1
|
Ribeiro FM, Arnaldo L, P Milhomem L, S Aguiar S, Franco OL. The intricate relationship between circadian rhythms and gastrointestinal peptides in obesity. Peptides 2025; 185:171356. [PMID: 39929256 DOI: 10.1016/j.peptides.2025.171356] [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: 11/10/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
There are different molecular pathways that regulate appetite, particularly the role of the hypothalamus, circadian rhythms, and gastrointestinal peptides. The hypothalamus integrates signals from orexigenic peptides like neuropeptide Y (NPY) and agouti-related protein (AgRP), which stimulate appetite, and anorexigenic peptides such as pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART), which promote satiety. These signals are influenced by peripheral hormones like leptin, ghrelin, insulin, and cortisol, as well as gut peptides including glucagon-like peptide-1 (GLP-1), peptide YY (PYY), and cholecystokinin (CCK). The circadian rhythm, regulated by proteins like circadian locomotor output cycles kaput (CLOCK) and brain and muscle ARNT-like 1 (BMAL1), modulates the secretion of these peptides, aligning feeding behaviors with the sleep-wake cycle. In obesity, these regulatory systems are disrupted, leading to leptin resistance, increased ghrelin sensitivity, and altered gut peptide secretion. This results in heightened appetite and impaired satiety, contributing to overeating and metabolic dysfunction. Additionally, circadian disruptions further impair metabolic processes, exacerbating obesity. The present article underscores the importance of understanding the molecular interplay between circadian rhythms and gastrointestinal peptides, particularly in the context of obesity. While some molecular interactions, such as the regulation of GLP-1 and PYY by reverberation of circadian rhythm α (REV-ERBα) and retinoic acid-related orphan receptor α (RORα), are well-established, clinical studies are scarce. Future research is expected to explore these pathways in obesity management, especially with the rise of incretin-based treatments like semaglutide. A deeper understanding of hypothalamic molecular mechanisms could lead to novel pharmacological and non-pharmacological therapies for obesity.
Collapse
Affiliation(s)
- Filipe M Ribeiro
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, DF, Brazil
| | - Luiz Arnaldo
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, DF, Brazil; Postgraduate Program in Molecular Pathology, University of Brasília, Brasília, DF, Brazil
| | - Lana P Milhomem
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, DF, Brazil
| | - Samuel S Aguiar
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, DF, Brazil
| | - Octavio L Franco
- Center for Proteomic and Biochemical Analysis, Post-Graduation in Genomic and Biotechnology Sciences, Catholic University of Brasilia, Brasília, DF, Brazil; Postgraduate Program in Molecular Pathology, University of Brasília, Brasília, DF, Brazil; S-Inova Biotech, Catholic University Dom Bosco, Biotechnology Program, Campo Grande, MS, Brazil.
| |
Collapse
|
2
|
Toyoshima Y, Nakamura K, Taguchi Y, Tokita R, Takeuchi S, Osawa H, Teramoto N, Sugihara H, Yoshizawa F, Yamanouchi K, Minami S. Deletion of IRS-1 leads to growth failure and insulin resistance with downregulation of liver and muscle insulin signaling in rats. Sci Rep 2025; 15:649. [PMID: 39779784 PMCID: PMC11711447 DOI: 10.1038/s41598-024-84234-1] [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: 01/29/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Insulin receptor substrate (IRS)-1 and IRS-2 are major molecules that transduce signals from insulin and insulin-like growth factor-I receptors. The physiological functions of these proteins have been intensively investigated in mice, while little is known in other animals. Our previous study showed that the disruption of IRS-2 impairs body growth but not glucose tolerance or insulin sensitivity in rats, which led us to hypothesize that IRS-1 plays more pivotal roles in insulin functions than IRS-2. Here, we created IRS-1 knockout (KO) rats to elucidate the physiological roles of IRS-1 in rats. The body weight of IRS-1 KO rats at birth was lower than that of wild-type (WT) littermates, and postnatal growth of IRS-1 KO rats was severely impaired. Compared with WT rats, IRS-1 KO rats displayed insulin resistance but maintained euglycemia because of compensatory hyperinsulinemia. In addition, despite the increased activity of insulin-stimulated IRS-2-associated phosphatidylinositol-3 kinase (PI3K), insulin-induced phosphorylation of the kinases downstream of PI3K was suppressed in the liver and skeletal muscle of IRS-1 KO rats. Taken together, these results indicate that in rats, IRS-1 is essential for normal growth and the glucose-lowering effects of insulin. IRS-1 appears to be more important than IRS-2 for insulin functions in rats.
Collapse
Affiliation(s)
- Yuka Toyoshima
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University, 350 Mine-machi, Utsunomiya, 321-8505, Tochigi, Japan.
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan.
| | - Katsuyuki Nakamura
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Chemistry and Biomolecular Science, Biomolecular Science Course, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Yusuke Taguchi
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Reiko Tokita
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Shiho Takeuchi
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hayato Osawa
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University, 350 Mine-machi, Utsunomiya, 321-8505, Tochigi, Japan
| | - Naomi Teramoto
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidetoshi Sugihara
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Fumiaki Yoshizawa
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University, 350 Mine-machi, Utsunomiya, 321-8505, Tochigi, Japan
| | - Keitaro Yamanouchi
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shiro Minami
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan
| |
Collapse
|
3
|
Ueki K. Rethinking Diabetes from the Perspective of Diverse Insulin Actions in Various Organs. JMA J 2024; 7:489-495. [PMID: 39513056 PMCID: PMC11543345 DOI: 10.31662/jmaj.2024-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/16/2024] [Indexed: 11/15/2024] Open
Abstract
Diabetes mellitus is defined as a group of metabolic diseases characterized by chronic hyperglycemia based on insufficient insulin action. At present, treatment for diabetes aims to prevent micro- and macrovascular complications. Although advances have been made in methods of controlling the risk factors of complications, including blood glucose management, there is still no effective treatment to cure diabetes. This is largely because we do not fully understand what diabetes is. To cure diabetes, it is necessary to elucidate the whole picture of insulin actions including those other than metabolic actions in various tissues and to understand what disorders are caused by its reduction or excess. This article reviews diverse insulin actions in various organs and the effects of their deficiency on diabetes, its complications, and associated diseases.
Collapse
Affiliation(s)
- Kohjiro Ueki
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Molecular Diabetology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Chen Q, Jiang FJ, Gao X, Li XY, Xia P. Steatotic hepatocyte-derived extracellular vesicles promote β-cell apoptosis and diabetes via microRNA-126a-3p. Liver Int 2023; 43:2560-2570. [PMID: 37337778 DOI: 10.1111/liv.15654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/21/2023]
Abstract
Extracellular vesicles (EVs) have emerged as a unique mediator of interorgan communications, playing important roles in the pathophysiologic process of various diseases, including diabetes and other metabolic diseases. Here, we reported that the EVs released by steatotic hepatocytes exerted a detrimental effect on pancreatic β cells, leading to β-cell apoptosis and dysfunction. The effect was profoundly attributable to an up-regulation of miR-126a-3p in the steatotic hepatocyte-derived EVs. Accordingly, overexpression of miR-126a-3p promoted, whereas inhibition of miR-126a-3p prevented β-cell apoptosis, through a mechanism related to its target gene, insulin receptor substrate-2. Moreover, inhibition of miR-126a-3p by its specific antagomir was able to partially reverse the loss of β-cell mass and ameliorate hyperglycaemia in diabetic mice. Thus, the findings reveal a novel pathogenic role of steatotic hepatocyte-derived EVs, which mechanistically links nonalcoholic fatty liver disease to the development of diabetes.
Collapse
Affiliation(s)
- Qi Chen
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fang-Jie Jiang
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Ying Li
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pu Xia
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Nishiyama K, Ono M, Tsuno T, Inoue R, Fukunaka A, Okuyama T, Kyohara M, Togashi Y, Fukushima S, Atsumi T, Sato A, Tsurumoto A, Sakai C, Fujitani Y, Terauchi Y, Ito S, Shirakawa J. Protective Effects of Imeglimin and Metformin Combination Therapy on β-Cells in db/db Male Mice. Endocrinology 2023; 164:bqad095. [PMID: 37314160 DOI: 10.1210/endocr/bqad095] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 06/15/2023]
Abstract
Imeglimin and metformin act in metabolic organs, including β-cells, via different mechanisms. In the present study, we investigated the impacts of imeglimin, metformin, or their combination (Imeg + Met) on β-cells, the liver, and adipose tissues in db/db mice. Imeglimin, metformin, or Imeg + Met treatment had no significant effects on glucose tolerance, insulin sensitivity, respiratory exchange ratio, or locomotor activity in db/db mice. The responsiveness of insulin secretion to glucose was recovered by Imeg + Met treatment. Furthermore, Imeg + Met treatment increased β-cell mass by enhancing β-cell proliferation and ameliorating β-cell apoptosis in db/db mice. Hepatic steatosis, the morphology of adipocytes, adiposity assessed by computed tomography, and the expression of genes related to glucose or lipid metabolism and inflammation in the liver and fat tissues showed no notable differences in db/db mice. Global gene expression analysis of isolated islets indicated that the genes related to regulation of cell population proliferation and negative regulation of cell death were enriched by Imeg + Met treatment in db/db islets. In vitro culture experiments confirmed the protective effects of Imeg + Met against β-cell apoptosis. The expression of Snai1, Tnfrsf18, Pdcd1, Mmp9, Ccr7, Egr3, and Cxcl12, some of which have been linked to apoptosis, in db/db islets was attenuated by Imeg + Met. Treatment of a β-cell line with Imeg + Met prevented apoptosis induced by hydrogen peroxide or palmitate. Thus, the combination of imeglimin and metformin is beneficial for the maintenance of β-cell mass in db/db mice, probably through direct action on β-cells, suggesting a potential strategy for protecting β-cells in the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Kuniyuki Nishiyama
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Masato Ono
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Takahiro Tsuno
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Ayako Fukunaka
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Setsuko Fukushima
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Takuto Atsumi
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Aoi Sato
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Asuka Tsurumoto
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Chisato Sakai
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Shuichi Ito
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| |
Collapse
|
6
|
Spezani R, Marinho TS, Macedo Cardoso LE, Aguila MB, Mandarim-de-Lacerda CA. Pancreatic islet remodeling in cotadutide-treated obese mice. Life Sci 2023; 327:121858. [PMID: 37315839 DOI: 10.1016/j.lfs.2023.121858] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/16/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) cause morphofunctional alterations in pancreatic islet alpha and beta cells. Therefore, we hypothesize that the new GLP-1/Glucagon receptor dual agonist cotadutide may benefit islet cell arrangement and function. Twelve-week-old C57BL/6 male mice were fed a control diet (C, 10 % kJ fat) or a high-fat diet (HF, 50 % kJ fat) for ten weeks. Then, the animals were divided into four groups for an additional 30 days and daily treated with subcutaneous cotadutide (30 nmol/kg) or vehicle: C, CC (control+cotadutide), HF, and HFC (high-fat+cotadutide). Cotadutide led to weight loss and reduced insulin resistance in the HFC group, increasing insulin receptor substrate 1 and solute carrier family 2 gene expressions in isolated islets. Also, cotadutide enhanced transcriptional factors related to islet cell transdifferentiation, decreasing aristaless-related homeobox and increasing the paired box 4 and 6, pancreatic and duodenal homeobox 1, v-maf musculoaponeurotic fibrosarcoma oncogene family protein A, neurogenin 3, and neurogenic differentiation 1. In addition, cotadutide improved the proliferating cell nuclear antigen, NK6 homeobox 1, B cell leukemia/lymphoma 2, but lessening caspase 3. Furthermore, cotadutide mitigated the endoplasmic reticulum (ER) stress-responsive genes, reducing transcription factor 4, DNA-damage-inducible transcript 3, and growth arrest and DNA-damage-inducible 45. In conclusion, our data demonstrated significant beneficial actions of cotadutide in DIO mice, such as weight loss, glycemic control, and insulin resistance improvement. In addition, cotadutide counteracted the pathological adaptive cellular arrangement of the pancreatic islet in obese mice, improving the markers of the transdifferentiating pathway, proliferation, apoptosis, and ER stress.
Collapse
Affiliation(s)
- Renata Spezani
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz E Macedo Cardoso
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
7
|
Girdhar K, Soto M, Huang Q, Orliaguet L, Cederquist C, Sundaresh B, Hu J, Figura M, Raisingani A, Canfora EE, Dirice E, Fujisaka S, Goossens GH, Blaak EE, Kulkarni RN, Kahn CR, Altindis E. Gut Microbiota Regulate Pancreatic Growth, Exocrine Function, and Gut Hormones. Diabetes 2022; 71:945-960. [PMID: 35212729 PMCID: PMC9044125 DOI: 10.2337/db21-0382] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022]
Abstract
Growing evidence indicates an important link between gut microbiota, obesity, and metabolic syndrome. Alterations in exocrine pancreatic function are also widely present in patients with diabetes and obesity. To examine this interaction, C57BL/6J mice were fed a chow diet, a high-fat diet (HFD), or an HFD plus oral vancomycin or metronidazole to modify the gut microbiome. HFD alone leads to a 40% increase in pancreas weight, decreased glucagon-like peptide 1 and peptide YY levels, and increased glucose-dependent insulinotropic peptide in the plasma. Quantitative proteomics identified 138 host proteins in fecal samples of these mice, of which 32 were significantly changed by the HFD. The most significant of these were the pancreatic enzymes. These changes in amylase and elastase were reversed by antibiotic treatment. These alterations could be reproduced by transferring gut microbiota from donor C57BL/6J mice to germ-free mice. By contrast, antibiotics had no effect on pancreatic size or exocrine function in C57BL/6J mice fed the chow diet. Further, 1 week vancomycin administration significantly increased amylase and elastase levels in obese men with prediabetes. Thus, the alterations in gut microbiota in obesity can alter pancreatic growth, exocrine function, and gut endocrine function and may contribute to the alterations observed in patients with obesity and diabetes.
Collapse
Affiliation(s)
| | - Marion Soto
- Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Qian Huang
- Biology Department Boston College, Chestnut Hill, MA
| | - Lucie Orliaguet
- Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Carly Cederquist
- Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | | | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | | | | | - Emanuel E. Canfora
- Department of Human Biology, Maastricht University, Maastricht, the Netherlands
| | - Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- Department of Pharmacology, School of Medicine, New York Medical College, Valhalla, NY
| | - Shiho Fujisaka
- Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Gijs H. Goossens
- Department of Human Biology, Maastricht University, Maastricht, the Netherlands
| | - Ellen E. Blaak
- Department of Human Biology, Maastricht University, Maastricht, the Netherlands
| | - Rohit N. Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women′s Hospital, Harvard Medical School, Boston, MA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA
| | - C. Ronald Kahn
- Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- Corresponding authors: Emrah Altindis, , and C. Ronald Kahn,
| | - Emrah Altindis
- Biology Department Boston College, Chestnut Hill, MA
- Corresponding authors: Emrah Altindis, , and C. Ronald Kahn,
| |
Collapse
|
8
|
Mechanistic Investigation of GHS-R Mediated Glucose-Stimulated Insulin Secretion in Pancreatic Islets. Biomolecules 2022; 12:biom12030407. [PMID: 35327599 PMCID: PMC8945998 DOI: 10.3390/biom12030407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/19/2022] [Accepted: 02/27/2022] [Indexed: 02/07/2023] Open
Abstract
Ghrelin receptor, a growth hormone secretagogue receptor (GHS-R), is expressed in the pancreas. Emerging evidence indicates that GHS-R is involved in the regulation of glucose-stimulated insulin secretion (GSIS), but the mechanism by which GHS-R regulates GSIS in the pancreas is unclear. In this study, we investigated the role of GHS-R on GSIS in detail using global Ghsr−/− mice (in vivo) and Ghsr-ablated pancreatic islets (ex vivo). GSIS was attenuated in both Ghsr−/− mice and Ghsr-ablated islets, while the islet morphology was similar between WT and Ghsr−/− mice. To elucidate the mechanism underpinning Ghsr-mediated GSIS, we investigated the key steps of the GSIS signaling cascade. The gene expression of glucose transporter 2 (Glut2) and the glucose-metabolic intermediate—glucose-6-phosphate (G6P) were reduced in Ghsr-ablated islets, supporting decreased glucose uptake. There was no difference in mitochondrial DNA content in the islets of WT and Ghsr−/− mice, but the ATP/ADP ratio in Ghsr−/− islets was significantly lower than that of WT islets. Moreover, the expression of pancreatic and duodenal homeobox 1 (Pdx1), as well as insulin signaling genes of insulin receptor (IR) and insulin receptor substrates 1 and 2 (IRS1/IRS2), was downregulated in Ghsr−/− islets. Akt is the key mediator of the insulin signaling cascade. Concurrently, Akt phosphorylation was reduced in the pancreas of Ghsr−/− mice under both insulin-stimulated and homeostatic conditions. These findings demonstrate that GHS-R ablation affects key components of the insulin signaling pathway in the pancreas, suggesting the existence of a cross-talk between GHS-R and the insulin signaling pathway in pancreatic islets, and GHS-R likely regulates GSIS via the Akt-Pdx1-GLUT2 pathway.
Collapse
|
9
|
Halperin F, Mezza T, Li P, Shirakawa J, Kulkarni RN, Goldfine AB. Insulin regulates arginine-stimulated insulin secretion in humans. Metabolism 2022; 128:155117. [PMID: 34999111 PMCID: PMC8821403 DOI: 10.1016/j.metabol.2021.155117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/16/2021] [Accepted: 12/26/2021] [Indexed: 11/30/2022]
Abstract
AIMS Insulin potentiates glucose-stimulated insulin secretion. These effects are attenuated in beta cell-specific insulin receptor knockout mice and insulin resistant humans. This investigation examines whether short duration insulin exposure regulates beta cell responsiveness to arginine, a non-glucose secretagogue, in healthy humans. MATERIALS AND METHODS Arginine-stimulated insulin secretion was studied in 10 healthy humans. In each subject arginine was administered as a bolus followed by continuous infusion on two occasions one month apart, after sham/saline or hyperinsulinemic-isoglycemic clamp, respectively providing low and high insulin pre-exposure conditions. Arginine-stimulated insulin secretion was measured by C-peptide deconvolution, and by a selective immunogenic (DAKO) assay for direct measurement of endogenous but not exogenous insulin. RESULTS Pre-exposure to exogenous insulin augmented arginine-stimulated insulin secretion. The effect was seen acutely following arginine bolus (endogenous DAKO insulin incremental AUC240-255min 311.6 ± 208.1 (post-insulin exposure) versus 120.6 ± 42.2 μU/ml•min (sham/saline) (t-test P = 0.021)), as well as in response to continuous arginine infusion (DAKO insulin incremental AUC260-290min 1095.3 ± 592.1 (sham/saline) versus 564.8 ± 207.1 μU/ml•min (high insulin)(P = 0.009)). Findings were similar when beta cell response was assessed using C-peptide, insulin secretion rates by deconvolution, and the C-peptide to glucose ratio. CONCLUSIONS We demonstrate a physiologic role of insulin in regulation of the beta cell secretory response to arginine.
Collapse
Affiliation(s)
- Florencia Halperin
- Joslin Diabetes Center, Boston, MA, United States of America; Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Teresa Mezza
- Joslin Diabetes Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America; Endocrinologia e Diabetologia, Fondazione Policlinico Universitario A. Gemelli IRCSS, Roma, Italy; Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ping Li
- Joslin Diabetes Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America; Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, PR China
| | - Jun Shirakawa
- Joslin Diabetes Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America; Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Rohit N Kulkarni
- Joslin Diabetes Center, Boston, MA, United States of America; Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America.
| | - Allison B Goldfine
- Joslin Diabetes Center, Boston, MA, United States of America; Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
10
|
Atmodjo WL, Larasati YO, Jo J, Nufika R, Naomi S, Winoto I. Relationship Between Insulin-Receptor Substrate 1 and Langerhans' Islet in a Rat Model of Type 2 Diabetes Mellitus. In Vivo 2021; 35:291-297. [PMID: 33402476 DOI: 10.21873/invivo.12258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND/AIM In vivo studies on pathogenesis of type 2 diabetes mellitus (T2DM) have been reported, however, the relationship between insulin-receptor substrate 1 (IRS1) and the area of Langerhans' islets was unknown. Therefore, a correlation between both parameters was assessed. MATERIALS AND METHODS Diabetic groups were fed with a high-fat diet (HFD) and injected with three different doses of streptozotocin, namely 25, 35 and 45 mg/kg, and compared to a control group after 9 weeks. RESULTS Administration of HFD/streptozotocin increased the level of fasting blood glucose but reduced the level of IRS1 and the area of Langerhans' islets in diabetic groups. The coefficient of correlation between IRS1 and area of Langerhans' islets was 0.259 (p=0.232). In addition, the coefficient of correlation for fasting blood glucose with the area of Langerhans' islets and IRS1 was -0.520 (p=0.011) and -0.603 (p=0.002), respectively. CONCLUSION The reduction of IRS1 was weakly correlated with the destruction of Langerhans' islets, suggesting there is an intermediate step between both parameters.
Collapse
Affiliation(s)
- Wahyuni Lukita Atmodjo
- Department of Immunopathology, Mochtar Riady Institute for Nanotechnology, Tangerang, Indonesia; .,Department of Anatomy, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
| | - Young Othiwi Larasati
- Department of Immunopathology, Mochtar Riady Institute for Nanotechnology, Tangerang, Indonesia
| | - Juandy Jo
- Program of Biology, Faculty of Science and Technology, Universitas Pelita Harapan, Tangerang, Indonesia
| | - Riska Nufika
- Department of Immunopathology, Mochtar Riady Institute for Nanotechnology, Tangerang, Indonesia
| | - Steffi Naomi
- Department of Biomedicine, Indonesia International Institute for Life Science, Jakarta, Indonesia
| | - Imelda Winoto
- Department of Immunopathology, Mochtar Riady Institute for Nanotechnology, Tangerang, Indonesia
| |
Collapse
|
11
|
Takatani T, Shirakawa J, Shibue K, Gupta MK, Kim H, Lu S, Hu J, White MF, Kennedy RT, Kulkarni RN. Insulin receptor substrate 1, but not IRS2, plays a dominant role in regulating pancreatic alpha cell function in mice. J Biol Chem 2021; 296:100646. [PMID: 33839150 PMCID: PMC8131928 DOI: 10.1016/j.jbc.2021.100646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/24/2021] [Accepted: 04/07/2021] [Indexed: 11/29/2022] Open
Abstract
Dysregulated glucagon secretion deteriorates glycemic control in type 1 and type 2 diabetes. Although insulin is known to regulate glucagon secretion via its cognate receptor (insulin receptor, INSR) in pancreatic alpha cells, the role of downstream proteins and signaling pathways underlying insulin's activities are not fully defined. Using in vivo (knockout) and in vitro (knockdown) studies targeting insulin receptor substrate (IRS) proteins, we compared the relative roles of IRS1 and IRS2 in regulating alpha cell function. Alpha cell-specific IRS1-knockout mice exhibited glucose intolerance and inappropriate glucagon suppression during glucose tolerance tests. In contrast, alpha cell-specific IRS2-knockout animals manifested normal glucose tolerance and suppression of glucagon secretion after glucose administration. Alpha cell lines with stable IRS1 knockdown could not repress glucagon mRNA expression and exhibited a reduction in phosphorylation of AKT Ser/Thr kinase (AKT, at Ser-473 and Thr-308). AlphaIRS1KD cells also displayed suppressed global protein translation, including reduced glucagon expression, impaired cytoplasmic Ca2+ response, and mitochondrial dysfunction. This was supported by the identification of novel IRS1-specific downstream target genes, Trpc3 and Cartpt, that are associated with glucagon regulation in alpha cells. These results provide evidence that IRS1, rather than IRS2, is a dominant regulator of pancreatic alpha cell function.
Collapse
Affiliation(s)
- Tomozumi Takatani
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Gunma, Japan
| | - Kimitaka Shibue
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manoj K Gupta
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Cell Therapy Translational Engine (CTTE), Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Hyunki Kim
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shusheng Lu
- Departments of Chemistry and Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Morris F White
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert T Kennedy
- Departments of Chemistry and Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
12
|
Rachdaoui N. Insulin: The Friend and the Foe in the Development of Type 2 Diabetes Mellitus. Int J Mol Sci 2020; 21:ijms21051770. [PMID: 32150819 PMCID: PMC7084909 DOI: 10.3390/ijms21051770] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/14/2022] Open
Abstract
Insulin, a hormone produced by pancreatic β-cells, has a primary function of maintaining glucose homeostasis. Deficiencies in β-cell insulin secretion result in the development of type 1 and type 2 diabetes, metabolic disorders characterized by high levels of blood glucose. Type 2 diabetes mellitus (T2DM) is characterized by the presence of peripheral insulin resistance in tissues such as skeletal muscle, adipose tissue and liver and develops when β-cells fail to compensate for the peripheral insulin resistance. Insulin resistance triggers a rise in insulin demand and leads to β-cell compensation by increasing both β-cell mass and insulin secretion and leads to the development of hyperinsulinemia. In a vicious cycle, hyperinsulinemia exacerbates the metabolic dysregulations that lead to β-cell failure and the development of T2DM. Insulin and IGF-1 signaling pathways play critical roles in maintaining the differentiated phenotype of β-cells. The autocrine actions of secreted insulin on β-cells is still controversial; work by us and others has shown positive and negative actions by insulin on β-cells. We discuss findings that support the concept of an autocrine action of secreted insulin on β-cells. The hypothesis of whether, during the development of T2DM, secreted insulin initially acts as a friend and contributes to β-cell compensation and then, at a later stage, becomes a foe and contributes to β-cell decompensation will be discussed.
Collapse
Affiliation(s)
- Nadia Rachdaoui
- Department of Animal Sciences, Room 108, Foran Hall, Rutgers, the State University of New Jersey, 59 Dudley Rd, New Brunswick, NJ 08901, USA
| |
Collapse
|
13
|
Li Y, Wang K, Zhang P, Huang J, Liu Y, Wang Z, Lu Y, Tan S, Yang F, Tan Y. Pyrosequencing analysis of IRS1 methylation levels in schizophrenia with tardive dyskinesia. Mol Med Rep 2020; 21:1702-1708. [PMID: 32319643 PMCID: PMC7057828 DOI: 10.3892/mmr.2020.10984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/17/2020] [Indexed: 12/16/2022] Open
Abstract
Tardive dyskinesia (TD) is a serious side effect of certain antipsychotic medications that are used to treat schizophrenia (SCZ) and other mental illnesses. The methylation status of the insulin receptor substrate 1 (IRS1) gene is reportedly associated with SCZ; however, no study, to the best of the authors' knowledge, has focused on the quantitative DNA methylation levels of the IRS1 gene using pyrosequencing in SCZ with or without TD. The present study aimed to quantify DNA methylation levels of 4 CpG sites in the IRS1 gene using a Chinese sample including SCZ patients with TD and without TD (NTD) and healthy controls (HCs). The general linear model (GLM) was used to detect DNA methylation levels among the 3 proposed groups (TD vs. NTD vs. HC). Mean DNA methylation levels of 4 CpG sites demonstrated normal distribution. Pearson's correlation analysis did not reveal any significant correlations between the DNA methylation levels of the 4 CpG sites and the severity of SCZ. GLM revealed significant differences between the 3 groups for CpG site 1 and the average of the 4 CpG sites (P=0.0001 and P=0.0126, respectively). Furthermore, the TD, NTD and TD + NTD groups demonstrated lower methylation levels in CpG site 1 (P=0.0003, P<0.0001 and P<0.0001, respectively) and the average of 4 CpG sites (P=0.0176, P=0.0063 and P=0.003, respectively) compared with the HC group. The results revealed that both NTD and TD patients had significantly decreased DNA methylation levels compared with healthy controls, which indicated a significant association between the DNA methylation levels of the IRS1 gene with SCZ and TD.
Collapse
Affiliation(s)
- Yanli Li
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Kesheng Wang
- Department of Family and Community Health, School of Nursing, Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| | - Ping Zhang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Junchao Huang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Ying Liu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN 37614, USA
| | - Zhiren Wang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Yongke Lu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Shuping Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Fude Yang
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| | - Yunlong Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, P.R. China
| |
Collapse
|
14
|
Sachs S, Bastidas-Ponce A, Tritschler S, Bakhti M, Böttcher A, Sánchez-Garrido MA, Tarquis-Medina M, Kleinert M, Fischer K, Jall S, Harger A, Bader E, Roscioni S, Ussar S, Feuchtinger A, Yesildag B, Neelakandhan A, Jensen CB, Cornu M, Yang B, Finan B, DiMarchi RD, Tschöp MH, Theis FJ, Hofmann SM, Müller TD, Lickert H. Targeted pharmacological therapy restores β-cell function for diabetes remission. Nat Metab 2020; 2:192-209. [PMID: 32694693 DOI: 10.1038/s42255-020-0171-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/15/2020] [Indexed: 12/27/2022]
Abstract
Dedifferentiation of insulin-secreting β cells in the islets of Langerhans has been proposed to be a major mechanism of β-cell dysfunction. Whether dedifferentiated β cells can be targeted by pharmacological intervention for diabetes remission, and ways in which this could be accomplished, are unknown as yet. Here we report the use of streptozotocin-induced diabetes to study β-cell dedifferentiation in mice. Single-cell RNA sequencing (scRNA-seq) of islets identified markers and pathways associated with β-cell dedifferentiation and dysfunction. Single and combinatorial pharmacology further show that insulin treatment triggers insulin receptor pathway activation in β cells and restores maturation and function for diabetes remission. Additional β-cell selective delivery of oestrogen by Glucagon-like peptide-1 (GLP-1-oestrogen conjugate) decreases daily insulin requirements by 60%, triggers oestrogen-specific activation of the endoplasmic-reticulum-associated protein degradation system, and further increases β-cell survival and regeneration. GLP-1-oestrogen also protects human β cells against cytokine-induced dysfunction. This study not only describes mechanisms of β-cell dedifferentiation and regeneration, but also reveals pharmacological entry points to target dedifferentiated β cells for diabetes remission.
Collapse
Affiliation(s)
- Stephan Sachs
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Miguel A Sánchez-Garrido
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Maximilian Kleinert
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Katrin Fischer
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| | - Sigrid Jall
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| | - Alexandra Harger
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Erik Bader
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Sara Roscioni
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Siegfried Ussar
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, Neuherberg, Germany
| | | | | | | | - Marion Cornu
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Bin Yang
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Matthias H Tschöp
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany.
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Department of Mathematics, Technical University of Munich, Munich, Germany.
| | - Susanna M Hofmann
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medical Clinic and Polyclinic IV, Ludwig Maximilian University of Munich, Munich, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany.
- Department of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
15
|
Dirice E, Kahraman S, De Jesus DF, El Ouaamari A, Basile G, Baker RL, Yigit B, Piehowski PD, Kim MJ, Dwyer AJ, Ng RWS, Schuster C, Vethe H, Martinov T, Ishikawa Y, Teo AKK, Smith RD, Hu J, Haskins K, Serwold T, Qian WJ, Fife BT, Kissler S, Kulkarni RN. Increased β-cell proliferation before immune cell invasion prevents progression of type 1 diabetes. Nat Metab 2019; 1:509-518. [PMID: 31423480 PMCID: PMC6696912 DOI: 10.1038/s42255-019-0061-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is characterized by pancreatic islet infiltration by autoreactive immune cells and a near-total loss of β-cells1. Restoration of insulin-producing β-cells coupled with immunomodulation to suppress the autoimmune attack has emerged as a potential approach to counter T1D2-4. Here we report that enhancing β-cell mass early in life, in two models of female NOD mice, results in immunomodulation of T-cells, reduced islet infiltration and lower β-cell apoptosis, that together protect them from developing T1D. The animals displayed altered β-cell antigens, and islet transplantation studies showed prolonged graft survival in the NOD-LIRKO model. Adoptive transfer of splenocytes from the NOD-LIRKOs prevented development of diabetes in pre-diabetic NOD mice. A significant increase in the splenic CD4+CD25+FoxP3+ regulatory T-cell (Treg) population was observed to underlie the protected phenotype since Treg depletion rendered NOD-LIRKO mice diabetic. The increase in Tregs coupled with activation of TGF-β/SMAD3 signaling pathway in pathogenic T-cells favored reduced ability to kill β-cells. These data support a previously unidentified observation that initiating β-cell proliferation, alone, prior to islet infiltration by immune cells alters the identity of β-cells, decreases pathologic self-reactivity of effector cells and increases Tregs to prevent progression of T1D.
Collapse
Affiliation(s)
- Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Sevim Kahraman
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Dario F. De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
- Graduate Program in Areas of Basic and Applied Biology
(GABBA), Abel Salazar Biomedical Sciences Institute, University of Porto, Porto,
Portugal
| | - Abdelfattah El Ouaamari
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Giorgio Basile
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Rocky L. Baker
- Department of Immunology, School of Medicine, University of
Colorado, Aurora, CO, USA
| | - Burcu Yigit
- Division of Immunology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, MA, USA
| | - Paul D. Piehowski
- Biological Sciences Division, Pacific Northwest National
Laboratory, Richland, WA, USA
| | - Mi-Jeong Kim
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Alexander J. Dwyer
- University of Minnesota, Center for Immunology, Department
of Medicine, Minneapolis, MN, USA
| | - Raymond W. S. Ng
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Heidrun Vethe
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
| | - Tijana Martinov
- University of Minnesota, Center for Immunology, Department
of Medicine, Minneapolis, MN, USA
| | - Yuki Ishikawa
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Adrian Kee Keong Teo
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National
Laboratory, Richland, WA, USA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn Haskins
- Department of Immunology, School of Medicine, University of
Colorado, Aurora, CO, USA
| | - Thomas Serwold
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National
Laboratory, Richland, WA, USA
| | - Brian T. Fife
- University of Minnesota, Center for Immunology, Department
of Medicine, Minneapolis, MN, USA
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston,
MA, USA
| | - Rohit N. Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes
Center, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Boston, MA, USA
| |
Collapse
|
16
|
Dirice E, De Jesus DF, Kahraman S, Basile G, Ng RW, El Ouaamari A, Teo AKK, Bhatt S, Hu J, Kulkarni RN. Human duct cells contribute to β cell compensation in insulin resistance. JCI Insight 2019; 4:99576. [PMID: 30996131 PMCID: PMC6538348 DOI: 10.1172/jci.insight.99576] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/05/2019] [Indexed: 12/19/2022] Open
Abstract
The identification of new sources of β cells is an important endeavor with therapeutic implications for diabetes. Insulin resistance, in physiological states such as pregnancy or in pathological states such as type 2 diabetes (T2D), is characterized by a compensatory increase in β cell mass. To explore the existence of a dynamic β cell reserve, we superimposed pregnancy on the liver-specific insulin receptor-KO (LIRKO) model of insulin resistance that already exhibits β cell hyperplasia and used lineage tracing to track the source of new β cells. Although both control and LIRKO mice displayed increased β cell mass in response to the relative insulin resistance of pregnancy, the further increase in mass in the latter supported a dynamic source that could be traced to pancreatic ducts. Two observations support the translational significance of these findings. First, NOD/SCID-γ LIRKO mice that became pregnant following cotransplantation of human islets and human ducts under the kidney capsule showed enhanced β cell proliferation and an increase in ductal cells positive for transcription factors expressed during β cell development. Second, we identified duct cells positive for immature β cell markers in pancreas sections from pregnant humans and in individuals with T2D. Taken together, during increased insulin demand, ductal cells contribute to the compensatory β cell pool by differentiation/neogenesis.
Collapse
Affiliation(s)
- Ercument Dirice
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Dario F. De Jesus
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
- Graduate Program in Areas of Basic and Applied Biology, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Sevim Kahraman
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Giorgio Basile
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Raymond W.S. Ng
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Abdelfattah El Ouaamari
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Adrian Kee Keong Teo
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Shweta Bhatt
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Jiang Hu
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Rohit N. Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Ye R, Onodera T, Scherer PE. Lipotoxicity and β Cell Maintenance in Obesity and Type 2 Diabetes. J Endocr Soc 2019; 3:617-631. [PMID: 30834357 PMCID: PMC6391718 DOI: 10.1210/js.2018-00372] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
Obesity and diabetes are often associated with lipotoxic conditions in multiple tissues. The insulin-producing β cells are susceptible to elevated lipid levels and the ensuing lipotoxicity. The preservation of β cell mass and function is one of the main goals of diabetes management under these metabolically stressful conditions. However, the adverse effects from the adaptive signaling pathways that β cells use to counteract lipotoxic stress have secondary negative effects in their own right. Antilipotoxic signaling cascades in β cells can contribute to their eventual failure. Such dual roles are seen for many other biological adaptive processes as well.
Collapse
Affiliation(s)
- Risheng Ye
- Department of Medical Education, Texas Tech University Health Sciences Center Paul L. Foster School of Medicine, El Paso, Texas
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Toshiharu Onodera
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
18
|
Rachdaoui N, Polo-Parada L, Ismail-Beigi F. Prolonged Exposure to Insulin Inactivates Akt and Erk 1/2 and Increases Pancreatic Islet and INS1E β-Cell Apoptosis. J Endocr Soc 2018; 3:69-90. [PMID: 30697602 PMCID: PMC6344346 DOI: 10.1210/js.2018-00140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic hyperinsulinemia, in vivo, increases the resistance of peripheral tissues to insulin by desensitizing insulin signaling. Insulin, in a heterologous manner, can also cause IGF-1 resistance. The aim of the current study was to investigate whether insulin-mediated insulin and IGF-1 resistance develops in pancreatic β-cells and whether this resistance results in β-cell decompensation. Chronic exposure of rat islets or INS1E β-cells to increasing concentrations of insulin decreased AktS473 phosphorylation in response to subsequent acute stimulation with 10 nM insulin or IGF-1. Prolonged exposure to high insulin levels not only inhibited AktS473 phosphorylation, but it also resulted in a significant inhibition of the phosphorylation of P70S6 kinase and Erk1/2 phosphorylation in response to the acute stimulation by glucose, insulin, or IGF-1. Decreased activation of Akt, P70S6K, and Erk1/2 was associated with decreased insulin receptor substrate 2 tyrosine phosphorylation and insulin receptor β-subunit abundance; neither IGF receptor β-subunit content nor its phosphorylation were affected. These signaling impairments were associated with decreased SERCA2 expression, perturbed plasma membrane calcium current and intracellular calcium handling, increased endoplasmic reticulum stress markers such as eIF2αS51 phosphorylation and Bip (GRP78) expression, and increased islet and β-cell apoptosis. We demonstrate that prolonged exposure to high insulin levels induces not only insulin resistance, but in a heterologous manner causes resistance to IGF-1 in rat islets and insulinoma cells resulting in decreased cell survival. These findings suggest the possibility that chronic exposure to hyperinsulinemia may negatively affect β-cell mass by increasing β-cell apoptosis.
Collapse
Affiliation(s)
- Nadia Rachdaoui
- Division of Clinical and Molecular Endocrinology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Luis Polo-Parada
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Faramarz Ismail-Beigi
- Division of Clinical and Molecular Endocrinology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
19
|
Kim C, Jeong DE, Heo S, Ji E, Rho JG, Jung M, Ahn S, Kim YJ, Kim YS, Nam SW, Kulkarni RN, Lee KB, Lee EK, Kim W. Reduced expression of the RNA-binding protein HuD in pancreatic neuroendocrine tumors correlates with low p27 Kip1 levels and poor prognosis. J Pathol 2018; 246:231-243. [PMID: 30014466 DOI: 10.1002/path.5135] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/14/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022]
Abstract
For the majority of patients diagnosed with pancreatic neuroendocrine tumors (NETs), there is significant malignant potential with a poor prognosis; however, the molecular abnormalities and pathogenesis of pancreatic NETs have not been firmly established. Here, we report that loss of expression of the RNA-binding protein HuD correlates with low p27Kip1 (p27) levels and poor prognosis in pancreatic NETs. HuD expression was frequently lost in many human pancreatic NETs, and these pancreatic NETs showed aggressive clinicopathological phenotypes with low p27 levels, increased tumor size, higher World Health Organization grade and pT stage of the tumor, and the presence of angioinvasion. Furthermore, loss of HuD was an independent, progression-free prognostic factor in multivariate survival analysis. However, the level of HuR, a member of the same Hu protein family as HuD, was not significantly correlated with pancreatic NET size and progression. Mechanistically, HuD enhanced p27 mRNA translation by interacting with both the 5'-untranslated region (UTR) and the 3'-UTR of p27 mRNA, and consequently suppressed cell cycle progression and tumor growth. In addition, HuD competed with miR-30a-3p for binding to the 3'-UTR of p27 mRNA, suggesting an interplay between HuD and miR-30a-3p in controlling p27 translation. Our results identify HuD as a pivotal suppressor of pancreatic NET growth, and suggest that HuD has potential value as a prognostic factor of pancreatic NETs. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Chongtae Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Da Eun Jeong
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sungeun Heo
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Eunbyul Ji
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jun Gi Rho
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Myeongwoo Jung
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sojin Ahn
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ye-Jin Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Rohit N Kulkarni
- Department of Islet Cell and Regenerative Biology, Joslin Diabetes Center and Department of Medicine, Harvard Medical School and Harvard Stem Cell Institute, Boston, MA, USA
| | - Kyoung Bun Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
20
|
Abstract
Insulin-like growth factors (IGFs) bind specifically to the IGF1 receptor on the cell surface of targeted tissues. Ligand binding to the α subunit of the receptor leads to a conformational change in the β subunit, resulting in the activation of receptor tyrosine kinase activity. Activated receptor phosphorylates several substrates, including insulin receptor substrates (IRSs) and Src homology collagen (SHC). Phosphotyrosine residues in these substrates are recognized by certain Src homology 2 (SH2) domain-containing signaling molecules. These include, for example, an 85 kDa regulatory subunit (p85) of phosphatidylinositol 3-kinase (PI 3-kinase), growth factor receptor-bound 2 (GRB2) and SH2-containing protein tyrosine phosphatase 2 (SHP2/Syp). These bindings lead to the activation of downstream signaling pathways, PI 3-kinase pathway and Ras-mitogen-activated protein kinase (MAP kinase) pathway. Activation of these signaling pathways is known to be required for the induction of various bioactivities of IGFs, including cell proliferation, cell differentiation and cell survival. In this review, the well-established IGF1 receptor signaling pathways required for the induction of various bioactivities of IGFs are introduced. In addition, we will discuss how IGF signals are modulated by the other extracellular stimuli or by themselves based on our studies.
Collapse
Affiliation(s)
- Fumihiko Hakuno
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Takahashi
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
21
|
Magnesium upregulates insulin receptor and glucose transporter-4 in streptozotocin-nicotinamide-induced type-2 diabetic rats. Endocr Regul 2018; 52:6-16. [DOI: 10.2478/enr-2018-0002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Objective. We investigated the effects of magnesium supplementation on glucose tolerance, insulin sensitivity, oxidative stress as well as the concentration of insulin receptor and glucose transporter-4 in streptozotocin-nicotinamide induced type-2 diabetic (T2D) rats. Methods. Rats were divided into four groups designated as: 1) control (CTR); 2) diabetic untreated (DU); 3) diabetic treated with 1 mg of Mg/kg diet (Mg1-D); and 4) diabetic treated with 2 mg of Mg/kg diet (Mg2-D). T2D was induced with a single intraperitoneal (i.p.) injection of freshly prepared streptozotocin (55 mg/kg) aft er an initial i.p. injection of nicotinamide (120 mg/kg). Glucose tolerance, insulin sensitivity, lipid profile, malondialdehyde (MAD) and glutathione content, insulin receptors (INSR) and glucose transporter-4 (GLUT4), fasting insulin and glucose levels were measured, and insulin resistance index was calculated using the homeostatic model assessment of insulin resistance (HOMA-IR). Results. Magnesium supplementation improved glucose tolerance and lowered blood glucose levels almost to the normal range. We also recorded a noticeable increase in insulin sensitivity in Mg-D groups when compared with DU rats. Lipid perturbations associated T2D were significantly attenuated by magnesium supplementation. Fasting glucose level was comparable to control values in the Mg-D groups while the HOMA-IR index was significantly lower compared with the DU rats. Magnesium reduced MDA but increased glutathione concentrations compared with DU group. Moreover, INSR and GLUT4 levels were elevated following magnesium supplementation in T2D rats. Conclusion. These findings demonstrate that magnesium may mediate effective metabolic control by stimulating the antioxidant defense, and increased levels of INSR and GLUT4 in diabetic rats.
Collapse
|
22
|
Rodríguez-Comas J, Moreno-Asso A, Moreno-Vedia J, Martín M, Castaño C, Marzà-Florensa A, Bofill-De Ros X, Mir-Coll J, Montané J, Fillat C, Gasa R, Novials A, Servitja JM. Stress-Induced MicroRNA-708 Impairs β-Cell Function and Growth. Diabetes 2017; 66:3029-3040. [PMID: 28970284 DOI: 10.2337/db16-1569] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 09/24/2017] [Indexed: 11/13/2022]
Abstract
The pancreatic β-cell transcriptome is highly sensitive to external signals such as glucose oscillations and stress cues. MicroRNAs (miRNAs) have emerged as key factors in gene expression regulation. Here, we aimed to identify miRNAs that are modulated by glucose in mouse pancreatic islets. We identified miR-708 as the most upregulated miRNA in islets cultured at low glucose concentrations, a setting that triggers a strong stress response. miR-708 was also potently upregulated by triggering endoplasmic reticulum (ER) stress with thapsigargin and in islets of ob/ob mice. Low-glucose induction of miR-708 was blocked by treatment with the chemical chaperone 4-phenylbutyrate, uncovering the involvement of ER stress in this response. An integrative analysis identified neuronatin (Nnat) as a potential glucose-regulated target of miR-708. Indeed, Nnat expression was inversely correlated with miR-708 in islets cultured at different glucose concentrations and in ob/ob mouse islets and was reduced after miR-708 overexpression. Consistent with the role of Nnat in the secretory function of β-cells, miR-708 overexpression impaired glucose-stimulated insulin secretion (GSIS), which was recovered by NNAT overexpression. Moreover, miR-708 inhibition recovered GSIS in islets cultured at low glucose. Finally, miR-708 overexpression suppressed β-cell proliferation and induced β-cell apoptosis. Collectively, our results provide a novel mechanism of glucose regulation of β-cell function and growth by repressing stress-induced miR-708.
Collapse
Affiliation(s)
- Júlia Rodríguez-Comas
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alba Moreno-Asso
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Juan Moreno-Vedia
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mercè Martín
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carlos Castaño
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Anna Marzà-Florensa
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Xavier Bofill-De Ros
- Gene Therapy and Cancer Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Joan Mir-Coll
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Joel Montané
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Cristina Fillat
- Gene Therapy and Cancer Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Rosa Gasa
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Anna Novials
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| | - Joan-Marc Servitja
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Barcelona, Spain
| |
Collapse
|
23
|
Caporarello N, Parrino C, Trischitta V, Frittitta L. Insulin receptor signaling and glucagon-like peptide 1 effects on pancreatic beta cells. PLoS One 2017; 12:e0181190. [PMID: 28767692 PMCID: PMC5540605 DOI: 10.1371/journal.pone.0181190] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/26/2017] [Indexed: 12/03/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a potent gluco-incretin hormone, which plays a central role on pancreatic beta cell proliferation, survival and insulin secreting activity and whose analogs are used for treating hyperglycemia in type 2 diabetes mellitus. Notably, abnormal insulin signaling affects all the above-mentioned aspects on pancreatic beta cells. The aim of our study was to investigate whether the protective effects of GLP1-1 on beta cells are affected by altered insulin receptor signaling. To this end, several effects of GLP-1 were studied in INS-1E rat beta cells transfected either with an inhibitor of insulin receptor function (i.e., the Ectonucleotide Pyrophosphatase Phosphodiesterase 1, ENPP1), or with insulin receptor small interfering RNA, as well as in control cells. Crucial experiments were carried out also in a second cell line, namely the βTC-1 mouse beta cells. Our data indicate that in insulin secreting beta cells in which either ENPP1 was up-regulated or insulin receptor was down-regulated, GLP-1 positive effects on several pancreatic beta cell activities, including glucose-induced insulin secretion, cell proliferation and cell survival, were strongly reduced. Further studies are needed to understand whether such a scenario occurs also in humans and, if so, if it plays a role of clinical relevance in diabetic patients with poor responsiveness to GLP-1 related treatments.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Cristina Parrino
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Vincenzo Trischitta
- IRCCS Casa Sollievo della Sofferenza, Research Unit of Diabetes and Endocrine Diseases, San Giovanni Rotondo, Italy
- Department of Experimental Medicine “Sapienza” University, Rome, Italy
| | - Lucia Frittitta
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- * E-mail:
| |
Collapse
|
24
|
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
|
25
|
Shirakawa J, Fernandez M, Takatani T, El Ouaamari A, Jungtrakoon P, Okawa ER, Zhang W, Yi P, Doria A, Kulkarni RN. Insulin Signaling Regulates the FoxM1/PLK1/CENP-A Pathway to Promote Adaptive Pancreatic β Cell Proliferation. Cell Metab 2017; 25:868-882.e5. [PMID: 28286049 PMCID: PMC5382039 DOI: 10.1016/j.cmet.2017.02.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/09/2017] [Accepted: 02/08/2017] [Indexed: 12/01/2022]
Abstract
Investigation of cell-cycle kinetics in mammalian pancreatic β cells has mostly focused on transition from the quiescent (G0) to G1 phase. Here, we report that centromere protein A (CENP-A), which is required for chromosome segregation during the M-phase, is necessary for adaptive β cell proliferation. Receptor-mediated insulin signaling promotes DNA-binding activity of FoxM1 to regulate expression of CENP-A and polo-like kinase-1 (PLK1) by modulating cyclin-dependent kinase-1/2. CENP-A deposition at the centromere is augmented by PLK1 to promote mitosis, while knocking down CENP-A limits β cell proliferation and survival. CENP-A deficiency in β cells leads to impaired adaptive proliferation in response to pregnancy, acute and chronic insulin resistance, and aging in mice. Insulin-stimulated CENP-A/PLK1 protein expression is blunted in islets from patients with type 2 diabetes. These data implicate the insulin-FoxM1/PLK1/CENP-A pathway-regulated mitotic cell-cycle progression as an essential component in the β cell adaptation to delay and/or prevent progression to diabetes.
Collapse
Affiliation(s)
- Jun Shirakawa
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Megan Fernandez
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Tomozumi Takatani
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Abdelfattah El Ouaamari
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Prapaporn Jungtrakoon
- Section on Genetics and Epidemiology, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215, USA
| | - Erin R Okawa
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Wei Zhang
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Peng Yi
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215, USA
| | - Rohit N Kulkarni
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA.
| |
Collapse
|
26
|
Mezza T, Shirakawa J, Martinez R, Hu J, Giaccari A, Kulkarni RN. Nuclear Export of FoxO1 Is Associated with ERK Signaling in β-Cells Lacking Insulin Receptors. J Biol Chem 2016; 291:21485-21495. [PMID: 27535223 DOI: 10.1074/jbc.m116.735738] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/06/2016] [Indexed: 12/17/2022] Open
Abstract
The insulin/insulin-like growth factor (IGF) signaling pathway plays a critical role in the regulation of islet cell biology. However, the signaling pathway(s) utilized by insulin to directly modulate β-cells is unclear. To interrogate whether insulin exerts endocrine effects in regulating proteins in the insulin/IGF-1 signaling cascade in vivo in physiological states via the insulin receptor, we designed two experimental approaches: 1) glucose gavage and 2) hyperinsulinemic intravenous infusion, for studies in either β-cell specific insulin receptor knock-out (βIRKO) or control mice. Immunostaining of sections of pancreas (collected immediately after glucose gavage or insulin infusion) from controls showed significant increases in pAKT+, p-p70S6K+, and pERK+ β-cells and a significant decrease in % nuclear FoxO1+ β-cells compared with corresponding vehicle-treated groups. In contrast, in βIRKOs, we observed no significant changes in pAKT+ or p-p70S6K+ β-cells in either experiment; however, pERK+ β-cells were significantly increased, and an attenuated decrease in % nuclear FoxO1+ β cells was evident in response to glucose gavage or insulin infusion. Treatment of control and βIRKO β-cell lines with glucose or insulin showed significantly decreased % nuclear FoxO1+ β-cells suggesting direct effects. Furthermore, blocking MAPK signaling had virtually no effect on FoxO1 nuclear export in controls, in contrast to attenuated export in βIRKO β-cells. These data suggest insulin acts on β-cells in an endocrine manner in the normal situation; and that in β-cells lacking insulin receptors, insulin and glucose minimally activate the Akt pathway, while ERK phosphorylation and FoxO1 nuclear export occur independently of insulin signaling.
Collapse
Affiliation(s)
- Teresa Mezza
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02215 and.,Center for Endocrine and Metabolic Diseases, Policlinico Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Jun Shirakawa
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02215 and
| | - Rachael Martinez
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02215 and
| | - Jiang Hu
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02215 and
| | - Andrea Giaccari
- Center for Endocrine and Metabolic Diseases, Policlinico Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Rohit N Kulkarni
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02215 and
| |
Collapse
|
27
|
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
|
28
|
Takatani T, Shirakawa J, Roe MW, Leech CA, Maier BF, Mirmira RG, Kulkarni RN. IRS1 deficiency protects β-cells against ER stress-induced apoptosis by modulating sXBP-1 stability and protein translation. Sci Rep 2016; 6:28177. [PMID: 27378176 PMCID: PMC4932502 DOI: 10.1038/srep28177] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/31/2016] [Indexed: 01/05/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is among several pathological features that underlie β-cell failure in the development of type 1 and type 2 diabetes. Adaptor proteins in the insulin/insulin-like-growth factor-1 signaling pathways, such as insulin receptor substrate-1 (IRS1) and IRS2, differentially impact β-cell survival but the underlying mechanisms remain unclear. Here we report that β-cells deficient in IRS1 (IRS1KO) are resistant, while IRS2 deficiency (IRS2KO) makes them susceptible to ER stress-mediated apoptosis. IRS1KOs exhibited low nuclear accumulation of spliced XBP-1 due to its poor stability, in contrast to elevated accumulation in IRS2KO. The reduced nuclear accumulation in IRS1KO was due to protein instability of Xbp1 secondary to proteasomal degradation. IRS1KO also demonstrated an attenuation in their general translation status in response to ER stress revealed by polyribosomal profiling. Phosphorylation of eEF2 was dramatically increased in IRS1KO enabling the β-cells to adapt to ER stress by blocking translation. Furthermore, significantly high ER calcium (Ca2+) was detected in IRS1KO β-cells even upon induction of ER stress. These observations suggest that IRS1 could be a therapeutic target for β-cell protection against ER stress-mediated cell death by modulating XBP-1 stability, protein synthesis, and Ca2+ storage in the ER.
Collapse
Affiliation(s)
- Tomozumi Takatani
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Jun Shirakawa
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Michael W Roe
- Department of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, USA
| | - Colin A Leech
- Department of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, USA
| | - Bernhard F Maier
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cellular and Integrative Physiology, Department of Biochemistry and Molecular Biology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
Kolic J, Manning Fox JE, Chepurny OG, Spigelman AF, Ferdaoussi M, Schwede F, Holz GG, MacDonald PE. PI3 kinases p110α and PI3K-C2β negatively regulate cAMP via PDE3/8 to control insulin secretion in mouse and human islets. Mol Metab 2016; 5:459-471. [PMID: 27408772 PMCID: PMC4921792 DOI: 10.1016/j.molmet.2016.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 04/26/2016] [Accepted: 05/04/2016] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES Phosphatidylinositol-3-OH kinase (PI3K) signalling in the endocrine pancreas contributes to glycaemic control. However, the mechanism by which PI3K modulates insulin secretion from the pancreatic beta cell is poorly understood. Thus, our objective was two-fold; to determine the signalling pathway by which acute PI3K inhibition enhances glucose-stimulated insulin secretion (GSIS) and to examine the role of this pathway in islets from type-2 diabetic (T2D) donors. METHODS Isolated islets from mice and non-diabetic or T2D human donors, or INS 832/13 cells, were treated with inhibitors of PI3K and/or phosphodiesterases (PDEs). The expression of PI3K-C2β was knocked down using siRNA. We measured insulin release, single-cell exocytosis, intracellular Ca(2+) responses ([Ca(2+)]i) and Ca(2+) channel currents, intracellular cAMP concentrations ([cAMP]i), and activation of cAMP-dependent protein kinase A (PKA) and protein kinase B (PKB/AKT). RESULTS The non-specific PI3K inhibitor wortmannin amplifies GSIS, raises [cAMP]i and activates PKA, but is without effect in T2D islets. Direct inhibition of specific PDE isoforms demonstrates a role for PDE3 (in humans and mice) and PDE8 (in mice) downstream of PI3K, and restores glucose-responsiveness of T2D islets. We implicate a role for the Class II PI3K catalytic isoform PI3K-C2β in this effect by limiting beta cell exocytosis. CONCLUSIONS PI3K limits GSIS via PDE3 in human islets. While inhibition of p110α or PIK-C2β signalling per se, may promote nutrient-stimulated insulin release, we now suggest that this signalling pathway is perturbed in islets from T2D donors.
Collapse
Affiliation(s)
- Jelena Kolic
- Department of Pharmacology, and the Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| | - Jocelyn E Manning Fox
- Department of Pharmacology, and the Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Oleg G Chepurny
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Aliya F Spigelman
- Department of Pharmacology, and the Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Mourad Ferdaoussi
- Department of Pharmacology, and the Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Frank Schwede
- BIOLOG Life Science Institute, 28199 Bremen, Germany
| | - George G Holz
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA; Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Patrick E MacDonald
- Department of Pharmacology, and the Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| |
Collapse
|
30
|
Fibroblast Growth Factor 21 (FGF21) Protects against High Fat Diet Induced Inflammation and Islet Hyperplasia in Pancreas. PLoS One 2016; 11:e0148252. [PMID: 26872145 PMCID: PMC4752212 DOI: 10.1371/journal.pone.0148252] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 01/16/2016] [Indexed: 11/30/2022] Open
Abstract
Fibroblast growth factor 21 (FGF21) is an important endocrine metabolic regulator expressed in multiple tissues including liver and adipose tissue. Although highest levels of expression are in pancreas, little is known about the function of FGF21 in this tissue. In order to understand the physiology of FGF21 in the pancreas, we analyzed its expression and regulation in both acinar and islet tissues. We found that acinar tissue express 20-fold higher levels than that observed in islets. We also observed that pancreatic FGF21 is nutritionally regulated; a marked reduction in FGF21 expression was noted with fasting while obesity is associated with 3–4 fold higher expression. Acinar and islet cells are targets of FGF21, which when systemically administered, leads to phosphorylation of the downstream target ERK 1/2 in about half of acinar cells and a small subset of islet cells. Chronic, systemic FGF21 infusion down-regulates its own expression in the pancreas. Mice lacking FGF21 develop significant islet hyperplasia and periductal lymphocytic inflammation when fed with a high fat obesogenic diet. Inflammatory infiltrates consist of TCRb+ Thy1+ T lymphocytes with increased levels of Foxp3+ regulatory T cells. Increased levels of inflammatory cells were coupled with elevated expression of cytokines such as TNFα, IFNγ and IL1β. We conclude that FGF21 acts to limit islet hyperplasia and may also prevent pancreatic inflammation.
Collapse
|
31
|
Shirakawa J, Okuyama T, Kyohara M, Yoshida E, Togashi Y, Tajima K, Yamazaki S, Kaji M, Koganei M, Sasaki H, Terauchi Y. DPP-4 inhibition improves early mortality, β cell function, and adipose tissue inflammation in db/db mice fed a diet containing sucrose and linoleic acid. Diabetol Metab Syndr 2016; 8:16. [PMID: 26937254 PMCID: PMC4774120 DOI: 10.1186/s13098-016-0138-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/17/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Diabetes therapy that not only lowers glucose levels but also lengthens life spans is required. We previously demonstrated that DPP-4 inhibition ameliorated β cell apoptosis and adipose tissue inflammation in β cell-specific glucokinase haploinsufficient mice fed a diet containing a combination of sucrose and linoleic acid (SL). METHODS In this study, we investigated the effects of DPP-4 inhibition in obese diabetic db/db mice fed an SL diet or a control diet containing sucrose and oleic acid (SO). We also examined the effects of DPP-4 inhibition in IRS-1-deficient mice fed an SL or SO diet as a model of insulin resistance. RESULTS DPP-4 inhibition efficiently increases the active GLP-1 levels in db/db mice. Unexpectedly, the SL diet, but not the SO diet, markedly increases mortality in the db/db mice. DPP-4 inhibition reduces the early lethality in SL-fed db/db mice. DPP-4 inhibition improves glucose tolerance, β cell function, and adipose tissue inflammation in db/db mice fed either diet. No significant changes in glycemic control or β cell mass were observed in any of the IRS-1-deficient mouse groups. CONCLUSIONS A diet containing a combination of sucrose and linoleic acid causes early lethality in obese diabetic db/db mice, but not in lean and insulin resistant IRS-1 knockout mice. DPP-4 inhibition has protective effects against the diet-induced lethality in db/db mice.
Collapse
Affiliation(s)
- Jun Shirakawa
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Tomoko Okuyama
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Mayu Kyohara
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Eiko Yoshida
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Yu Togashi
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Kazuki Tajima
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Shunsuke Yamazaki
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Mitsuyo Kaji
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| | - Megumi Koganei
- />Food Science Research Laboratories, R&D Division, Meiji Co., Ltd., Odawara, Japan
| | - Hajime Sasaki
- />Food Science Research Laboratories, R&D Division, Meiji Co., Ltd., Odawara, Japan
- />Department of Nutritional and Life Sciences, Kanagawa Institute of Technology, Atsugi, Japan
| | - Yasuo Terauchi
- />Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004 Japan
| |
Collapse
|
32
|
Cataldo LR, Mizgier ML, Busso D, Olmos P, Galgani JE, Valenzuela R, Mezzano D, Aranda E, Cortés VA, Santos JL. Serotonin- and Dopamine-Related Gene Expression in db/db Mice Islets and in MIN6 β-Cells Treated with Palmitate and Oleate. J Diabetes Res 2016; 2016:3793781. [PMID: 27366756 PMCID: PMC4913013 DOI: 10.1155/2016/3793781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/26/2016] [Accepted: 05/10/2016] [Indexed: 12/20/2022] Open
Abstract
High circulating nonesterified fatty acids (NEFAs) concentration, often reported in diabetes, leads to impaired glucose-stimulated insulin secretion (GSIS) through not yet well-defined mechanisms. Serotonin and dopamine might contribute to NEFA-dependent β-cell dysfunction, since extracellular signal of these monoamines decreases GSIS. Moreover, palmitate-treated β-cells may enhance the expression of the serotonin receptor Htr2c, affecting insulin secretion. Additionally, the expression of monoamine-oxidase type B (Maob) seems to be lower in islets from humans and mice with diabetes compared to nondiabetic islets, which may lead to increased monoamine concentrations. We assessed the expression of serotonin- and dopamine-related genes in islets from db/db and wild-type (WT) mice. In addition, the effect of palmitate and oleate on the expression of such genes, 5HT content, and GSIS in MIN6 β-cell was determined. Lower Maob expression was found in islets from db/db versus WT mice and in MIN6 β-cells in response to palmitate and oleate treatment compared to vehicle. Reduced 5HT content and impaired GSIS in response to palmitate (-25%; p < 0.0001) and oleate (-43%; p < 0.0001) were detected in MIN6 β-cells. In conclusion, known defects of GSIS in islets from db/db mice and MIN6 β-cells treated with NEFAs are accompanied by reduced Maob expression and reduced 5HT content.
Collapse
Affiliation(s)
- L. R. Cataldo
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- Facultad de Medicina, Universidad de los Andes, 7620001 Santiago, Chile
| | - M. L. Mizgier
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - D. Busso
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - P. Olmos
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - J. E. Galgani
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- UDA-Ciencias de la Salud, Carrera de Nutrición y Dietética, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - R. Valenzuela
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile, 7550367 Santiago, Chile
| | - D. Mezzano
- Laboratorio de Hemostasia, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - E. Aranda
- Laboratorio de Hemostasia, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - V. A. Cortés
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - J. L. Santos
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- *J. L. Santos:
| |
Collapse
|
33
|
Lee YJ, Liu C, Liao M, Sukhova GK, Shirakawa J, Abdennour M, Iamarene K, Andre S, Inouye K, Clement K, Kulkarni RN, Banks AS, Libby P, Shi GP. Deficiency of FcϵR1 Increases Body Weight Gain but Improves Glucose Tolerance in Diet-Induced Obese Mice. Endocrinology 2015; 156:4047-58. [PMID: 26295369 PMCID: PMC4606759 DOI: 10.1210/en.2015-1184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prior studies demonstrated increased plasma IgE in diabetic patients, but the direct participation of IgE in diabetes or obesity remains unknown. This study found that plasma IgE levels correlated inversely with body weight, body mass index, and body fat mass among a population of randomly selected obese women. IgE receptor FcϵR1-deficient (Fcer1a(-/-)) mice and diet-induced obesity (DIO) mice demonstrated that FcϵR1 deficiency in DIO mice increased food intake, reduced energy expenditure, and increased body weight gain but improved glucose tolerance and glucose-induced insulin secretion. White adipose tissue from Fcer1a(-/-) mice showed an increased expression of phospho-AKT, CCAAT/enhancer binding protein-α, peroxisome proliferator-activated receptor-γ, glucose transporter-4 (Glut4), and B-cell lymphoma 2 (Bcl2) but reduced uncoupling protein 1 (UCP1) and phosphorylated c-Jun N-terminal kinase (JNK) expression, tissue macrophage accumulation, and apoptosis, suggesting that IgE reduces adipogenesis and glucose uptake but induces energy expenditure, adipocyte apoptosis, and white adipose tissue inflammation. In 3T3-L1 cells, IgE inhibited the expression of CCAAT/enhancer binding protein-α and peroxisome proliferator-activated receptor-γ, and preadipocyte adipogenesis and induced adipocyte apoptosis. IgE reduced the 3T3-L1 cell expression of Glut4, phospho-AKT, and glucose uptake, which concurred with improved glucose tolerance in Fcer1a(-/-) mice. This study established two novel pathways of IgE in reducing body weight gain in DIO mice by suppressing adipogenesis and inducing adipocyte apoptosis while worsening glucose tolerance by reducing Glut4 expression, glucose uptake, and insulin secretion.
Collapse
Affiliation(s)
- Yun-Jung Lee
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Conglin Liu
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Mengyang Liao
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Galina K Sukhova
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Jun Shirakawa
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Meriem Abdennour
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karine Iamarene
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Sebastien Andre
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karen Inouye
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karine Clement
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Rohit N Kulkarni
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Alexander S Banks
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Peter Libby
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Guo-Ping Shi
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| |
Collapse
|
34
|
Alvarez-Perez JC, Rosa TC, Casinelli GP, Valle SR, Lakshmipathi J, Rosselot C, Rausell-Palamos F, Vasavada RC, García-Ocaña A. Hepatocyte growth factor ameliorates hyperglycemia and corrects β-cell mass in IRS2-deficient mice. Mol Endocrinol 2015; 28:2038-48. [PMID: 25361392 DOI: 10.1210/me.2014-1207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Insulin resistance, when combined with decreased β-cell mass and relative insufficient insulin secretion, leads to type 2 diabetes. Mice lacking the IRS2 gene (IRS2(-/-) mice) develop diabetes due to uncompensated insulin resistance and β-cell failure. Hepatocyte growth factor (HGF) activates the phosphatidylinositol 3-kinase/Akt signaling pathway in β-cells without recruitment of IRS1 or IRS2 and increases β-cell proliferation, survival, mass, and function when overexpressed in β-cells of transgenic (TG) mice. We therefore hypothesized that HGF may protect against β-cell failure in IRS2 deficiency. For that purpose, we cross-bred TG mice overexpressing HGF in β-cells with IRS2 knockout (KO) mice. Glucose homeostasis analysis revealed significantly reduced hyperglycemia, compensatory hyperinsulinemia, and improved glucose tolerance in TG/KO mice compared with those in KO mice in the context of similar insulin resistance. HGF overexpression also increased glucose-stimulated insulin secretion in IRS2(-/-) islets. To determine whether this glucose homeostasis improvement correlated with alterations in β-cells, we measured β-cell mass, proliferation, and death in these mice. β-Cell proliferation was increased and death was decreased in TG/KO mice compared with those in KO mice. As a result, β-cell mass was significantly increased in TG/KO mice compared with that in KO mice, reaching levels similar to those in wild-type mice. Analysis of the intracellular targets involved in β-cell failure in IRS2 deficiency showed Pdx-1 up-regulation, Akt/FoxO1 phosphorylation, and p27 down-regulation in TG/KO mouse islets. Taken together, these results indicate that HGF can compensate for IRS2 deficiency and subsequent insulin resistance by normalizing β-cell mass and increasing circulating insulin. HGF may be of value as a therapeutic agent against β-cell failure.
Collapse
Affiliation(s)
- Juan C Alvarez-Perez
- Diabetes, Obesity and Metabolism Institute (J.C.A.-P., J.L., C.R., F.R.-P., R.C.V., A.G.-O.), Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Department of Medicine (T.C.R., G.P.C., S.R.V.), Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
El Ouaamari A, Zhou JY, Liew CW, Shirakawa J, Dirice E, Gedeon N, Kahraman S, De Jesus DF, Bhatt S, Kim JS, Clauss TR, Camp DG, Smith RD, Qian WJ, Kulkarni RN. Compensatory Islet Response to Insulin Resistance Revealed by Quantitative Proteomics. J Proteome Res 2015; 14:3111-3122. [PMID: 26151086 DOI: 10.1021/acs.jproteome.5b00587] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Compensatory islet response is a distinct feature of the prediabetic insulin-resistant state in humans and rodents. To identify alterations in the islet proteome that characterize the adaptive response, we analyzed islets from 5 month old male control, high-fat diet fed (HFD), or obese ob/ob mice by LC-MS/MS and quantified ~1100 islet proteins (at least two peptides) with a false discovery rate < 1%. Significant alterations in abundance were observed for ~350 proteins among groups. The majority of alterations were common to both models, and the changes of a subset of ~40 proteins and 12 proteins were verified by targeted quantification using selected reaction monitoring and western blots, respectively. The insulin-resistant islets in both groups exhibited reduced expression of proteins controlling energy metabolism, oxidative phosphorylation, hormone processing, and secretory pathways. Conversely, an increased expression of molecules involved in protein synthesis and folding suggested effects in endoplasmic reticulum stress response, cell survival, and proliferation in both insulin-resistant models. In summary, we report a unique comparison of the islet proteome that is focused on the compensatory response in two insulin-resistant rodent models that are not overtly diabetic. These data provide a valuable resource of candidate proteins to the scientific community to undertake further studies aimed at enhancing β-cell mass in patients with diabetes. The data are available via the MassIVE repository, under accession no. MSV000079093.
Collapse
Affiliation(s)
- Abdelfattah El Ouaamari
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Jian-Ying Zhou
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Chong Wee Liew
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jun Shirakawa
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Ercument Dirice
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Nicholas Gedeon
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Sevim Kahraman
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Dario F De Jesus
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Shweta Bhatt
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Jong-Seo Kim
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Therese Rw Clauss
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - David G Camp
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Richard D Smith
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Wei-Jun Qian
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Rohit N Kulkarni
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
36
|
Escribano O, Gómez-Hernández A, Díaz-Castroverde S, Nevado C, García G, Otero YF, Perdomo L, Beneit N, Benito M. Insulin receptor isoform A confers a higher proliferative capability to pancreatic beta cells enabling glucose availability and IGF-I signaling. Mol Cell Endocrinol 2015; 409:82-91. [PMID: 25797178 DOI: 10.1016/j.mce.2015.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 02/16/2015] [Accepted: 03/14/2015] [Indexed: 11/29/2022]
Abstract
The main compensatory response to insulin resistance is the pancreatic beta cell hyperplasia to account for increased insulin secretion. In fact, in a previous work we proposed a liver-pancreas endocrine axis with IGF-I (insulin-like growth factor type I) secreted by the liver acting on IRA insulin receptor in beta cells from iLIRKO mice (inducible Liver Insulin Receptor KnockOut) that showed a high IRA/IRB ratio. However, the role of insulin receptor isoforms in the IGF-I-induced beta cell proliferation as well as the underlying molecular mechanisms remain poorly understood. For this purpose, we have used four immortalized mouse beta cell lines: bearing IR (IRLoxP), lacking IR (IRKO), expressing exclusively IRA (IRA), or alternatively expressing IRB (IRB). Pancreatic beta cell proliferation studies showed that IRA cells are more sensitive than those expressing IRB to the mitogenic response induced by IGF-I, acting through the pathway IRA/IRS-1/2/αp85/Akt/mTORC1/p70S6K. More importantly, IRA beta cells, but not IRB, showed an increased glucose uptake as compared with IRLoxP cells, this effect being likely owing to an enhanced association between Glut-1 and Glut-2 with IRA. Overall, our results strongly suggest a prevalent role of IRA in glucose availability and IGF-I-induced beta cell proliferation mainly through mTORC1. These results could explain, at least partially, the role played by the liver-secreted IGF-I in the compensatory beta cell hyperplasia observed in response to severe hepatic insulin resistance in iLIRKO mice.
Collapse
Affiliation(s)
- Oscar Escribano
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| | - Almudena Gómez-Hernández
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Sabela Díaz-Castroverde
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Carmen Nevado
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Gema García
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Yolanda F Otero
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Liliana Perdomo
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Nuria Beneit
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| |
Collapse
|
37
|
Santulli G, Pagano G, Sardu C, Xie W, Reiken S, D’Ascia SL, Cannone M, Marziliano N, Trimarco B, Guise TA, Lacampagne A, Marks AR. Calcium release channel RyR2 regulates insulin release and glucose homeostasis. J Clin Invest 2015; 125:1968-1978. [PMID: 25844899 PMCID: PMC4463204 DOI: 10.1172/jci79273] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/27/2015] [Indexed: 12/21/2022] Open
Abstract
The type 2 ryanodine receptor (RyR2) is a Ca2+ release channel on the endoplasmic reticulum (ER) of several types of cells, including cardiomyocytes and pancreatic β cells. In cardiomyocytes, RyR2-dependent Ca2+ release is critical for excitation-contraction coupling; however, a functional role for RyR2 in β cell insulin secretion and diabetes mellitus remains controversial. Here, we took advantage of rare RyR2 mutations that were identified in patients with a genetic form of exercise-induced sudden death (catecholaminergic polymorphic ventricular tachycardia [CPVT]). As these mutations result in a "leaky" RyR2 channel, we exploited them to assess RyR2 channel function in β cell dynamics. We discovered that CPVT patients with mutant leaky RyR2 present with glucose intolerance, which was heretofore unappreciated. In mice, transgenic expression of CPVT-associated RyR2 resulted in impaired glucose homeostasis, and an in-depth evaluation of pancreatic islets and β cells from these animals revealed intracellular Ca2+ leak via oxidized and nitrosylated RyR2 channels, activated ER stress response, mitochondrial dysfunction, and decreased fuel-stimulated insulin release. Additionally, we verified the effects of the pharmacological inhibition of intracellular Ca2+ leak in CPVT-associated RyR2-expressing mice, in human islets from diabetic patients, and in an established murine model of type 2 diabetes mellitus. Taken together, our data indicate that RyR2 channels play a crucial role in the regulation of insulin secretion and glucose homeostasis.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| | - Gennaro Pagano
- Department of Medicine, Imperial College, London, United Kingdom
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
| | - Celestino Sardu
- Department of Electrophysiology, the Leiden University Medical Center (LUMC), Leiden, Netherlands
- Department of Medical, Surgical, Neurological, Metabolic, and Geriatric Sciences, Second University of Naples, Naples, Italy
- Department of Cardiovascular Disorders, Catholic University of the Sacred Heart, “John Paul II” Foundation for Research and Treatment, Campobasso, Italy
| | - Wenjun Xie
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| | - Salvatore Luca D’Ascia
- Department of Cardiology and Arrhythmology, Clinical Institute “Città Studi” Hospital, Milan, Italy
| | - Michele Cannone
- Division of Cardiology, G. Tatarella Hospital, Cerignola (Foggia), Italy
| | - Nicola Marziliano
- Division of Molecular Pathology, Niguarda Ca’ Granda Hospital, Milan, Italy
- Divisions of Medical Genetics and Cardiology, University Hospital of Parma, Parma, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
| | - Theresa A. Guise
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Alain Lacampagne
- U1046 INSERM, UMR 9214, CNRS, CHRU Montpellier, Montpellier, France
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
- Department of Medicine, Columbia University, New York, New York, USA
| |
Collapse
|
38
|
SALVATIERRA CRISTIANAS, REIS SÍLVIAR, PESSOA ANAF, SOUZA LETÍCIAMDE, STOPPIGLIA LUIZF, VELOSO ROBERTOV, REIS MARISEA, CARNEIRO EVERARDOM, BOSCHERO ANTONIOC, COLODEL EDSONM, ARANTES VANESSAC, LATORRACA MÁRCIAQ. Short-term low-protein diet during pregnancy alters islet area and protein content of phosphatidylinositol 3-kinase pathway in rats. ACTA ACUST UNITED AC 2015; 87:1007-18. [PMID: 25860970 DOI: 10.1590/0001-3765201520140251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 12/15/2014] [Indexed: 12/31/2022]
Abstract
The phosphatidylinositol 3-kinase and mitogen-activated protein kinase pathways mediate β cell growth, proliferation, survival and death. We investigated whether protein restriction during pregnancy alters islet morphometry or the expression and phosphorylation of several proteins involved in the phosphatidylinositol 3-kinase and mitogen-activated protein kinase pathways. As controls, adult pregnant and non-pregnant rats were fed a normal-protein diet (17%). Pregnant and non-pregnant rats in the experimental groups were fed a low-protein diet (6%) for 15 days. Low protein diet during pregnancy increased serum prolactin level, reduced serum corticosterone concentration and the expression of both protein kinase B/AKT1 (AKT1) and p70 ribosomal protein S6 kinase (p70S6K), as well as the islets area, but did not alter the insulin content of pancreatic islets. Pregnancy increased the expression of the Src homology/collagen (SHC) protein and the extracellular signal-regulated kinases 1/2 (ERK1/2) independent of diet. ERK1/2 phosphorylation (pERK1/2) was similar in islets from pregnant and non-pregnant rats fed a low-protein diet, and was higher in islets from pregnant rats than in islets from non-pregnant rats fed a normal-protein diet. Thus, a short-term, low-protein diet during pregnancy was sufficient to reduce the levels of proteins in the phosphatidylinositol 3-kinase pathway and affect islet morphometry.
Collapse
|
39
|
Hakuno F, Fukushima T, Yoneyama Y, Kamei H, Ozoe A, Yoshihara H, Yamanaka D, Shibano T, Sone-Yonezawa M, Yu BC, Chida K, Takahashi SI. The Novel Functions of High-Molecular-Mass Complexes Containing Insulin Receptor Substrates in Mediation and Modulation of Insulin-Like Activities: Emerging Concept of Diverse Functions by IRS-Associated Proteins. Front Endocrinol (Lausanne) 2015; 6:73. [PMID: 26074875 PMCID: PMC4443775 DOI: 10.3389/fendo.2015.00073] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/25/2015] [Indexed: 12/25/2022] Open
Abstract
Insulin-like peptides, such as insulin-like growth factors (IGFs) and insulin, induce a variety of bioactivities, such as growth, differentiation, survival, increased anabolism, and decreased catabolism in many cell types and in vivo. In general, IGFs or insulin bind to IGF-I receptor (IGF-IR) or insulin receptor (IR), activating the receptor tyrosine kinase. Insulin receptor substrates (IRSs) are known to be major substrates of receptor kinases, mediating IGF/insulin signals to direct bioactivities. Recently, we discovered that IRSs form high-molecular-mass complexes (referred to here as IRSomes) even without IGF/insulin stimulation. These complexes contain proteins (referred to here as IRSAPs; IRS-associated proteins), which modulate tyrosine phosphorylation of IRSs by receptor kinases, control IRS stability, and determine intracellular localization of IRSs. In addition, in these complexes, we found not only proteins that are involved in RNA metabolism but also RNAs themselves. Thus, IRSAPs possibly contribute to modulation of IGF/insulin bioactivities. Since it is established that disorder of modulation of insulin-like activities causes various age-related diseases including cancer, we could propose that the IRSome is an important target for treatment of these diseases.
Collapse
Affiliation(s)
- Fumihiko Hakuno
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Fukushima
- Laboratory of Biomedical Chemistry, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Biological Sciences, Faculty of Bioscience and Biotechnology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Yosuke Yoneyama
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyasu Kamei
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Atsufumi Ozoe
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidehito Yoshihara
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Daisuke Yamanaka
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takashi Shibano
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Meri Sone-Yonezawa
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Bu-Chin Yu
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Chida
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Takahashi
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- *Correspondence: Shin-Ichiro Takahashi, Laboratory of Cell Regulation, Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan,
| |
Collapse
|
40
|
Liu S, Xi Y, Bettaieb A, Matsuo K, Matsuo I, Kulkarni RN, Haj FG. Disruption of protein-tyrosine phosphatase 1B expression in the pancreas affects β-cell function. Endocrinology 2014; 155:3329-38. [PMID: 24956127 PMCID: PMC4138572 DOI: 10.1210/en.2013-2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Protein-tyrosine phosphatase 1B (PTP1B) is a physiological regulator of glucose homeostasis and energy balance. However, the role of PTP1B in pancreatic endocrine function remains largely unknown. To investigate the metabolic role of pancreatic PTP1B, we generated mice with pancreas PTP1B deletion (panc-PTP1B KO). Mice were fed regular chow or a high-fat diet, and metabolic parameters, insulin secretion and glucose tolerance were determined. On regular chow, panc-PTP1B KO and control mice exhibited comparable glucose tolerance whereas aged panc-PTP1B KO exhibited mild glucose intolerance. Furthermore, high-fat feeding promoted earlier impairment of glucose tolerance and attenuated glucose-stimulated insulin secretion in panc-PTP1B KO mice. The secretory defect in glucose-stimulated insulin secretion was recapitulated in primary islets ex vivo, suggesting that the effects were likely cell-autonomous. At the molecular level, PTP1B deficiency in vivo enhanced basal and glucose-stimulated tyrosyl phosphorylation of EphA5 in islets. Consistently, PTP1B overexpression in the glucose-responsive MIN6 β-cell line attenuated EphA5 tyrosyl phosphorylation, and substrate trapping identified EphA5 as a PTP1B substrate. In summary, these studies identify a novel role for PTP1B in pancreatic endocrine function.
Collapse
Affiliation(s)
- Siming Liu
- Nutrition Department (S.L., Y.X., A.B., K.M., I.M., F.G.H.), University of California Davis, Davis, California 95616; Joslin Diabetes Center (R.N.K.), Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215; and Division of Endocrinology, Diabetes and Metabolism (F.G.H.), Department of Internal Medicine, and Comprehensive Cancer Center, University of California Davis, Sacramento, California 95817
| | | | | | | | | | | | | |
Collapse
|
41
|
Zhang J, Liu F. Tissue-specific insulin signaling in the regulation of metabolism and aging. IUBMB Life 2014; 66:485-95. [PMID: 25087968 DOI: 10.1002/iub.1293] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/14/2014] [Indexed: 12/30/2022]
Abstract
In mammals, insulin signaling regulates glucose homeostasis and plays an essential role in metabolism, organ growth, development, fertility, and lifespan. The defects in this signaling pathway contribute to various metabolic diseases such as type 2 diabetes, polycystic ovarian disease, hypertension, hyperlipidemia, and atherosclerosis. However, reducing the insulin signaling pathway has been found to increase longevity and delay the aging-associated diseases in various animals, ranging from nematodes to mice. These seemly paradoxical findings raise an interesting question as to how modulation of the insulin signaling pathway could be an effective approach to improve metabolism and aging. In this review, we summarize current understanding on tissue-specific functions of insulin signaling in the regulation of metabolism and lifespan. We also discuss the potential benefits and limitations in modulating tissue-specific insulin signaling pathway to improve metabolism and healthspan.
Collapse
Affiliation(s)
- Jingjing Zhang
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education; Diabetes Center, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | |
Collapse
|
42
|
Paternally induced transgenerational inheritance of susceptibility to diabetes in mammals. Proc Natl Acad Sci U S A 2014; 111:1873-8. [PMID: 24449870 DOI: 10.1073/pnas.1321195111] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The global prevalence of prediabetes and type 2 diabetes (T2D) is increasing, and it is contributing to the susceptibility to diabetes and its related epidemic in offspring. Although the impacts of paternal impaired fasting blood glucose and glucose intolerance on the metabolism of offspring have been well established, the exact molecular and mechanistic basis that mediates these impacts remains largely unclear. Here we show that paternal prediabetes increases the susceptibility to diabetes in offspring through gametic epigenetic alterations. In our findings, paternal prediabetes led to glucose intolerance and insulin resistance in offspring. Relative to controls, offspring of prediabetic fathers exhibited altered gene expression patterns in the pancreatic islets, with down-regulation of several genes involved in glucose metabolism and insulin signaling pathways. Epigenomic profiling of offspring pancreatic islets revealed numerous changes in cytosine methylation depending on paternal prediabetes, including reproducible changes in methylation over several insulin signaling genes. Paternal prediabetes altered overall methylome patterns in sperm, with a large portion of differentially methylated genes overlapping with that of pancreatic islets in offspring. Our study uniquely revealed that prediabetes can be inherited transgenerationally through the mammalian germ line by an epigenetic mechanism.
Collapse
|
43
|
Dirice E, Kahraman S, Jiang W, El Ouaamari A, De Jesus DF, Teo AK, Hu J, Kawamori D, Gaglia JL, Mathis D, Kulkarni RN. Soluble factors secreted by T cells promote β-cell proliferation. Diabetes 2014; 63:188-202. [PMID: 24089508 PMCID: PMC3868047 DOI: 10.2337/db13-0204] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Type 1 diabetes is characterized by infiltration of pancreatic islets with immune cells, leading to insulin deficiency. Although infiltrating immune cells are traditionally considered to negatively impact β-cells by promoting their death, their contribution to proliferation is not fully understood. Here we report that islets exhibiting insulitis also manifested proliferation of β-cells that positively correlated with the extent of lymphocyte infiltration. Adoptive transfer of diabetogenic CD4(+) and CD8(+) T cells, but not B cells, selectively promoted β-cell proliferation in vivo independent from the effects of blood glucose or circulating insulin or by modulating apoptosis. Complementary to our in vivo approach, coculture of diabetogenic CD4(+) and CD8(+) T cells with NOD.RAG1(-/-) islets in an in vitro transwell system led to a dose-dependent secretion of candidate cytokines/chemokines (interleukin-2 [IL-2], IL-6, IL-10, MIP-1α, and RANTES) that together enhanced β-cell proliferation. These data suggest that soluble factors secreted from T cells are potential therapeutic candidates to enhance β-cell proliferation in efforts to prevent and/or delay the onset of type 1 diabetes.
Collapse
Affiliation(s)
- Ercument Dirice
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Sevim Kahraman
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Wenyu Jiang
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Abdelfattah El Ouaamari
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Dario F. De Jesus
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Adrian K.K. Teo
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Jiang Hu
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Dan Kawamori
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Jason L. Gaglia
- Section of Immunobiology, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA
| | - Rohit N. Kulkarni
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA
- Corresponding author: Rohit N. Kulkarni,
| |
Collapse
|
44
|
Zeng N, Yang KT, Bayan JA, He L, Aggarwal R, Stiles JW, Hou X, Medina V, Abad D, Palian BM, Al-Abdullah I, Kandeel F, Johnson DL, Stiles BL. PTEN controls β-cell regeneration in aged mice by regulating cell cycle inhibitor p16ink4a. Aging Cell 2013; 12:1000-11. [PMID: 23826727 DOI: 10.1111/acel.12132] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2013] [Indexed: 12/31/2022] Open
Abstract
Tissue regeneration diminishes with age, concurrent with declining hormone levels including growth factors such as insulin-like growth factor-1 (IGF-1). We investigated the molecular basis for such decline in pancreatic β-cells where loss of proliferation occurs early in age and is proposed to contribute to the pathogenesis of diabetes. We studied the regeneration capacity of β-cells in mouse model where PI3K/AKT pathway downstream of insulin/IGF-1 signaling is upregulated by genetic deletion of Pten (phosphatase and tensin homologue deleted on chromosome 10) specifically in insulin-producing cells. In this model, PTEN loss prevents the decline in proliferation capacity in aged β-cells and restores the ability of aged β-cells to respond to injury-induced regeneration. Using several animal and cell models where we can manipulate PTEN expression, we found that PTEN blocks cell cycle re-entry through a novel pathway leading to an increase in p16(ink4a), a cell cycle inhibitor characterized for its role in cellular senescence/aging. A downregulation in p16(ink4a) occurs when PTEN is lost as a result of cyclin D1 induction and the activation of E2F transcription factors. The activation of E2F transcriptional factors leads to methylation of p16(ink4a) promoter, an event that is mediated by the upregulation of polycomb protein, Ezh2. These analyses establish a novel PTEN/cyclin D1/E2F/Ezh2/p16(ink4a) signaling network responsible for the aging process and provide specific evidence for a molecular paradigm that explain how decline in growth factor signals such as IGF-1 (through PTEN/PI3K signaling) may control regeneration and the lack thereof in aging cells.
Collapse
Affiliation(s)
- Ni Zeng
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
| | - Kai-Ting Yang
- Department of Biochemistry; Keck School of Medicine; University of Southern California; Los Angeles CA 90033 USA
| | - Jennifer-Ann Bayan
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
| | - Lina He
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
| | - Richa Aggarwal
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
| | - Joseph W. Stiles
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
| | - Xiaogang Hou
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
| | - Vivian Medina
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
| | - Danny Abad
- Islet Transplant Center; City of Hope; Duarte CA 91010 USA
| | - Beth M. Palian
- Department of Biochemistry; Keck School of Medicine; University of Southern California; Los Angeles CA 90033 USA
| | | | - Fouad Kandeel
- Islet Transplant Center; City of Hope; Duarte CA 91010 USA
| | - Deborah L. Johnson
- Department of Biochemistry; Keck School of Medicine; University of Southern California; Los Angeles CA 90033 USA
| | - Bangyan L. Stiles
- Pharmacology and Pharmaceutical Sciences; School of Pharmacy; University of Southern California; Los Angeles CA 90089 USA
- Department of Pathology; Keck School of Medicine; University of Southern California; Los Angeles CA 90033 USA
| |
Collapse
|
45
|
Chowdhury A, Dyachok O, Tengholm A, Sandler S, Bergsten P. Functional differences between aggregated and dispersed insulin-producing cells. Diabetologia 2013; 56:1557-68. [PMID: 23604550 PMCID: PMC3671110 DOI: 10.1007/s00125-013-2903-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 03/12/2013] [Indexed: 11/27/2022]
Abstract
AIMS/HYPOTHESIS Beta cells situated in the islet of Langerhans respond more vigorously to glucose than do dissociated beta cells. Mechanisms for this discrepancy were studied by comparing insulin-producing MIN6 cells aggregated into pseudoislets with MIN6 monolayer cells and mouse and human islets. METHODS MIN6 monolayers, pseudoislets and mouse and human islets were exposed to glucose, α-ketoisocaproic acid (KIC), pyruvate, KIC plus glutamine and the phosphatidylinositol 3-kinase (PI3K) inhibitors LY294002 or wortmannin. Insulin secretion (ELISA), cytoplasmic Ca(2+) concentration ([Ca(2+)]c; microfluorometry), glucose oxidation (radiolabelling), the expression of genes involved in mitochondrial metabolism (PCR) and the phosphorylation of insulin receptor signalling proteins (western blotting) were measured. RESULTS Insulin secretory responses to glucose, pyruvate, KIC and glutamine were higher in pseudoislets than monolayers and comparable to those of human islets. Glucose oxidation and genes for mitochondrial metabolism were upregulated in pseudoislets compared with single cells and monolayers, respectively. Phosphorylation at the inhibitory S636/639 site of IRS-1 was significantly higher in monolayers and dispersed human and mouse cells than pseudoislets and intact human and mouse islets. PI3K inhibition only slightly attenuated glucose-stimulated insulin secretion from monolayers, but substantially reduced that from pseudoislets and human and mouse islets without suppressing the glucose-induced [Ca(2+)]c response. CONCLUSIONS/INTERPRETATION We propose that islet architecture is critical for proper beta cell mitochondrial metabolism and IRS-1 signalling, and that PI3K regulates insulin secretion at a step distal to the elevation of [Ca(2+)]c.
Collapse
Affiliation(s)
- A Chowdhury
- Department of Medical Cell Biology, Uppsala University, Box 571, 75123, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
46
|
Ezzat S, Zheng L, Florez JC, Stefan N, Mayr T, Hliang MM, Jablonski K, Harden M, Stančáková A, Laakso M, Haring HU, Ullrich A, Asa SL. The cancer-associated FGFR4-G388R polymorphism enhances pancreatic insulin secretion and modifies the risk of diabetes. Cell Metab 2013; 17:929-940. [PMID: 23747250 PMCID: PMC4005358 DOI: 10.1016/j.cmet.2013.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 01/18/2013] [Accepted: 04/05/2013] [Indexed: 12/11/2022]
Abstract
The fibroblast growth factor receptor 4 (FGFR4)-R388 single-nucleotide polymorphism has been associated with cancer risk and prognosis. Here we show that the FGFR4-R388 allele yields a receptor variant that preferentially promotes STAT3/5 signaling. This STAT activation transcriptionally induces Grb14 in pancreatic endocrine cells to promote insulin secretion. Knockin mice with the FGFR4 variant allele develop pancreatic islets that secrete more insulin, a feature that is reversed through Grb14 deletion and enhanced with FGF19 administration. We also show in humans that the FGFR4-R388 allele enhances islet function and may protect against type 2 diabetes. These data support a common genetic link underlying cancer and hyperinsulinemia.
Collapse
Affiliation(s)
- Shereen Ezzat
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada.
| | - Lei Zheng
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Jose C Florez
- Center for Human Genetic Research and Diabetes Research Center (Diabetes Unit), Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02142, USA
| | | | - Thomas Mayr
- Department of Molecular Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Maw Maw Hliang
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Kathleen Jablonski
- The Biostatistics Center, George Washington University, Rockville, MD 20852, USA
| | - Maegan Harden
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | | | - Axel Ullrich
- Department of Molecular Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Sylvia L Asa
- Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
47
|
Kolic J, Spigelman AF, Plummer G, Leung E, Hajmrle C, Kin T, Shapiro AMJ, Manning Fox JE, MacDonald PE. Distinct and opposing roles for the phosphatidylinositol 3-OH kinase catalytic subunits p110α and p110β in the regulation of insulin secretion from rodent and human beta cells. Diabetologia 2013; 56:1339-1349. [PMID: 23568272 DOI: 10.1007/s00125-013-2882-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 02/18/2013] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Phosphatidylinositol 3-OH kinases (PI3Ks) regulate beta cell mass, gene transcription, and function, although the contribution of the specific isoforms is unknown. As reduced type 1A PI3K signalling is thought to contribute to impaired insulin secretion, we investigated the role of the type 1A PI3K catalytic subunits α and β (p110α and -β) in insulin granule recruitment and exocytosis in rodent and human islets. METHODS The p110α and p110β subunits were inhibited pharmacologically or by small hairpin (sh)RNA-mediated knockdown, and were directly infused or overexpressed in mouse and human islets, beta cells and INS-1 832/13 cells. Glucose-stimulated insulin secretion (GSIS), single-cell exocytosis, Ca(2+) signalling, plasma membrane granule localisation, and actin density were monitored. RESULTS Inhibition or knockdown of p110α increased GSIS. This was not due to altered Ca(2+) responses, depolymerisation of cortical actin or increased cortical granule density, but to enhanced Ca(2+)-dependent exocytosis. Intracellular infusion of recombinant PI3Kα (p110α/p85β) blocked exocytosis. Conversely, knockdown (but not pharmacological inhibition) of p110β blunted GSIS, reduced cortical granule density and impaired exocytosis. Exocytosis was rescued by direct intracellular infusion of recombinant PI3Kβ (p110β/p85β) even when p110β catalytic activity was inhibited. Conversely, both the wild-type p110β and a catalytically inactive mutant directly facilitated exocytosis. CONCLUSIONS/INTERPRETATION Type 1A PI3K isoforms have distinct and opposing roles in the acute regulation of insulin secretion. While p110α acts as a negative regulator of beta cell exocytosis and insulin secretion, p110β is a positive regulator of insulin secretion through a mechanism separate from its catalytic activity.
Collapse
Affiliation(s)
- J Kolic
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada T6G 2E1
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Prudente S, Morini E, Marselli L, Baratta R, Copetti M, Mendonca C, Andreozzi F, Chandalia M, Pellegrini F, Bailetti D, Alberico F, Shah H, Abate N, Sesti G, Frittitta L, Marchetti P, Doria A, Trischitta V. Joint effect of insulin signaling genes on insulin secretion and glucose homeostasis. J Clin Endocrinol Metab 2013; 98:E1143-7. [PMID: 23633196 PMCID: PMC6618023 DOI: 10.1210/jc.2012-4282] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CONTEXT Reduced insulin signaling in insulin secreting β-cells causes defective insulin secretion and hyperglycemia in mice. OBJECTIVE We investigated whether functional polymorphisms affecting insulin signaling (ie, ENPP1 K121Q, rs1044498; IRS1 G972R, rs1801278; and TRIB3 Q84R, rs2295490) exert a joint effect on insulin secretion and abnormal glucose homeostasis (AGH). DESIGN Insulin secretion was evaluated by 1) the disposition index (DI) from an oral glucose tolerance test (OGTT) in 829 individuals; 2) insulin secretion stimulation index (SI) in islets from nondiabetic donors after glucose (n = 92) or glibenclamide (n = 89) stimulation. AGH (including impaired fasting glucose and/or impaired glucose tolerance or type 2 diabetes; T2D) was evaluated in case-control studies from the GENetics of Type 2 Diabetes in Italy and the United States (GENIUS T2D) Consortium (n = 6607). RESULTS Genotype risk score, obtained by totaling individual weighted risk allele effects, was associated with the following: 1) DI (P = .005); 2) glucose and glibenclamide SI (P = .046 and P = .009); or 3) AGH (odds ratio 1.08, 95% confidence interval 1.03-1.13; P = .001). We observed an inverse relationship between genetic effect and age at AGH onset, as indicated by a linear correlation between AGH-genotype risk score odds ratios and age-at-diagnosis cutoffs (R(2) = 0.80, P < .001). CONCLUSIONS Functional polymorphisms affecting insulin signaling exert a joint effect on both in vivo and in vitro insulin secretion as well as on early-onset AGH. Our data provide further evidence that abnormal insulin signaling reduces β-cell function and impairs glucose homeostasis.
Collapse
Affiliation(s)
- Sabrina Prudente
- Casa Sollievo della Sofferenza-Mendel Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Timing of maternal exposure to a high fat diet and development of obesity and hyperinsulinemia in male rat offspring: same metabolic phenotype, different developmental pathways? J Nutr Metab 2013; 2013:517384. [PMID: 23762542 PMCID: PMC3666195 DOI: 10.1155/2013/517384] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/08/2013] [Accepted: 04/20/2013] [Indexed: 12/22/2022] Open
Abstract
Objective. Offspring born to mothers either fed an obesogenic diet throughout their life or restricted to pregnancy and lactation demonstrate obesity, hyperinsulinemia, and hyperleptinemia, irrespective of their postweaning diet. We examined whether timing of a maternal obesogenic diet results in differential regulation of pancreatic adipoinsular and inflammatory signaling pathways in offspring. Methods. Female Wistar rats were randomized into 3 groups: (1) control (CONT): fed a control diet preconceptionally and during pregnancy and lactation; (2) maternal high fat (MHF): fed an HF diet throughout their life and during pregnancy and lactation; (3) pregnancy and lactation HF (PLHF): fed a control diet throughout life until mating, then HF diet during pregnancy and lactation. Male offspring were fed the control diet postweaning. Plasma and pancreatic tissue were collected, and mRNA concentrations of key factors regulating adipoinsular axis signaling were determined. Results. MHF and PLHF offspring exhibited increased adiposity and were hyperinsulinemic and hyperleptinemic compared to CONT. Despite a similar anthropometric phenotype, MHF and PLHF offspring exhibited distinctly different expression for key pancreatic genes, dependent upon maternal preconceptional nutritional background. Conclusions. These data suggest that despite using differential signaling pathways, obesity in offspring may be an adaptive outcome of early life exposure to HF during critical developmental windows.
Collapse
|
50
|
Feng X, Tucker KL, Parnell LD, Shen J, Lee YC, Ordovás JM, Ling WH, Lai CQ. Insulin receptor substrate 1 (IRS1) variants confer risk of diabetes in the Boston Puerto Rican Health Study. Asia Pac J Clin Nutr 2013; 22:150-9. [PMID: 23353623 DOI: 10.6133/apjcn.2013.22.1.09] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Published data concerning associations between IRS1 variants and type 2 diabetes and related traits have been inconsistent. We examined the relationship between common variants in IRS1, type 2 diabetes, and related traits including insulin resistance, hyperglycemia and DNA damage in the Boston Puerto Rican Health Study. METHODS We genotyped six common IRS1 variants in an adult Puerto Rican population (n=1132) and tested for association with risk of type 2 diabetes and related traits. RESULTS SNPs rs934167 and rs1801123 showed significant association with fasting glucose concentrations (p = 0.005 and p = 0.016, respectively) and rs934167 showed significant association with plasma insulin levels (p = 0.005). Carriers of the rs934167 minor allele had significantly higher HOMA-IR and lower QUICKI (p = 0.001 and p = 0.001, respectively), and a 40% and 58% greater likelihood of being hyperglycaemic or hyperinsulinemic (OR = 1.40 and 1.58; p = 0.013 and 0.002, respectively). However, they exhibited only a marginally significant trend towards having type 2 diabetes (OR=1.27, p = 0.077). Furthermore, carriers of the haplotype C-T of the rs934167 and rs1801123 minor alleles showed consistent patterns of associations after correction for multiple testing. In addition, the G972R (rs1801278) minor allele was significantly associated with higher urinary 8-OHdG concentrations (p = 0.020) and plasma CRP levels (p = 0.035). CONCLUSIONS Our results support IRS1 variants associated with type 2 diabetes risk in adult Puerto Ricans. Moreover, we report the novel finding that IRS1 variant G972R (rs1801278) may contribute to oxidative DNA damage and inflammation.
Collapse
Affiliation(s)
- Xiang Feng
- Departmennt of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|