boston Children's Hospital and Harvard Medical School, Boston, MA, 02215, USA.
| | - C Ronald Kahn
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
16
|
Inceptor counteracts insulin signalling in β-cells to control glycaemia. Nature 2021; 590:326-331. [DOI: 10.1038/s41586-021-03225-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022]
|
17
|
Henquin JC. Paracrine and autocrine control of insulin secretion in human islets: evidence and pending questions. Am J Physiol Endocrinol Metab 2021; 320:E78-E86. [PMID: 33103455 DOI: 10.1152/ajpendo.00485.2020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Insulin secretion by β-cells is largely controlled by circulating nutrients, hormones, and neurotransmitters. However, recent years have witnessed the multiplication of studies investigating whether local regulation also takes place within pancreatic islets, in which β-cells cohabit with several other cell types. The cell composition and architectural organization of human islets differ from those of rodent islets and are particularly favorable to cellular interactions. An impressive number of hormonal (glucagon, glucagon-like peptide-1, somatostatin, etc.) and nonhormonal products (ATP, acetylcholine, γ-aminobutyric acid, dopamine, etc.) are released by islet cells and have been implicated in a local control of insulin secretion. This review analyzes reports directly testing paracrine and autocrine control of insulin secretion in isolated human islets. Many of these studies were designed on background information collected in rodent islets. However, the perspective of the review is not to highlight species similarities or specificities but to contrast established and speculative mechanisms in human islets. It will be shown that the current evidence is convincing only for a minority of candidates for a paracrine function whereas arguments supporting a physiological role of others do not stand up to scrutiny. Several pending questions await further investigation.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium
| |
Collapse
|
18
|
Arous C, Mizgier ML, Rickenbach K, Pinget M, Bouzakri K, Wehrle-Haller B. Integrin and autocrine IGF2 pathways control fasting insulin secretion in β-cells. J Biol Chem 2020; 295:16510-16528. [PMID: 32934005 PMCID: PMC7864053 DOI: 10.1074/jbc.ra120.012957] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/09/2020] [Indexed: 12/20/2022] Open
Abstract
Elevated levels of fasting insulin release and insufficient glucose-stimulated insulin secretion (GSIS) are hallmarks of diabetes. Studies have established cross-talk between integrin signaling and insulin activity, but more details of how integrin-dependent signaling impacts the pathophysiology of diabetes are needed. Here, we dissected integrin-dependent signaling pathways involved in the regulation of insulin secretion in β-cells and studied their link to the still debated autocrine regulation of insulin secretion by insulin/insulin-like growth factor (IGF) 2-AKT signaling. We observed for the first time a cooperation between different AKT isoforms and focal adhesion kinase (FAK)-dependent adhesion signaling, which either controlled GSIS or prevented insulin secretion under fasting conditions. Indeed, β-cells form integrin-containing adhesions, which provide anchorage to the pancreatic extracellular matrix and are the origin of intracellular signaling via FAK and paxillin. Under low-glucose conditions, β-cells adopt a starved adhesion phenotype consisting of actin stress fibers and large peripheral focal adhesion. In contrast, glucose stimulation induces cell spreading, actin remodeling, and point-like adhesions that contain phospho-FAK and phosphopaxillin, located in small protrusions. Rat primary β-cells and mouse insulinomas showed an adhesion remodeling during GSIS resulting from autocrine insulin/IGF2 and AKT1 signaling. However, under starving conditions, the maintenance of stress fibers and the large adhesion phenotype required autocrine IGF2-IGF1 receptor signaling mediated by AKT2 and elevated FAK-kinase activity and ROCK-RhoA levels but low levels of paxillin phosphorylation. This starved adhesion phenotype prevented excessive insulin granule release to maintain low insulin secretion during fasting. Thus, deregulation of the IGF2 and adhesion-mediated signaling may explain dysfunctions observed in diabetes.
Collapse
Affiliation(s)
- Caroline Arous
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.
| | - Maria Luisa Mizgier
- UMR DIATHEC, Centre Européen d'Etude du Diabète, UMR DIATHEC, Strasbourg, France
| | - Katharina Rickenbach
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Michel Pinget
- UMR DIATHEC, Centre Européen d'Etude du Diabète, UMR DIATHEC, Strasbourg, France
| | - Karim Bouzakri
- UMR DIATHEC, Centre Européen d'Etude du Diabète, UMR DIATHEC, Strasbourg, France
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| |
Collapse
|
19
|
Sevrin T, Boquien CY, Gandon A, Grit I, de Coppet P, Darmaun D, Alexandre-Gouabau MC. Fenugreek Stimulates the Expression of Genes Involved in Milk Synthesis and Milk Flow through Modulation of Insulin/GH/IGF-1 Axis and Oxytocin Secretion. Genes (Basel) 2020; 11:E1208. [PMID: 33081164 PMCID: PMC7602737 DOI: 10.3390/genes11101208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/06/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
We previously demonstrated galactagogue effect of fenugreek in a rat model of lactation challenge, foreshadowing its use in women's breastfeeding management. To assess longitudinal molecular mechanisms involved in milk synthesis/secretion in dams submitted to fenugreek supplementation, inguinal mammary, pituitary glands and plasma were isolated in forty-three rats nursing large 12 pups-litters and assigned to either a control (CTL) or a fenugreek-supplemented (FEN) diet during lactation. RT-PCR were performed at days 12 and 18 of lactation (L12 and L18) and the first day of involution (Inv1) to measure the relative expression of genes related to both milk synthesis and its regulation in the mammary gland and lactogenic hormones in the pituitary gland. Plasma hormone concentrations were measured by ELISA. FEN diet induced 2- to 3-times higher fold change in relative expression of several genes related to macronutrient synthesis (Fasn, Acaca, Fabp3, B4galt1, Lalba and Csn2) and energy metabolism (Cpt1a, Acads) and in IGF-1 receptor in mammary gland, mainly at L12. Pituitary oxytocin expression and plasma insulin concentration (+77.1%) were also significantly increased. Altogether, these findings suggest fenugreek might extend duration of peak milk synthesis through modulation of the insulin/GH/IGF-1 axis and increase milk ejection by activation of oxytocin secretion.
Collapse
Affiliation(s)
- Thomas Sevrin
- FRANCE Bébé Nutrition (FBN) Laboratory, 53000 Laval, France;
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Clair-Yves Boquien
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Alexis Gandon
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Isabelle Grit
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Pierre de Coppet
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Dominique Darmaun
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
- Nantes University Hospital (CHU) Nantes, F-44000 Nantes, France
| | - Marie-Cécile Alexandre-Gouabau
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| |
Collapse
|
20
|
Nagao M, Esguerra JLS, Asai A, Ofori JK, Edlund A, Wendt A, Sugihara H, Wollheim CB, Oikawa S, Eliasson L. Potential Protection Against Type 2 Diabetes in Obesity Through Lower CD36 Expression and Improved Exocytosis in β-Cells. Diabetes 2020; 69:1193-1205. [PMID: 32198214 PMCID: PMC7243297 DOI: 10.2337/db19-0944] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/09/2020] [Indexed: 12/18/2022]
Abstract
Obesity is a risk factor for type 2 diabetes (T2D); however, not all obese individuals develop the disease. In this study, we aimed to investigate the cause of differential insulin secretion capacity of pancreatic islets from donors with T2D and non-T2D (ND), especially obese donors (BMI ≥30 kg/m2). Islets from obese donors with T2D had reduced insulin secretion, decreased β-cell exocytosis, and higher expression of fatty acid translocase CD36. We tested the hypothesis that CD36 is a key molecule in the reduced insulin secretion capacity. Indeed, CD36 overexpression led to decreased insulin secretion, impaired exocytosis, and reduced granule docking. This was accompanied by reduced expression of the exocytotic proteins SNAP25, STXBP1, and VAMP2, likely because CD36 induced downregulation of the insulin receptor substrate (IRS) proteins, suppressed the insulin-signaling phosphatidylinositol 3-kinase/AKT pathway, and increased nuclear localization of the transcription factor FoxO1. CD36 antibody treatment of the human β-cell line EndoC-βH1 increased IRS1 and exocytotic protein levels, improved granule docking, and enhanced insulin secretion. Our results demonstrate that β-cells from obese donors with T2D have dysfunctional exocytosis likely due to an abnormal lipid handling represented by differential CD36 expression. Hence, CD36 could be a key molecule to limit β-cell function in T2D associated with obesity.
Collapse
Affiliation(s)
- Mototsugu Nagao
- Department of Clinical Sciences, Malmö, Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Clinical Research Centre, Malmö, Sweden
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Jonathan L S Esguerra
- Department of Clinical Sciences, Malmö, Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Akira Asai
- Department of Clinical Sciences, Malmö, Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Clinical Research Centre, Malmö, Sweden
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Jones K Ofori
- Department of Clinical Sciences, Malmö, Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Anna Edlund
- Department of Clinical Sciences, Malmö, Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Anna Wendt
- Department of Clinical Sciences, Malmö, Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Hitoshi Sugihara
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Claes B Wollheim
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Shinichi Oikawa
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Lena Eliasson
- Department of Clinical Sciences, Malmö, Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Clinical Research Centre, Malmö, Sweden
| |
Collapse
|
21
|
Kataru RP, Park HJ, Baik JE, Li C, Shin J, Mehrara BJ. Regulation of Lymphatic Function in Obesity. Front Physiol 2020; 11:459. [PMID: 32499718 PMCID: PMC7242657 DOI: 10.3389/fphys.2020.00459] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022] Open
Abstract
The lymphatic system has many functions, including macromolecules transport, fat absorption, regulation and modulation of adaptive immune responses, clearance of inflammatory cytokines, and cholesterol metabolism. Thus, it is evident that lymphatic function can play a key role in the regulation of a wide array of biologic phenomenon, and that physiologic changes that alter lymphatic function may have profound pathologic effects. Recent studies have shown that obesity can markedly impair lymphatic function. Obesity-induced pathologic changes in the lymphatic system result, at least in part, from the accumulation of inflammatory cells around lymphatic vessel leading to impaired lymphatic collecting vessel pumping capacity, leaky initial and collecting lymphatics, alterations in lymphatic endothelial cell (LEC) gene expression, and degradation of junctional proteins. These changes are important since impaired lymphatic function in obesity may contribute to the pathology of obesity in other organ systems in a feed-forward manner by increasing low-grade tissue inflammation and the accumulation of inflammatory cytokines. More importantly, recent studies have suggested that interventions that inhibit inflammatory responses, either pharmacologically or by lifestyle modifications such as aerobic exercise and weight loss, improve lymphatic function and metabolic parameters in obese mice. The purpose of this review is to summarize the pathologic effects of obesity on the lymphatic system, the cellular mechanisms that regulate these responses, the effects of impaired lymphatic function on metabolic syndrome in obesity, and the interventions that may improve lymphatic function in obesity.
Collapse
Affiliation(s)
- Raghu P Kataru
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hyeong Ju Park
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jung Eun Baik
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Claire Li
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jinyeon Shin
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Babak J Mehrara
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
22
|
Rumala CZ, Liu J, Locasale JW, Corkey BE, Deeney JT, Rameh LE. Exposure of Pancreatic β-Cells to Excess Glucose Results in Bimodal Activation of mTORC1 and mTOR-Dependent Metabolic Acceleration. iScience 2020; 23:100858. [PMID: 32058969 PMCID: PMC7005503 DOI: 10.1016/j.isci.2020.100858] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/28/2019] [Accepted: 01/16/2020] [Indexed: 01/03/2023] Open
Abstract
Chronic exposure of pancreatic β-cells to excess glucose can lead to metabolic acceleration and loss of stimulus-secretion coupling. Here, we examined how exposure to excess glucose (defined here as concentrations above 5 mM) affects mTORC1 signaling and the metabolism of β-cells. Acute exposure to excess glucose stimulated glycolysis-dependent mTORC1 signaling, without changes in the PI3K or AMPK pathways. Prolonged exposure to excess glucose led to hyperactivation of mTORC1 and metabolic acceleration, characterized by higher basal respiration and maximal respiratory capacity, increased energy demand, and enhanced flux through mitochondrial pyruvate metabolism. Inhibition of pyruvate transport to the mitochondria decelerated the metabolism of β-cells chronically exposed to excess glucose and re-established glucose-dependent mTORC1 signaling, disrupting a positive feedback loop for mTORC1 hyperactivation. mTOR inhibition had positive and negative impacts on various metabolic pathways and insulin secretion, demonstrating a role for mTOR signaling in the long-term metabolic adaptation of β-cells to excess glucose. Acute glucose stimulates mTORC1 in β-cells through a glycolysis-dependent mechanism Chronic excess glucose promotes mTOR-dependent metabolic acceleration of β-cells Metabolic acceleration activates a positive feedback loop for mTORC1 hyperactivation mTOR hyperactivation disturbs the metabolism and insulin secretion patterns of β-cells
Collapse
Affiliation(s)
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Jason Wei Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Barbara Ellen Corkey
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jude Thaddeus Deeney
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Lucia Egydio Rameh
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
23
|
Hou G, Tang Y, Ren L, Guan Y, Hou X, Song G. The ANGPTL8 rs2278426 (C/T) Polymorphism Is Associated with Prediabetes and Type 2 Diabetes in a Han Chinese Population in Hebei Province. Int J Endocrinol 2020; 2020:1621239. [PMID: 33343659 PMCID: PMC7728483 DOI: 10.1155/2020/1621239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/14/2020] [Accepted: 11/23/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Our aim was to investigate the association between the genetics of the angiopoietin protein-like 8 (ANGPTL8) rs2278426 (C/T) polymorphism with prediabetes (pre-DM) and type 2 diabetes (T2DM) in a Han Chinese population in Hebei Province, China. METHODS We enrolled 1,460 participants into this case-control study: healthy controls, n = 524; pre-DM, n = 460; and T2DM: n = 460. Ligase assays on blood samples from all participants were used to identify polymorphisms. Differences in genotype and allele distributions were compared by the chi-square test and one-way analysis of variance, and a post hoc pairwise analysis was performed using the Bonferroni test. The logistic regression technique was adjusted for age, sex, and body mass index. RESULTS The frequency of the TT (10.9%) genotype was significantly higher in pre-DM patients than in controls (odds ratio [OR] = 1.696, 95% confidence interval [CI] = 1.026-2.802, P=0.039). In the T2DM group, the CT (48%) and TT (15%) genotypes were significantly higher compared with those in the control group (CT : OR = 1.384, 95% CI = 1.013-1.890, P=0.041; TT : OR = 2.530, 95% CI = 1.476-4.334, P=0.001). The frequency of the T allele was significantly higher in the pre-DM (32.8%) and T2DM (39%) groups compared with the control group (26.9%) and was significantly associated with an increased risk of pre-DM (OR = 1.253, 95% CI = 1.017-1.544, P=0.034) and T2DM (OR = 1.518, 95% CI = 1.214-1.897, P=0.001). Furthermore, insulin levels in the pre-DM and T2DM groups were significantly decreased in those with the TT genotype compared with the CC and CT genotypes. CONCLUSION ANGPTL8 rs2278426 may be involved in the mechanism of insulin secretion and could lead to an increased risk of pre-DM and T2DM.
Collapse
Affiliation(s)
- Guangsen Hou
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Yong Tang
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Luping Ren
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Yunpeng Guan
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Xiaoyu Hou
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| |
Collapse
|
24
|
Ježek P, Jabůrek M, Plecitá-Hlavatá L. Contribution of Oxidative Stress and Impaired Biogenesis of Pancreatic β-Cells to Type 2 Diabetes. Antioxid Redox Signal 2019; 31:722-751. [PMID: 30450940 PMCID: PMC6708273 DOI: 10.1089/ars.2018.7656] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022]
Abstract
Significance: Type 2 diabetes development involves multiple changes in β-cells, related to the oxidative stress and impaired redox signaling, beginning frequently by sustained overfeeding due to the resulting lipotoxicity and glucotoxicity. Uncovering relationships among the dysregulated metabolism, impaired β-cell "well-being," biogenesis, or cross talk with peripheral insulin resistance is required for elucidation of type 2 diabetes etiology. Recent Advances: It has been recognized that the oxidative stress, lipotoxicity, and glucotoxicity cannot be separated from numerous other cell pathology events, such as the attempted compensation of β-cell for the increased insulin demand and dynamics of β-cell biogenesis and its "reversal" at dedifferentiation, that is, from the concomitantly decreasing islet β-cell mass (also due to transdifferentiation) and low-grade islet or systemic inflammation. Critical Issues: At prediabetes, the compensation responses of β-cells, attempting to delay the pathology progression-when exaggerated-set a new state, in which a self-checking redox signaling related to the expression of Ins gene expression is impaired. The resulting altered redox signaling, diminished insulin secretion responses to various secretagogues including glucose, may lead to excretion of cytokines or chemokines by β-cells or excretion of endosomes. They could substantiate putative stress signals to the periphery. Subsequent changes and lasting glucolipotoxicity promote islet inflammatory responses and further pathology spiral. Future Directions: Should bring an understanding of the β-cell self-checking and related redox signaling, including the putative stress signal to periphery. Strategies to cure or prevent type 2 diabetes could be based on the substitution of the "wrong" signal by the "correct" self-checking signal.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Jabůrek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
25
|
Hasannejad M, Samsamshariat SZ, Esmaili A, Jahanian-Najafabadi A. Klotho induces insulin resistance possibly through interference with GLUT4 translocation and activation of Akt, GSK3β, and PFKfβ3 in 3T3-L1 adipocyte cells. Res Pharm Sci 2019; 14:369-377. [PMID: 31516514 PMCID: PMC6714116 DOI: 10.4103/1735-5362.263627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Klotho is considered as an anti-aging factor inducing insulin resistance and involved in type 2 diabetes. However, mechanisms by which klotho induces insulin resistance remain to be understood. Thus, in this study, we aimed to evaluate possible interference points of klotho with insulin signaling pathways in 3T3-L1 adipocyte cells by focusing on phosphorylation levels of Akt, GSK3β, PFK-fβ3, and GLUT4 translocation. Differentiation of 3T3-L1 cells to the adipocyte-like cells were performed using specific differentiation kit and confirmed by mRNA expression assay of PPARγ using qRT-PCR, and Sudan black staining of lipid droplets. Then cells were co-treated with klotho and insulin. Expression and translocation of GLUT4 mRNA were evaluated using qRT-PCR and Alexa flour 488 conjugated GLUT4 antibody, respectively. P-Akt/Akt, p-GSK3β/GSK3β, and p-PFKfβ3/PFKfβ3 ratios were determined in insulin and klotho/insulin treated cells using western blot. Our result indicated that GLUT4 expression were decreased to 0.72 ± 0.16 fold in insulin treated cells, however it was calculated 1.12 ± 0.25 fold in klotho/insulin treated cells. In addition, klotho prevented GLUT4 membrane translocation by 27.2% in comparison with insulin-treated cells (P < 0.05). Interestingly, in insulin/klotho co-treated cells, phospho-levels of Akt, GSK3β, and PFKfβ3 proteins was decreased to 2.34 ± 0.14, 2.29 ± 0.63, and 1.95 ± 0.37 fold in comparison with the insulin cells, (P < 0.05). In conclusion, our study indicated that klotho induces insulin resistance in adipocytes possibly through prevention of GLUT4 translocation, and interfere with phosphorylation of Akt, GSK3β, and PFKf3β intracellular signaling mediators by insulin.
Collapse
Affiliation(s)
- Mohamad Hasannejad
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Seyed Ziaaldin Samsamshariat
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Armita Esmaili
- Department of Pathology, Mehrgan hospital, Kerman University of Medical Sciences and Health services, Kerman, I.R. Iran
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
26
|
Keller MP, Rabaglia ME, Schueler KL, Stapleton DS, Gatti DM, Vincent M, Mitok KA, Wang Z, Ishimura T, Simonett SP, Emfinger CH, Das R, Beck T, Kendziorski C, Broman KW, Yandell BS, Churchill GA, Attie AD. Gene loci associated with insulin secretion in islets from non-diabetic mice. J Clin Invest 2019; 129:4419-4432. [PMID: 31343992 DOI: 10.1172/jci129143] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Genetic susceptibility to type 2 diabetes is primarily due to β-cell dysfunction. However, a genetic study to directly interrogate β-cell function ex vivo has never been previously performed. We isolated 233,447 islets from 483 Diversity Outbred (DO) mice maintained on a Western-style diet, and measured insulin secretion in response to a variety of secretagogues. Insulin secretion from DO islets ranged >1,000-fold even though none of the mice were diabetic. The insulin secretory response to each secretagogue had a unique genetic architecture; some of the loci were specific for one condition, whereas others overlapped. Human loci that are syntenic to many of the insulin secretion QTL from mouse are associated with diabetes-related SNPs in human genome-wide association studies. We report on three genes, Ptpn18, Hunk and Zfp148, where the phenotype predictions from the genetic screen were fulfilled in our studies of transgenic mouse models. These three genes encode a non-receptor type protein tyrosine phosphatase, a serine/threonine protein kinase, and a Krϋppel-type zinc-finger transcription factor, respectively. Our results demonstrate that genetic variation in insulin secretion that can lead to type 2 diabetes is discoverable in non-diabetic individuals.
Collapse
Affiliation(s)
- Mark P Keller
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Mary E Rabaglia
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Kathryn L Schueler
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Donnie S Stapleton
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | | | | | - Kelly A Mitok
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Ziyue Wang
- University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, Madison, Wisconsin, USA
| | | | - Shane P Simonett
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | | | - Rahul Das
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Tim Beck
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christina Kendziorski
- University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, Madison, Wisconsin, USA
| | - Karl W Broman
- University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, Madison, Wisconsin, USA
| | - Brian S Yandell
- University of Wisconsin-Madison, Department of Horticulture, Madison, Wisconsin, USA
| | | | - Alan D Attie
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| |
Collapse
|
27
|
Abstract
Controlling the excess and shortage of energy is a fundamental task for living organisms. Diabetes is a representative metabolic disease caused by the malfunction of energy homeostasis. The islets of Langerhans in the pancreas release long-range messengers, hormones, into the blood to regulate the homeostasis of the primary energy fuel, glucose. The hormone and glucose levels in the blood show rhythmic oscillations with a characteristic period of 5-10 min, and the functional roles of the oscillations are not clear. Each islet has [Formula: see text] and [Formula: see text] cells that secrete glucagon and insulin, respectively. These two counter-regulatory hormones appear sufficient to increase and decrease glucose levels. However, pancreatic islets have a third cell type, [Formula: see text] cells, which secrete somatostatin. The three cell populations have a unique spatial organization in islets, and they interact to perturb their hormone secretions. The mini-organs of islets are scattered throughout the exocrine pancreas. Considering that the human pancreas contains approximately a million islets, the coordination of hormone secretion from the multiple sources of islets and cells within the islets should have a significant effect on human physiology. In this review, we introduce the hierarchical organization of tripartite cell networks, and recent biophysical modeling to systematically understand the oscillations and interactions of [Formula: see text], [Formula: see text], and [Formula: see text] cells. Furthermore, we discuss the functional roles and clinical implications of hormonal oscillations and their phase coordination for the diagnosis of type II diabetes.
Collapse
Affiliation(s)
- Taegeun Song
- Department of Physics, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | | |
Collapse
|
28
|
Santo-Domingo J, Galindo AN, Cominetti O, De Marchi U, Cutillas P, Dayon L, Wiederkehr A. Glucose-dependent phosphorylation signaling pathways and crosstalk to mitochondrial respiration in insulin secreting cells. Cell Commun Signal 2019; 17:14. [PMID: 30786936 PMCID: PMC6381748 DOI: 10.1186/s12964-019-0326-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 02/08/2019] [Indexed: 01/13/2023] Open
Abstract
Background Glucose is the main secretagogue of pancreatic beta-cells. Uptake and metabolism of the nutrient stimulates the beta-cell to release the blood glucose lowering hormone insulin. This metabolic activation is associated with a pronounced increase in mitochondrial respiration. Glucose stimulation also initiates a number of signal transduction pathways for the coordinated regulation of multiple biological processes required for insulin secretion. Methods Shotgun proteomics including TiO2 enrichment of phosphorylated peptides followed by liquid chromatography tandem mass spectrometry on lysates from glucose-stimulated INS-1E cells was used to identify glucose regulated phosphorylated proteins and signal transduction pathways. Kinase substrate enrichment analysis (KSEA) was applied to identify key regulated kinases and phosphatases. Glucose-induced oxygen consumption was measured using a XF96 Seahorse instrument to reveal cross talk between glucose-regulated kinases and mitochondrial activation. Results Our kinetic analysis of substrate phosphorylation reveal the molecular mechanism leading to rapid activation of insulin biogenesis, vesicle trafficking, insulin granule exocytosis and cytoskeleton remodeling. Kinase-substrate enrichment identified upstream kinases and phosphatases and time-dependent activity changes during glucose stimulation. Activity trajectories of well-known glucose-regulated kinases and phosphatases are described. In addition, we predict activity changes in a number of kinases including NUAK1, not or only poorly studied in the context of the pancreatic beta-cell. Furthermore, we pharmacologically tested whether signaling pathways predicted by kinase-substrate enrichment analysis affected glucose-dependent acceleration of mitochondrial respiration. We find that phosphoinositide 3-kinase, Ca2+/calmodulin dependent protein kinase and protein kinase C contribute to short-term regulation of energy metabolism. Conclusions Our results provide a global view into the regulation of kinases and phosphatases in insulin secreting cells and suggest cross talk between glucose-induced signal transduction and mitochondrial activation. Electronic supplementary material The online version of this article (10.1186/s12964-019-0326-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jaime Santo-Domingo
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park Bâtiment G, 1015, Lausanne, Switzerland.
| | - Antonio Núñez Galindo
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park Bâtiment G, 1015, Lausanne, Switzerland
| | - Ornella Cominetti
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park Bâtiment G, 1015, Lausanne, Switzerland
| | - Umberto De Marchi
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park Bâtiment G, 1015, Lausanne, Switzerland
| | - Pedro Cutillas
- Analytical Signalling Group, Centre for Cell Signalling, Queen Mary University of London, London, UK
| | - Loïc Dayon
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park Bâtiment G, 1015, Lausanne, Switzerland
| | - Andreas Wiederkehr
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park Bâtiment G, 1015, Lausanne, Switzerland
| |
Collapse
|
29
|
Liang H, Nie J, Van Skike CE, Valentine JM, Orr ME. Mammalian Target of Rapamycin at the Crossroad Between Alzheimer's Disease and Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:185-225. [PMID: 31062331 DOI: 10.1007/978-981-13-3540-2_10] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that Alzheimer's disease may manifest as a metabolic disorder with pathology and/or dysfunction in numerous tissues. Adults with Alzheimer's disease suffer with significantly more comorbidities than demographically matched Medicare beneficiaries (Zhao et al, BMC Health Serv Res 8:108, 2008b). Reciprocally, comorbid health conditions increase the risk of developing Alzheimer's disease (Haaksma et al, PLoS One 12(5):e0177044, 2017). Type 2 diabetes mellitus is especially notable as the disease shares many overlapping pathologies observed in patients with Alzheimer's disease, including hyperglycemia, hyperinsulinemia, insulin resistance, glucose intolerance, dyslipidemia, inflammation, and cognitive dysfunction, as described in Chap. 8 of this book (Yoshitake et al, Neurology 45(6):1161-1168, 1995; Leibson et al, Am J Epidemiol 145(4):301-308, 1997; Ott et al, Neurology 53(9):1937-1942, 1999; Voisin et al, Rev Med Interne 24(Suppl 3):288s-291s, 2003; Janson et al. Diabetes 53(2):474-481, 2004; Ristow M, J Mol Med (Berl) 82(8):510-529, 2004; Whitmer et al, BMJ 330(7504):1360, 2005, Curr Alzheimer Res 4(2):103-109, 2007; Ohara et al, Neurology 77(12):1126-1134, 2011). Although nondiabetic older adults also experience age-related cognitive decline, diabetes is uniquely associated with a twofold increased risk of Alzheimer's disease, as described in Chap. 2 of this book (Yoshitake et al, Neurology 45(6):1161-1168, 1995; Leibson et al, Am J Epidemiol 145(4):301-308, 1997; Ott et al. Neurology 53(9):1937-1942, 1999; Ohara et al, Neurology 77(12):1126-1134, 2011). Good glycemic control has been shown to improve cognitive status (Cukierman-et al, Diabetes Care 32(2):221-226, 2009), and the use of insulin sensitizers is correlated with a lower rate of cognitive decline in older adults (Morris JK, Burns JM, Curr Neurol Neurosci Rep 12(5):520-527, 2012). At the molecular level, the mechanistic/mammalian target of rapamycin (mTOR) plays a key role in maintaining energy homeostasis. Nutrient availability and cellular stress information, both extracellular and intracellular, are integrated and transduced through mTOR signaling pathways. Aberrant regulation of mTOR occurs in the brains of patients with Alzheimer's disease and in numerous tissues of individuals with type 2 diabetes (Mannaa et al, J Mol Med (Berl) 91(10):1167-1175, 2013). Moreover, modulating mTOR activity with a pharmacological inhibitor, rapamycin, provides wide-ranging health benefits, including healthy life span extension in numerous model organisms (Vellai et al, Nature 426(6967):620, 2003; Jia et al, Development 131(16):3897-3906, 2004; Kapahi et al, Curr Biol 14(10):885-890, 2004; Kaeberlein et al, Science 310(5751):1193-1196, 2005; Powers et al, Genes Dev 20(2):174-184, 2006; Harrison et al, Nature 460(7253):392-395, 2009; Selman et al, Science 326(5949):140-144, 2009; Sharp ZD, Strong R, J Gerontol A Biol Sci Med Sci 65(6):580-589, 2010), which underscores its importance to overall organismal health and longevity. In this chapter, we discuss the physiological role of mTOR signaling and the consequences of mTOR dysregulation in the brain and peripheral tissues, with emphasis on its relevance to the development of Alzheimer's disease and link to type 2 diabetes.
Collapse
Affiliation(s)
- Hanyu Liang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jia Nie
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Candice E Van Skike
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Joseph M Valentine
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Miranda E Orr
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- San Antonio Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, San Antonio, TX, USA.
| |
Collapse
|
30
|
Reduced expression of Twist 1 is protective against insulin resistance of adipocytes and involves mitochondrial dysfunction. Sci Rep 2018; 8:12590. [PMID: 30135600 PMCID: PMC6105588 DOI: 10.1038/s41598-018-30820-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/07/2018] [Indexed: 01/06/2023] Open
Abstract
Insulin resistance (IR) has become a global epidemic that represents a serious hazard to public health. However, the precise mechanisms modulating IR have not been fully elucidated. The present study aimed to investigate the role of transcriptional factor Twist 1 in adipocyte IR and to further explore the molecular mechanism. An in vitro IR model based on cultured 3T3-L1 adipocytes was established under high glucose/insulin stimulation and an in vivo IR model in C57/BL6J mice induced by a high fat diet (HFD) was also developed. Lentivirus targeting Twist 1 silencing was introduced. The relationships between Twist 1 expression and IR state, mitochondrial dysfunction and the downstream insulin signaling pathway were assayed. Our results firstly showed the elevation of Twist 1 in IR adipocytes, and Twist 1 silencing attenuated IR. Then mitochondrial ultra-structural damage, elevated ROS, decreased MMP and ATP, and changes in mitochondrial biosynthesis-related genes in IR group indicated mitochondrial dysfunction. Further, the downstream IRS/PI3K/AKT/GluT4 pathway was showed involved in Twist 1-mediated IR. In total, we provide evidence of a protective role of Twist 1 silencing in relieving the IR state of adipocytes. Mitochondrial dysfunction and the downstream IRS/PI3K/AKT/GluT4 pathway were involved in this Twist 1-mediated IR.
Collapse
|
31
|
Kitamoto T, Sakurai K, Lee EY, Yokote K, Accili D, Miki T. Distinct roles of systemic and local actions of insulin on pancreatic β-cells. Metabolism 2018; 82:100-110. [PMID: 29320716 PMCID: PMC7391221 DOI: 10.1016/j.metabol.2017.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/16/2017] [Accepted: 12/31/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Pancreatic β-cell mass and function are critical in glucose homeostasis. Their regulatory mechanisms have been studied principally under experimental conditions of reduced β-cell numbers, such as β-cell ablation and partial pancreatectomy. In the present study, we generated an opposite mouse model with an excessive amount of ectopic β-cells, and analyzed its consequence on β-cell mass and survival. METHODS Mice underwent sub-renal transplantation (SRT) of pseudo-islets generated from a pancreatic β-cell line MIN6 or intra-pancreatic transplantation (IPT) of MIN6 cells, and morphological and functional changes of their endocrine pancreata were analyzed. Cellular fate of pancreatic β-cells after transplantation was traced using RipCre:Rosa26-tdTomato mice. By using MIN6 cells, we evaluated the roles of extracellular glucose, membrane potential, and insulin signaling on β-cell survival. RESULTS SRT mice developed severe, progressive hypoglycemia associated with marked reduction in insulin-positive (Ins+) cell mass and apparent increase in apoptotic Ins+ cells. In in vitro experiments of MIN6 cells, insulin signaling blockade potently induced cell death, suggesting that local insulin action is required for β-cell survival. In fact, IPT (i.e. transplantation close to endogenous β-cells) resulted in fewer apoptotic Ins+ cells compared with those induced by SRT. On the other hand, β-cell mass was decreased in proportion to the decrease in blood glucose levels in both SRT and IPT mice, suggesting a contribution of hypoglycemia induced by systemic hyperinsulinemia. CONCLUSION Insulin plays distinct roles in β-cell survival and β-cell mass regulation through its local and systemic actions on β-cells, respectively.
Collapse
Affiliation(s)
- Takumi Kitamoto
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan; Department of Clinical Cell Biology and Medicine, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan; Division of Endocrinology, Department of Medicine, Columbia University, New York 10032, USA
| | - Kenichi Sakurai
- Center for Preventive Medical Sciences, Chiba University, Chiba 263-8522, Japan
| | - Eun Young Lee
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Domenico Accili
- Division of Endocrinology, Department of Medicine, Columbia University, New York 10032, USA
| | - Takashi Miki
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan.
| |
Collapse
|
32
|
Torii S, Kubota C, Saito N, Kawano A, Hou N, Kobayashi M, Torii R, Hosaka M, Kitamura T, Takeuchi T, Gomi H. The pseudophosphatase phogrin enables glucose-stimulated insulin signaling in pancreatic β cells. J Biol Chem 2018; 293:5920-5933. [PMID: 29483197 DOI: 10.1074/jbc.ra117.000301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/14/2018] [Indexed: 12/18/2022] Open
Abstract
Autocrine insulin signaling is critical for pancreatic β-cell growth and activity and is at least partially controlled by protein-tyrosine phosphatases (PTPs) that act on insulin receptors (IRs). The receptor-type PTP phogrin primarily localizes on insulin secretory granules in pancreatic β cells. We recently reported that phogrin knockdown decreases the protein levels of insulin receptor substrate 2 (IRS2), whereas high-glucose stimulation promotes formation of a phogrin-IR complex that stabilizes IRS2. However, the underlying molecular mechanisms by which phogrin affects IRS2 levels are unclear. Here, we found that relative to wildtype mice, IRS2 levels in phogrin-knockout mice islets decreased by 44%. When phogrin was silenced by shRNA in pancreatic β-cell lines, glucose-induced insulin signaling led to proteasomal degradation of IRS2 via a negative feedback mechanism. Phogrin overexpression in a murine hepatocyte cell line consistently prevented chronic insulin treatment-induced IRS2 degradation. In vitro, phogrin directly bound the IR without the assistance of other proteins and protected recombinant PTP1B from oxidation to potentiate its activity toward the IR. Furthermore, phogrin expression suppressed insulin-induced local generation of hydrogen peroxide and subsequent PTP1B oxidation, which allowed progression of IR dephosphorylation. Together, these results suggest that a transient interaction of phogrin with the IR enables glucose-stimulated autocrine insulin signaling through the regulation of PTP1B activity, which is essential for suppressing feedback-mediated IRS2 degradation in pancreatic β cells.
Collapse
Affiliation(s)
| | | | | | | | - Ni Hou
- From the Biosignal Research Center and
| | - Masaki Kobayashi
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma 371-8512, Japan
| | | | - Masahiro Hosaka
- the Department of Biotechnology, Akita Prefectural University, Akita 010-0195, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma 371-8512, Japan
| | - Toshiyuki Takeuchi
- From the Biosignal Research Center and.,the Administration Office, Gunma University, Maebashi, Gunma 371-8512, Japan, and
| | - Hiroshi Gomi
- the Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa 252-8510, Japan
| |
Collapse
|
33
|
Ardestani A, Lupse B, Kido Y, Leibowitz G, Maedler K. mTORC1 Signaling: A Double-Edged Sword in Diabetic β Cells. Cell Metab 2018; 27:314-331. [PMID: 29275961 DOI: 10.1016/j.cmet.2017.11.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/11/2017] [Accepted: 11/15/2017] [Indexed: 12/21/2022]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a central regulator of metabolic and nutrient cues that integrates environmental inputs into downstream signaling pathways to control cellular metabolism, growth, and survival. While numerous in vitro and in vivo studies reported the positive functions of mTORC1 in the regulation of β cell survival and proliferation under physiological conditions, more recent work demonstrates the opposite in the long term; this is exemplified by the constitutive inappropriate hyper-activation of mTORC1 in diabetic islets or β cells under conditions of increased β cell stress and metabolic demands. These recent findings uncover mTORC1's importance as an emerging significant player in the development and progression of β cell failure in type 2 diabetes and suggest that mTORC1 may act as a "double edge sword" in the regulation of β cell mass and function in response to metabolic stress such as nutrient overload and insulin resistance.
Collapse
Affiliation(s)
- Amin Ardestani
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany.
| | - Blaz Lupse
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe 654-0142, Japan
| | - Gil Leibowitz
- Endocrinology and Metabolism Service and the Hadassah Diabetes Unit, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Kathrin Maedler
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany.
| |
Collapse
|
34
|
Ramzy A, Mojibian M, Kieffer TJ. Insulin-Deficient Mouse β-Cells Do Not Fully Mature but Can Be Remedied Through Insulin Replacement by Islet Transplantation. Endocrinology 2018; 159:83-102. [PMID: 29029025 DOI: 10.1210/en.2017-00263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 09/22/2017] [Indexed: 12/17/2022]
Abstract
Insulin receptor (IR) insufficiency in β-cells leads to impaired insulin secretion and reduced β-cell hyperplasia in response to hyperglycemia. Selective IR deficiency in β-cells in later embryological development may lead to compensatory β-cell hyperplasia. Although these findings suggest insulin signaling on the β-cell is important for β-cell function, they are confounded by loss of signaling by the insulinlike growth factors through the IR. To determine whether insulin itself is necessary for β-cell development and maturation, we performed a characterization of pancreatic islets in mice with deletions of both nonallelic insulin genes (Ins1-/-Ins2-/-). We immunostained neonatal Ins1-/-Ins2-/- and Ins1+/+Ins2+/+ pancreata and performed quantitative polymerase chain reaction on isolated neonatal islets. Insulin-deficient islets had reduced expression of factors normally expressed in maturing β-cells, including muscoloaponeurotic fibrosarcoma oncogene homolog A, homeodomain transcription factor 6.1, and glucose transporter 2. Ins1-/-Ins2-/-β-cells expressed progenitor factors associated with stem cells or dedifferentiated β-cells, including v-myc avian myolocytomatosis viral oncogene lung carcinoma derived and homeobox protein NANOG. We replaced insulin by injection or islet transplantation to keep mice alive into adulthood to determine whether insulin replacement was sufficient for the completed maturation of insulin-deficient β-cells. Short-term insulin glargine (Lantus®) injections partially rescued the β-cell phenotype, whereas long-term replacement of insulin by isogenic islet transplantation supported the formation of more mature β-cells. Our findings suggest that tightly regulated glycemia, insulin species, or other islet factors are necessary for β-cell maturation.
Collapse
Affiliation(s)
- Adam Ramzy
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Majid Mojibian
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
35
|
Nagy L, Márton J, Vida A, Kis G, Bokor É, Kun S, Gönczi M, Docsa T, Tóth A, Antal M, Gergely P, Csóka B, Pacher P, Somsák L, Bai P. Glycogen phosphorylase inhibition improves beta cell function. Br J Pharmacol 2018; 175:301-319. [PMID: 28409826 PMCID: PMC5758390 DOI: 10.1111/bph.13819] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Glycogen phosphorylase (GP) is the key enzyme for glycogen degradation. GP inhibitors (GPi-s) are glucose lowering agents that cause the accumulation of glucose in the liver as glycogen. Glycogen metabolism has implications in beta cell function. Glycogen degradation can maintain cellular glucose levels, which feeds into catabolism to maintain insulin secretion, and elevated glycogen degradation levels contribute to glucotoxicity. The purpose of this study was to assess whether influencing glycogen metabolism in beta cells by GPi-s affects the function of these cells. EXPERIMENTAL APPROACH The effects of structurally different GPi-s were investigated on MIN6 insulinoma cells and in a mouse model of diabetes. KEY RESULTS GPi treatment increased glycogen content and, consequently, the surface area of glycogen in MIN6 cells. Furthermore, GPi treatment induced insulin receptor β (InsRβ), Akt and p70S6K phosphorylation, as well as pancreatic and duodenal homeobox 1(PDX1) and insulin expression. In line with these findings, GPi-s enhanced non-stimulated and glucose-stimulated insulin secretion in MIN6 cells. The InsRβ was shown to co-localize with glycogen particles as confirmed by in silico screening, where components of InsR signalling were identified as glycogen-bound proteins. GPi-s also activated the pathway of insulin secretion, indicated by enhanced glycolysis, mitochondrial oxidation and calcium signalling. Finally, GPi-s increased the size of islets of Langerhans and improved glucose-induced insulin release in mice. CONCLUSION AND IMPLICATIONS These data suggest that GPi-s also target beta cells and can be repurposed as agents to preserve beta cell function or even ameliorate beta cell dysfunction in different forms of diabetes. LINKED ARTICLES This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.
Collapse
Affiliation(s)
- Lilla Nagy
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- MTA‐DE Cell Biology and Signaling Research GroupDebrecenHungary
| | - Judit Márton
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - András Vida
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- MTA‐DE Lendület Laboratory of Cellular MetabolismDebrecenHungary
| | - Gréta Kis
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Éva Bokor
- Department of Organic Chemistry, Faculty of Science and TechnologyUniversity of DebrecenDebrecenHungary
| | - Sándor Kun
- Department of Organic Chemistry, Faculty of Science and TechnologyUniversity of DebrecenDebrecenHungary
| | - Mónika Gönczi
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Tibor Docsa
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Attila Tóth
- Department of Cardiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- MTA‐DE Neuroscience Research GroupDebrecenHungary
| | - Pál Gergely
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Balázs Csóka
- Department of SurgeryRutgers – New Jersey Medical SchoolNewarkNJUSA
- Center for Immunity and InflammationRutgers – New Jersey Medical SchoolNewarkNJUSA
| | - Pal Pacher
- NIAAA, National Institutes of Health, Laboratory of Physiologic StudiesRockvilleMDUSA
| | - László Somsák
- Department of Organic Chemistry, Faculty of Science and TechnologyUniversity of DebrecenDebrecenHungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- MTA‐DE Lendület Laboratory of Cellular MetabolismDebrecenHungary
- Research Center for Molecular MedicineUniversity of DebrecenDebrecenHungary
| |
Collapse
|
36
|
Skelin Klemen M, Dolenšek J, Slak Rupnik M, Stožer A. The triggering pathway to insulin secretion: Functional similarities and differences between the human and the mouse β cells and their translational relevance. Islets 2017; 9:109-139. [PMID: 28662366 PMCID: PMC5710702 DOI: 10.1080/19382014.2017.1342022] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In β cells, stimulation by metabolic, hormonal, neuronal, and pharmacological factors is coupled to secretion of insulin through different intracellular signaling pathways. Our knowledge about the molecular machinery supporting these pathways and the patterns of signals it generates comes mostly from rodent models, especially the laboratory mouse. The increased availability of human islets for research during the last few decades has yielded new insights into the specifics in signaling pathways leading to insulin secretion in humans. In this review, we follow the most central triggering pathway to insulin secretion from its very beginning when glucose enters the β cell to the calcium oscillations it produces to trigger fusion of insulin containing granules with the plasma membrane. Along the way, we describe the crucial building blocks that contribute to the flow of information and focus on their functional role in mice and humans and on their translational implications.
Collapse
Affiliation(s)
- Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Institute of Physiology; Center for Physiology and Pharmacology; Medical University of Vienna; Vienna, Austria
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
37
|
Abu-Farha M, Abubaker J, Tuomilehto J. ANGPTL8 (betatrophin) role in diabetes and metabolic diseases. Diabetes Metab Res Rev 2017; 33. [PMID: 28722798 DOI: 10.1002/dmrr.2919] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 05/30/2017] [Accepted: 07/04/2017] [Indexed: 12/22/2022]
Abstract
Diabetes is a major disease worldwide that is reaching epidemic levels. Its increased prevalence as well as its association with a high number of complications such as cardiovascular diseases, nephropathy, and retinopathy makes it an important disease for investigation. ANGPTL8 is a recently identified hormone that has been associated with two functionally important processes in the development of type 2 diabetes, insulin resistance as well as lipid metabolism. Initial work has shown that ANGPTL8 was expressed in liver, white adipose, and brown adipose tissues. ANGPTL8 regulates the activity of lipoprotein lipase, which is a key enzyme in lipoprotein lipolysis pathway through its direct interaction with ANGPTL3. It has been also reported that it regulates the replication of β-cells in response to insulin resistance. As a result, many recent studies have focused on the association of ANGPTL8 with diabetes and obesity as well as its association with various metabolic markers in order to better understand its physiological role in glucose homeostasis and lipid metabolism. In this review, we will highlight some of the key clinical findings, mainly from human studies, that investigated the role of ANGPTL8 in metabolic diseases such as diabetes, obesity, and the metabolic syndrome.
Collapse
Affiliation(s)
- Mohamed Abu-Farha
- Biochemistry and Molecular Biology Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Jehad Abubaker
- Biochemistry and Molecular Biology Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | |
Collapse
|
38
|
Moullé VS, Ghislain J, Poitout V. Nutrient regulation of pancreatic β-cell proliferation. Biochimie 2017; 143:10-17. [PMID: 28987628 DOI: 10.1016/j.biochi.2017.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022]
Abstract
Excess consumption of energy-dense foods combined with a sedentary lifestyle is driving an obesity epidemic. Although obesity is closely associated with insulin resistance, most individuals meet the insulin demand by increasing their functional β-cell mass. Those who eventually develop type 2 diabetes are distinguished by a failure in this compensatory process. Although a causal role of insulin resistance in compensatory β-cell responses has received considerable experimental support, precisely how the β cell senses changes in the metabolic environment is still unknown. As metabolism of glucose, lipids and amino acids is profoundly altered in obesity, it is not surprising that these nutrients are conspicuous among the factors proposed to contribute. In this review we summarise our understanding of the role of nutrients, in particular glucose, fatty acids and amino acids in β-cell compensation with a particular emphasis on their relation to insulin resistance-induced factors and their underlying mechanism of action. Finally, we describe the concept of epigenetic programming and review recent studies illustrating how the status of the β cell epigenome is a product of its nutrient environment, and how metabolic programming of the β cell contributes to diabetes risk.
Collapse
Affiliation(s)
- Valentine S Moullé
- Montreal Diabetes Research Center, University of Montreal, QC, Canada; CRCHUM, University of Montreal, QC, Canada.
| | - Julien Ghislain
- Montreal Diabetes Research Center, University of Montreal, QC, Canada; CRCHUM, University of Montreal, QC, Canada.
| | - Vincent Poitout
- Montreal Diabetes Research Center, University of Montreal, QC, Canada; CRCHUM, University of Montreal, QC, Canada; Department of Medicine, University of Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, QC, Canada.
| |
Collapse
|
39
|
Wang S, Wu J, Wang N, Zeng L, Wu Y. The role of growth hormone receptor in β cell function. Growth Horm IGF Res 2017; 36:30-35. [PMID: 28915386 DOI: 10.1016/j.ghir.2017.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/10/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022]
Abstract
Growth hormone (GH) exerts numerous effects on tissues through binding to its receptor, GHR, which resides on cell membranes in many different organs and tissues. Endocrine pancreatic β cells are the only source of insulin secretion in response to metabolic demand, thereby regulating blood glucose and maintaining metabolic homeostasis. β cell dysfunction is the main composition of diabetes mellitus. Numerous studies have provided strong evidence that GHR signaling plays an independent role in β cell function. In this review, we focus on the role of GHR signaling in β cell actions and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Shuang Wang
- Institute of Genome Engineered Animal Models for Human Diseases, Dalian Medical University, Dalian 116044, China
| | - Jin Wu
- Institute of Genome Engineered Animal Models for Human Diseases, Dalian Medical University, Dalian 116044, China
| | - Ning Wang
- Institute of Genome Engineered Animal Models for Human Diseases, Dalian Medical University, Dalian 116044, China
| | - Li Zeng
- Institute of Genome Engineered Animal Models for Human Diseases, Dalian Medical University, Dalian 116044, China.
| | - Yingjie Wu
- Institute of Genome Engineered Animal Models for Human Diseases, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
40
|
Lee B, Song T, Lee K, Kim J, Berggren PO, Ryu SH, Jo J. Insulin modulates the frequency of Ca2+ oscillations in mouse pancreatic islets. PLoS One 2017; 12:e0183569. [PMID: 28846705 PMCID: PMC5573301 DOI: 10.1371/journal.pone.0183569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/07/2017] [Indexed: 11/20/2022] Open
Abstract
Pancreatic islets can adapt to oscillatory glucose to produce synchronous insulin pulses. Can islets adapt to other oscillatory stimuli, specifically insulin? To answer this question, we stimulated islets with pulses of exogenous insulin and measured their Ca2+ oscillations. We observed that sufficiently high insulin (> 500 nM) with an optimal pulse period (~ 4 min) could make islets to produce synchronous Ca2+ oscillations. Glucose and insulin, which are key stimulatory factors of islets, modulate islet Ca2+ oscillations differently. Glucose increases the active-to-silent ratio of phases, whereas insulin increases the period of the oscillation. To examine the dual modulation, we adopted a phase oscillator model that incorporated the phase and frequency modulations. This mathematical model showed that out-of-phase oscillations of glucose and insulin were more effective at synchronizing islet Ca2+ oscillations than in-phase stimuli. This finding suggests that a phase shift in glucose and insulin oscillations can enhance inter-islet synchronization.
Collapse
Affiliation(s)
- Boah Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Taegeun Song
- Asia Pacific Center for Theoretical Physics, Pohang, Gyeongbuk, Korea
| | - Kayoung Lee
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Jaeyoon Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Per-Olof Berggren
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institute, Stockholm, Sweden
| | - Sung Ho Ryu
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Junghyo Jo
- Asia Pacific Center for Theoretical Physics, Pohang, Gyeongbuk, Korea
- Department of Physics, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
- * E-mail:
| |
Collapse
|
41
|
Zhang L, Zhou L, Song X, Liang G, Xu Z, Wang F, Huang F, Jiang G. Involvement of exogenous 3‑deoxyglucosone in β‑cell dysfunction induces impaired glucose regulation. Mol Med Rep 2017; 16:2976-2984. [PMID: 28656301 DOI: 10.3892/mmr.2017.6856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 05/04/2017] [Indexed: 11/05/2022] Open
Abstract
β‑cell dysfunction is the primary cause of type 2 diabetes mellitus (T2DM). 1,2‑dicarbonyl compounds, such as 3‑deoxyglucosone (3DG) have been reported to increase the risk of T2DM. Abnormal elevation of plasma 3DG may impair β‑cell function and thereby, it is linked to T2DM. Previous findings suggest that exogenous 3DG may serve an important role in the development of pre‑diabetes. In the present study, the authors examine whether exogenous 3DG induces impaired glucose regulation in mice by decreasing β‑cell function involving of accumulation of plasma 3DG. At two weeks following administration of 3DG, fasting blood glucose (FBG) levels, oral glucose tolerance (by a glucose meter) and plasma levels of 3DG (by HPLC) and insulin (by radioimmunoassay) were measured. Glucose‑stimulated insulin secretion in cultured pancreas islets and INS‑1 cells was measured by radioimmunoassay. Western blotting was used to examine the expression of the key molecules of the insulin‑PI3K signaling pathway. 3DG treatment increased FBG and fasting blood insulin levels, reduced oral glucose tolerance in conjunction with decreased ∆Ins30‑0/∆G30‑0. In 3DG‑treated mice, an increase in the plasma 3DG level was observed, which was most likely the mechanism for decreased β‑cell function. This idea was further supported by these results that non‑cytotoxic 3DG concentration obviously decreased glucose‑stimulated insulin secretion in cultured pancreas islets and INS‑1 cells exposure to high glucose (25.5 mM). 3DG decreased the expression of GLUT2 and phosphorylation of IRS‑1, PI3K‑p85 and Akt in high glucose‑induced INS‑1 cells. To the best of the authors' knowledge, the present study is the first to demonstrate that exogenous 3DG induced normal mice to develop IGR, resulting from β‑cell dysfunction. Exogenous 3DG administration increased plasma 3DG levels, which participates in inducing β‑cell dysfunction, at least in part, through impairing IRS‑1/PI3K/GLUT2 signaling.
Collapse
Affiliation(s)
- Lurong Zhang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| | - Liang Zhou
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| | - Xiudao Song
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| | - Guoqiang Liang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| | - Zhongrui Xu
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| | - Fei Wang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| | - Fei Huang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| | - Guorong Jiang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215003, P.R. China
| |
Collapse
|
42
|
Edgerton DS, Kraft G, Smith M, Farmer B, Williams PE, Coate KC, Printz RL, O'Brien RM, Cherrington AD. Insulin's direct hepatic effect explains the inhibition of glucose production caused by insulin secretion. JCI Insight 2017; 2:e91863. [PMID: 28352665 DOI: 10.1172/jci.insight.91863] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Insulin can inhibit hepatic glucose production (HGP) by acting directly on the liver as well as indirectly through effects on adipose tissue, pancreas, and brain. While insulin's indirect effects are indisputable, their physiologic role in the suppression of HGP seen in response to increased insulin secretion is not clear. Likewise, the mechanisms by which insulin suppresses lipolysis and pancreatic α cell secretion under physiologic circumstances are also debated. In this study, insulin was infused into the hepatic portal vein to mimic increased insulin secretion, and insulin's indirect liver effects were blocked either individually or collectively. During physiologic hyperinsulinemia, plasma free fatty acid (FFA) and glucagon levels were clamped at basal values and brain insulin action was blocked, but insulin's direct effects on the liver were left intact. Insulin was equally effective at suppressing HGP when its indirect effects were absent as when they were present. In addition, the inhibition of lipolysis, as well as glucagon and insulin secretion, did not require CNS insulin action or decreased plasma FFA. This indicates that the rapid suppression of HGP is attributable to insulin's direct effect on the liver and that its indirect effects are redundant in the context of a physiologic increase in insulin secretion.
Collapse
Affiliation(s)
- Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Guillaume Kraft
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Marta Smith
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ben Farmer
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Phillip E Williams
- Division of Surgical Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Katie C Coate
- Samford University, Department of Nutrition and Dietetics, Birmingham, Alabama, USA
| | - Richard L Printz
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Richard M O'Brien
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
43
|
Ikonomov OC, Sbrissa D, Delvecchio K, Rillema JA, Shisheva A. Unexpected severe consequences of Pikfyve deletion by aP2- or Aq-promoter-driven Cre expression for glucose homeostasis and mammary gland development. Physiol Rep 2016; 4:4/11/e12812. [PMID: 27273882 PMCID: PMC4908490 DOI: 10.14814/phy2.12812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/04/2016] [Indexed: 01/03/2023] Open
Abstract
Systemic deficiency of PIKfyve, the evolutionarily conserved phosphoinositide kinase synthesizing cellular PtdIns5P and PtdIns(3,5)P2 and implicated in insulin signaling, causes early embryonic death in mice. In contrast, mice with muscle‐specific Pikfyve disruption have normal lifespan but exhibit early‐age whole‐body glucose intolerance and muscle insulin resistance, thus establishing the key role of muscle PIKfyve in glucose homeostasis. Fat and muscle tissues control postprandial glucose clearance through different mechanisms, raising questions as to whether adipose Pikfyve disruption will also trigger whole‐body metabolic abnormalities, and if so, what the mechanism might be. To clarify these issues, here we have characterized two new mouse models with adipose tissue disruption of Pikfyve through Cre recombinase expression driven by adipose‐specific aP2‐ or adiponectin (Aq) promoters. Whereas both mouse lines were ostensibly normal until adulthood, their glucose homeostasis and systemic insulin sensitivity were severely dysregulated. These abnormalities stemmed in part from accelerated fat‐cell lipolysis and elevated serum FFA. Intriguingly, aP2‐Cre‐PIKfyvefl/fl but not Aq‐Cre‐PIKfyvefl/fl females had severely impaired pregnancy‐induced mammary gland differentiation and lactogenesis, consistent with aP2‐Cre‐mediated Pikfyve excision in nonadipogenic tissues underlying this defect. Intriguingly, whereas mammary glands from postpartum control and Aq‐Cre‐PIKfyvefl/fl mice or ex vivo mammary gland explants showed profound upregulation of PIKfyve protein levels subsequent to prolactin receptor activation, such increases were not apparent in aP2‐Cre‐PIKfyvefl/fl females. Collectively, our data identify for the first time that adipose tissue Pikfyve plays a key role in the mechanisms regulating glucose homeostasis and that the PIKfyve pathway is critical in mammary epithelial differentiation during pregnancy and lactogenesis downstream of prolactin receptor signaling.
Collapse
Affiliation(s)
- Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Diego Sbrissa
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Khortnal Delvecchio
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - James A Rillema
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
44
|
Benthuysen JR, Carrano AC, Sander M. Advances in β cell replacement and regeneration strategies for treating diabetes. J Clin Invest 2016; 126:3651-3660. [PMID: 27694741 DOI: 10.1172/jci87439] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In the past decade, new approaches have been explored that are aimed at restoring functional β cell mass as a treatment strategy for diabetes. The two most intensely pursued strategies are β cell replacement through conversion of other cell types and β cell regeneration by enhancement of β cell replication. The approach closest to clinical implementation is the replacement of β cells with human pluripotent stem cell-derived (hPSC-derived) cells, which are currently under investigation in a clinical trial to assess their safety in humans. In addition, there has been success in reprogramming developmentally related cell types into β cells. Reprogramming approaches could find therapeutic applications by inducing β cell conversion in vivo or by reprogramming cells ex vivo followed by implantation. Finally, recent studies have revealed novel pharmacologic targets for stimulating β cell replication. Manipulating these targets or the pathways they regulate could be a strategy for promoting the expansion of residual β cells in diabetic patients. Here, we provide an overview of progress made toward β cell replacement and regeneration and discuss promises and challenges for clinical implementation of these strategies.
Collapse
|
45
|
Insulin receptor alternative splicing is regulated by insulin signaling and modulates beta cell survival. Sci Rep 2016; 6:31222. [PMID: 27526875 PMCID: PMC4985653 DOI: 10.1038/srep31222] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/14/2016] [Indexed: 12/03/2022] Open
Abstract
Type 2 Diabetes (T2DM) affects more than 300 million people worldwide. One of the hallmarks of T2DM is peripheral insulin resistance, in part due to unproductive insulin signaling through the insulin receptor. The insulin receptor (INSR) exists as two isoforms, INSR-A and INSR-B, which results from skipping or inclusion of exon 11 respectively. What determines the relative abundance of the different insulin receptor splice variants is unknown. Moreover, it is not yet clear what the physiological roles of each of the isoforms are in normal and diseased beta cells. In this study, we show that insulin induces INSR exon 11 inclusion in pancreatic beta cells in both human and mouse. This occurs through activation of the Ras-MAPK/ERK signaling pathway and up-regulation of the splicing factor SRSF1. Induction of exon 11 skipping by a splice-site competitive antisense oligonucleotide inhibited the MAPK-ERK signaling pathway downstream of the insulin receptor, sensitizing the pancreatic β-cell line MIN6 to stress-induced apoptosis and lipotoxicity. These results assign to insulin a regulatory role in INSR alternative splicing through the Ras-MAPK/ERK signaling pathway. We suggest that in beta cells, INSR-B has a protective role, while INSR-A expression sensitizes beta cells to programmed cell death.
Collapse
|
46
|
Stamateris RE, Sharma RB, Kong Y, Ebrahimpour P, Panday D, Ranganath P, Zou B, Levitt H, Parambil NA, O'Donnell CP, García-Ocaña A, Alonso LC. Glucose Induces Mouse β-Cell Proliferation via IRS2, MTOR, and Cyclin D2 but Not the Insulin Receptor. Diabetes 2016; 65:981-95. [PMID: 26740601 PMCID: PMC5314707 DOI: 10.2337/db15-0529] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022]
Abstract
An important goal in diabetes research is to understand the processes that trigger endogenous β-cell proliferation. Hyperglycemia induces β-cell replication, but the mechanism remains debated. A prime candidate is insulin, which acts locally through the insulin receptor. Having previously developed an in vivo mouse hyperglycemia model, we tested whether glucose induces β-cell proliferation through insulin signaling. By using mice lacking insulin signaling intermediate insulin receptor substrate 2 (IRS2), we confirmed that hyperglycemia-induced β-cell proliferation requires IRS2 both in vivo and ex vivo. Of note, insulin receptor activation was not required for glucose-induced proliferation, and insulin itself was not sufficient to drive replication. Glucose and insulin caused similar acute signaling in mouse islets, but chronic signaling differed markedly, with mammalian target of rapamycin (MTOR) and extracellular signal-related kinase (ERK) activation by glucose and AKT activation by insulin. MTOR but not ERK activation was required for glucose-induced proliferation. Cyclin D2 was necessary for glucose-induced β-cell proliferation. Cyclin D2 expression was reduced when either IRS2 or MTOR signaling was lost, and restoring cyclin D2 expression rescued the proliferation defect. Human islets shared many of these regulatory pathways. Taken together, these results support a model in which IRS2, MTOR, and cyclin D2, but not the insulin receptor, mediate glucose-induced proliferation.
Collapse
Affiliation(s)
- Rachel E Stamateris
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Rohit B Sharma
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Yahui Kong
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Pantea Ebrahimpour
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Deepika Panday
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Pavana Ranganath
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Baobo Zou
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Helena Levitt
- Division of Endocrinology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Christopher P O'Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Disease, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Laura C Alonso
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
47
|
Kuznetsova A, Yu Y, Hollister-Lock J, Opare-Addo L, Rozzo A, Sadagurski M, Norquay L, Reed JE, El Khattabi I, Bonner-Weir S, Weir GC, Sharma A, White MF. Trimeprazine increases IRS2 in human islets and promotes pancreatic β cell growth and function in mice. JCI Insight 2016; 1. [PMID: 27152363 PMCID: PMC4854304 DOI: 10.1172/jci.insight.80749] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The capacity of pancreatic β cells to maintain glucose homeostasis during chronic physiologic and immunologic stress is important for cellular and metabolic homeostasis. Insulin receptor substrate 2 (IRS2) is a regulated adapter protein that links the insulin and IGF1 receptors to downstream signaling cascades. Since strategies to maintain or increase IRS2 expression can promote β cell growth, function, and survival, we conducted a screen to find small molecules that can increase IRS2 mRNA in isolated human pancreatic islets. We identified 77 compounds, including 15 that contained a tricyclic core. To establish the efficacy of our approach, one of the tricyclic compounds, trimeprazine tartrate, was investigated in isolated human islets and in mouse models. Trimeprazine is a first-generation antihistamine that acts as a partial agonist against the histamine H1 receptor (H1R) and other GPCRs, some of which are expressed on human islets. Trimeprazine promoted CREB phosphorylation and increased the concentration of IRS2 in islets. IRS2 was required for trimeprazine to increase nuclear Pdx1, islet mass, β cell replication and function, and glucose tolerance in mice. Moreover, trimeprazine synergized with anti-CD3 Abs to reduce the progression of diabetes in NOD mice. Finally, it increased the function of human islet transplants in streptozotocin-induced (STZ-induced) diabetic mice. Thus, trimeprazine, its analogs, or possibly other compounds that increase IRS2 in islets and β cells without adverse systemic effects might provide mechanism-based strategies to prevent the progression of diabetes.
Collapse
Affiliation(s)
- Alexandra Kuznetsova
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yue Yu
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Hollister-Lock
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Lynn Opare-Addo
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aldo Rozzo
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianna Sadagurski
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lisa Norquay
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica E Reed
- Housey Pharmaceutical Research Laboratories, Southfield, Michigan, USA
| | - Ilham El Khattabi
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Bonner-Weir
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Gordon C Weir
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Arun Sharma
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Morris F White
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Abstract
In this issue of Developmental Cell, Okamoto and Nishimura (2015) identify a positive feedback loop between neuronal cells that maintains insulin signaling and growth under restricted nutritional conditions.
Collapse
Affiliation(s)
- Rebecca A S Palu
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Carl S Thummel
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
49
|
Proinsulin and heat shock protein 90 as biomarkers of beta-cell stress in the early period after onset of type 1 diabetes. Transl Res 2016; 168:96-106.e1. [PMID: 26397425 PMCID: PMC4839287 DOI: 10.1016/j.trsl.2015.08.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/12/2015] [Accepted: 08/28/2015] [Indexed: 11/23/2022]
Abstract
Rapid evaluation of therapies designed to preserve β cells in persons with type 1 diabetes (T1D) is hampered by limited availability of sensitive β-cell health biomarkers. In particular, biomarkers elucidating the presence and degree of β-cell stress are needed. We characterized β-cell secretory activity and stress in 29 new-onset T1D subjects (10.6 ± 3.0 years, 55% male) at diagnosis and then 8.2 ± 1.2 weeks later at first clinic follow-up. We did comparisons with 16 matched healthy controls. We evaluated hemoglobin A1c (HbA1c), β-cell function (random C-peptide [C] and proinsulin [PI]), β-cell stress (PI:C ratio), and the β-cell stress marker heat shock protein (HSP)90 and examined these parameters' relationships with clinical and laboratory characteristics at diagnosis. Mean diagnosis HbA1c was 11.3% (100 mmol/mol) and 7.6% (60 mmol/mol) at follow-up. C-peptide was low at diagnosis (P < 0.001 vs controls) and increased at follow-up (P < 0.001) to comparable with controls. PI did not differ from controls at diagnosis but increased at follow-up (P = 0.003) signifying increased release of PI alongside improved insulin secretion. PI:C ratios and HSP90 concentrations were elevated at both time points. Younger subjects had lower C-peptide and greater PI, PI:C, and HSP90. We also examined islets isolated from prediabetic nonobese diabetic mice and found that HSP90 levels were increased ∼4-fold compared with those in islets isolated from matched CD1 controls, further substantiating HSP90 as a marker of β-cell stress in T1D. Our data indicate that β-cell stress can be assessed using PI:C and HSP90. This stress persists after T1D diagnosis. Therapeutic approaches to reduce β-cell stress in new-onset T1D should be considered.
Collapse
|
50
|
White MF, Copps KD. The Mechanisms of Insulin Action. ENDOCRINOLOGY: ADULT AND PEDIATRIC 2016:556-585.e13. [DOI: 10.1016/b978-0-323-18907-1.00033-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|