1
|
Brito Nunes C, Borges MC, Freathy RM, Lawlor DA, Qvigstad E, Evans DM, Moen GH. Understanding the Genetic Landscape of Gestational Diabetes: Insights into the Causes and Consequences of Elevated Glucose Levels in Pregnancy. Metabolites 2024; 14:508. [PMID: 39330515 PMCID: PMC11434570 DOI: 10.3390/metabo14090508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Background/Objectives: During pregnancy, physiological changes in maternal circulating glucose levels and its metabolism are essential to meet maternal and fetal energy demands. Major changes in glucose metabolism occur throughout pregnancy and consist of higher insulin resistance and a compensatory increase in insulin secretion to maintain glucose homeostasis. For some women, this change is insufficient to maintain normoglycemia, leading to gestational diabetes mellitus (GDM), a condition characterized by maternal glucose intolerance and hyperglycaemia first diagnosed during the second or third trimester of pregnancy. GDM is diagnosed in approximately 14.0% of pregnancies globally, and it is often associated with short- and long-term adverse health outcomes in both mothers and offspring. Although recent studies have highlighted the role of genetic determinants in the development of GDM, research in this area is still lacking, hindering the development of prevention and treatment strategies. Methods: In this paper, we review recent advances in the understanding of genetic determinants of GDM and glycaemic traits during pregnancy. Results/Conclusions: Our review highlights the need for further collaborative efforts as well as larger and more diverse genotyped pregnancy cohorts to deepen our understanding of the genetic aetiology of GDM, address research gaps, and further improve diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Caroline Brito Nunes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Rachel M. Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 4PY, UK;
| | - Deborah A. Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Elisabeth Qvigstad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - David M. Evans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Frazer Institute, University of Queensland, Brisbane 4102, Australia
| | - Gunn-Helen Moen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Frazer Institute, University of Queensland, Brisbane 4102, Australia
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
2
|
Stamateris RE, Landa-Galvan HV, Sharma RB, Darko C, Redmond D, Rane SG, Alonso LC. Noncanonical CDK4 signaling rescues diabetes in a mouse model by promoting β cell differentiation. J Clin Invest 2023; 133:e166490. [PMID: 37712417 PMCID: PMC10503800 DOI: 10.1172/jci166490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Abstract
Expanding β cell mass is a critical goal in the fight against diabetes. CDK4, an extensively characterized cell cycle activator, is required to establish and maintain β cell number. β cell failure in the IRS2-deletion mouse type 2 diabetes model is, in part, due to loss of CDK4 regulator cyclin D2. We set out to determine whether replacement of endogenous CDK4 with the inhibitor-resistant mutant CDK4-R24C rescued the loss of β cell mass in IRS2-deficient mice. Surprisingly, not only β cell mass but also β cell dedifferentiation was effectively rescued, despite no improvement in whole body insulin sensitivity. Ex vivo studies in primary islet cells revealed a mechanism in which CDK4 intervened downstream in the insulin signaling pathway to prevent FOXO1-mediated transcriptional repression of critical β cell transcription factor Pdx1. FOXO1 inhibition was not related to E2F1 activity, to FOXO1 phosphorylation, or even to FOXO1 subcellular localization, but rather was related to deacetylation and reduced FOXO1 abundance. Taken together, these results demonstrate a differentiation-promoting activity of the classical cell cycle activator CDK4 and support the concept that β cell mass can be expanded without compromising function.
Collapse
Affiliation(s)
- Rachel E. Stamateris
- MD/PhD Program, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Huguet V. Landa-Galvan
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| | - Rohit B. Sharma
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| | - Christine Darko
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| | - David Redmond
- Hartman Institute for Therapeutic Regenerative Medicine, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Sushil G. Rane
- Integrative Cellular Metabolism Section, Diabetes, Endocrinology and Obesity Branch, National Institute for Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Laura C. Alonso
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| |
Collapse
|
3
|
Li M, Zhang R, Ge Q, Yue L, Ma D, Khattab F, Xie W, Cui Y, Gilon P, Zhao X, Li X, Cheng R. Chemerin as an Inducer of β Cell Proliferation Mediates Mitochondrial Homeostasis and Promotes β Cell Mass Expansion. Int J Mol Sci 2023; 24:ijms24119136. [PMID: 37298086 DOI: 10.3390/ijms24119136] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Loss of the β cell population is a crucial feature of type 2 diabetes. Restoring the β cell mass by stimulating β cell proliferation and preventing its apoptosis was proposed as a therapeutic approach to treating diabetes. Therefore, researchers have been increasingly interested in identifying exogenous factors that can stimulate β cell proliferation in situ and in vitro. Adipokine chemerin, which is secreted from adipose tissue and the liver, has been identified as a chemokine that plays a critical role in the regulation of metabolism. In this study, we demonstrate that chemerin as a circulating adipokine promotes β cell proliferation in vivo and in vitro. Chemerin serum levels and the expression of the main receptors within islets are highly regulated under a variety of challenging conditions, including obesity and type 2 diabetes. As compared to their littermates, mice overexpressing chemerin had a larger islet area and increased β cell mass with both a normal and high-fat diet. Moreover, in chemerin-overexpressed mice, we observed improved mitochondrial homeostasis and increased insulin synthesis. In summary, our findings confirm the potential role of chemerin as an inducer of β cell proliferation, and they provide novel insights into the helpful strategy to expand β cell population.
Collapse
Affiliation(s)
- Min Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ruifan Zhang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qian Ge
- The First Clinical College, Chongqing Medical University, Chongqing 400016, China
| | - Lingzhi Yue
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Dan Ma
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Firas Khattab
- Pôle d'Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Wenhua Xie
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yewei Cui
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Patrick Gilon
- Pôle d'Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Xueya Zhao
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Rui Cheng
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
4
|
Lopes JB, Małz M, Senko AN, Zocher S, Kempermann G. Loss of individualized behavioral trajectories in adult neurogenesis-deficient cyclin D2 knockout mice. Hippocampus 2023; 33:360-372. [PMID: 36880417 DOI: 10.1002/hipo.23522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/14/2023] [Accepted: 02/19/2023] [Indexed: 03/08/2023]
Abstract
There is still limited mechanistic insight into how the interaction of individuals with their environment results in the emergence of individuality in behavior and brain structure. Nevertheless, the idea that personal activity shapes the brain is implicit in strategies for healthy cognitive aging as well as in the idea that individuality is reflected in the brain's connectome. We have shown that even isogenic mice kept in a shared enriched environment (ENR) developed divergent and stable social and exploratory trajectories. As these trajectories-measured as roaming entropy (RE)-positively correlated with adult hippocampal neurogenesis, we hypothesized that a feedback between behavioral activity and adult hippocampal neurogenesis might be a causal factor in brain individualization. We used cyclin D2 knockout mice with constitutively extremely low levels of adult hippocampal neurogenesis and their wild-type littermates. We housed them for 3 months in a novel ENR paradigm, consisting of 70 connected cages equipped with radio frequency identification antennae for longitudinal tracking. Cognitive performance was evaluated in the Morris Water Maze task (MWM). With immunohistochemistry we confirmed that adult neurogenesis correlated with RE in both genotypes and that D2 knockout mice had the expected impaired performance in the reversal phase of the MWM. But whereas the wild-type animals developed stable exploratory trajectories with increasing variance, correlating with adult neurogenesis, this individualizing phenotype was absent in D2 knockout mice. Here the behaviors started out more random and revealed less habituation and low variance. Together, these findings suggest that adult neurogenesis contributes to experience-dependent brain individualization.
Collapse
Affiliation(s)
- Jadna Bogado Lopes
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Monika Małz
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Anna N Senko
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Sara Zocher
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
| |
Collapse
|
5
|
Lei Z, Chen Y, Wang J, Zhang Y, Shi W, Wang X, Xing D, Li D, Jiao X. Txnip deficiency promotes β-cell proliferation in the HFD-induced obesity mouse model. Endocr Connect 2022; 11:EC-21-0641. [PMID: 35294398 PMCID: PMC9066588 DOI: 10.1530/ec-21-0641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/25/2022]
Abstract
Elucidating the mechanisms of regulation of β-cell proliferation is key to understanding the pathogenesis of diabetes mellitus. Txnip is a tumor suppressor that is upregulated in diabetes and plays an important role in the regulation of insulin sensitivity; however, its potential effect on pancreatic β-cell proliferation remains unclear. Here, we evaluated the role of Txnip in pancreatic β-cell compensatory proliferation by subjecting WT and Txnip knockout (KO) mice to a high-fat diet (HFD). Our results demonstrate that Txnip deficiency improves glucose tolerance and increases insulin sensitivity in HFD-induced obesity. The antidiabetogenic effect of Txnip deficiency was accompanied by increased β-cell proliferation and enhanced β-cell mass expansion. Furthermore, Txnip deficiency modulated the expression of a set of transcription factors with key roles in β-cell proliferation and cell cycle regulation. Txnip KO in HFD mice also led to activated levels of p-PI3K, p-AKT, p-mTOR and p-GSK3β, suggesting that Txnip may act via PI3K/AKT signaling to suppress β-cell proliferation. Thus, our work provides a theoretical basis for Txnip as a new therapeutic target for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Zhandong Lei
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
- Department of Anatomy, Shanxi Medical University, Taiyuan, China
| | - Yunfei Chen
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Jin Wang
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Yan Zhang
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Wenjuan Shi
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Xuejiao Wang
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Dehai Xing
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Dongxue Li
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Xiangying Jiao
- Key Laboratory of Cellular Physiology, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
- Correspondence should be addressed to X Jiao:
| |
Collapse
|
6
|
Lee JH, Lee J. Endoplasmic Reticulum (ER) Stress and Its Role in Pancreatic β-Cell Dysfunction and Senescence in Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23094843. [PMID: 35563231 PMCID: PMC9104816 DOI: 10.3390/ijms23094843] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023] Open
Abstract
An increased life span and accompanying nutritional affluency have led to a rapid increase in diseases associated with aging, such as obesity and type 2 diabetes, imposing a tremendous economic and health burden on society. Pancreatic β-cells are crucial for controlling glucose homeostasis by properly producing and secreting the glucose-lowering hormone insulin, and the dysfunction of β-cells determines the outcomes for both type 1 and type 2 diabetes. As the native structure of insulin is formed within the endoplasmic reticulum (ER), ER homeostasis should be appropriately maintained to allow for the proper metabolic homeostasis and functioning of β-cells. Recent studies have found that cellular senescence is critically linked with cellular stresses, including ER stress, oxidative stress, and mitochondrial stress. These studies implied that β-cell senescence is caused by ER stress and other cellular stresses and contributes to β-cells’ dysfunction and the impairment of glucose homeostasis. This review documents and discusses the current understanding of cellular senescence, β-cell function, ER stress, its associated signaling mechanism (unfolded protein response), and the effect of ER stress on β-cell senescence and dysfunction.
Collapse
Affiliation(s)
- Ji-Hye Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Correspondence:
| |
Collapse
|
7
|
Hwang YJ, Jung GS, Jeon WB, Lee KM. Arg-Gly-Asp-modified elastin-like polypeptide regulates cell proliferation and cell cycle proteins via the phosphorylation of Erk and Akt in pancreatic β-cell. Heliyon 2020; 6:e04918. [PMID: 32995613 PMCID: PMC7501433 DOI: 10.1016/j.heliyon.2020.e04918] [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: 05/13/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 11/24/2022] Open
Abstract
Objective Enhancement of β-cell proliferation plays an important role in maintaining β-cell mass and function, and in improving pancreatic β-cell survival before transplantation. Extracellular matrix (ECM) components increase the adhesion and proliferation of β-cells, and the RGD-modified elastin-like polypeptide (RGD-ELP, REP) has been described as a bioactive matrix. In this study, we investigated whether REP could enhance β-cell adhesion and proliferation and elucidated the signaling pathways involved. Methods We investigated the effect of REP on cell adhesion, proliferation and insulin secretion via assays using Rin-m and rat islets. Crystal violet, CCK-8, and BrdU assay, FACS, western blot, real time q-PCR analyses and insulin ELISA were examined. To explain the associated mechanisms, phosphorylation of Akt and extracellular signal-regulated kinase (Erk) were measured. Results REP more increased the adhesion, proliferation and survival of Rin-m cells compared to elastin-like poly peptide (ELP) without RGD-motif. The enhancement of β-cell proliferation by REP was associated with increased cyclin D1, cyclin D2 and cdk6, and decreased p27 levels. When β-cells were cultured on REP, Erk and the phosphatidylinositol 3-kinase (PI3-kinase) downstream effector, Akt was stimulated. Treatment with the Erk pathway inhibitor and PI3-kinase inhibitor decreased REP-induced β-cell adhesion and proliferation, and regulated REP-induced cell cycle proteins. Additionally, REP increased the mRNA and protein levels of insulin and its transcription factor, PDX-1, and insulin secretion. Conclusions Our results demonstrate that the up-regulation of the PI3K/Akt and Erk signaling pathways and the regulation of cell cycle proteins by REP could serve as effective strategies for improving pancreatic β-cell adhesion and proliferation.
Collapse
Affiliation(s)
- Yeo Jin Hwang
- Division of Electronics & Information System, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Gwon-Soo Jung
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Won Bae Jeon
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Kyeong-Min Lee
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| |
Collapse
|
8
|
Oliveira KM, Figueiredo LS, Araujo TR, Freitas IN, Silva JN, Boschero AC, Ribeiro RA. Prolonged bisphenol-A exposure decreases endocrine pancreatic proliferation in response to obesogenic diet in ovariectomized mice. Steroids 2020; 160:108658. [PMID: 32442623 DOI: 10.1016/j.steroids.2020.108658] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/24/2020] [Accepted: 05/14/2020] [Indexed: 12/23/2022]
Abstract
Research on the deleterious actions of bisphenol (BP)-A have focused on its effects on insulin secretion during pre/perinatal periods or adulthood. Estrogens also modulate endocrine pancreas physiology in females during aging; however, the effects of BPA on islet morphophysiology after menopause have not been investigated. We evaluated the effects of BPA exposure on glucose homeostasis and islet morphofunction in ovariectomized (OVX) mice fed on a high-fat diet (HFD). Adult Swiss female mice were underwent to bilateral ovariectomy, and with the confirmation of the establishment of surgical menopause, the females were then submitted, or not,to a normolipidic diet or HFD [control (CTL) and HFD groups, respectively] without or with 1 μg/mL BPA in their drinking water (CBPA and HBPA groups) for 90 days. HFD females displayed obesity, hyperglycemia, hyperinsulinemia, glucose intolerance and insulin resistance. BPA did not modulate HFD-induced obesity or body glucose impairments in HBPA females, and islets isolated from both the HFD and HBPA groups exhibited insulin hypersecretion. The HBPA islets, however, displayed enlarged islet cells and reduced proliferation, in association with the downregulation of mRNAs encoding PDX-1, NGN3 and CCND2 and upregulation of mRNAs encoding ER-β, GPR30, TNF-α and IL-1β in HBPA islets. BPA consumption in OVX mice impaired the islet-cell hyperplasia response to the HFD, partly mediated by increased expression of ER-β and GPR30, which impaired the expression of major genes involved in islet-cell survival and functionality. Together with higher pro-inflammatory cytokines expression in the islet milieu, these alterations may accelerate β-cell failure in postmenopause.
Collapse
Affiliation(s)
- Kênia M Oliveira
- Federal University of Rio de Janeiro, Campus UFRJ-Macaé, Macaé, RJ, Brazil
| | | | - Thiago R Araujo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Israelle N Freitas
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Juliana N Silva
- Federal University of Rio de Janeiro, Campus UFRJ-Macaé, Macaé, RJ, Brazil
| | - Antonio C Boschero
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Rosane A Ribeiro
- Federal University of Rio de Janeiro, Campus UFRJ-Macaé, Macaé, RJ, Brazil.
| |
Collapse
|
9
|
Abstract
BACKGROUND Pancreatic β-cells adapt to high metabolic demand by expanding their β-cell mass and/or enhancing insulin secretion to maintain glucose homeostasis. Type 2 diabetes (T2D) is typically characterized by β-cell decompensation. SCOPE OF THE REVIEW The current review focuses on summarizing the "omics" and "epi-omics" approaches that particularly focus on addressing the β-cell adaptation to insulin resistance and T2D. MAJOR CONCLUSIONS The molecular mechanisms underlying successful versus compromised β-cell adaptation to insulin resistance are not entirely understood. The last decade has seen an exponential increase in the use of "omics" and "epi-omics" approaches to dissect pathophysiology of metabolic diseases. One recent example is the emergence of m6A mRNA methylation as a new layer of regulation of gene expression with the potential to impact diverse physiological processes in metabolic cells.
Collapse
Affiliation(s)
- Dario F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Harvard Stem Cell Institute, Boston, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Harvard Stem Cell Institute, Boston, USA.
| |
Collapse
|
10
|
Rosselot C, Kumar A, Lakshmipathi J, Zhang P, Lu G, Katz LS, Prochownik EV, Stewart AF, Lambertini L, Scott DK, Garcia-Ocaña A. Myc Is Required for Adaptive β-Cell Replication in Young Mice but Is Not Sufficient in One-Year-Old Mice Fed With a High-Fat Diet. Diabetes 2019; 68:1934-1949. [PMID: 31292135 PMCID: PMC6754239 DOI: 10.2337/db18-1368] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 07/02/2019] [Indexed: 12/18/2022]
Abstract
Failure to expand pancreatic β-cells in response to metabolic stress leads to excessive workload resulting in β-cell dysfunction, dedifferentiation, death, and development of type 2 diabetes. In this study, we demonstrate that induction of Myc is required for increased pancreatic β-cell replication and expansion during metabolic stress-induced insulin resistance with short-term high-fat diet (HFD) in young mice. β-Cell-specific Myc knockout mice fail to expand adaptively and show impaired glucose tolerance and β-cell dysfunction. Mechanistically, PKCζ, ERK1/2, mTOR, and PP2A are key regulators of the Myc response in this setting. DNA methylation analysis shows hypomethylation of cell cycle genes that are Myc targets in islets from young mice fed with a short-term HFD. Importantly, DNA hypomethylation of Myc response elements does not occur in islets from 1-year-old mice fed with a short-term HFD, impairing both Myc recruitment to cell cycle regulatory genes and β-cell replication. We conclude that Myc is required for metabolic stress-mediated β-cell expansion in young mice, but with aging, Myc upregulation is not sufficient to induce β-cell replication by, at least partially, an epigenetically mediated resistance to Myc action.
Collapse
Affiliation(s)
- Carolina Rosselot
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anil Kumar
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jayalakshmi Lakshmipathi
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Pili Zhang
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Geming Lu
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liora S Katz
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Edward V Prochownik
- Division of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA
- Department of Microbiology & Molecular Genetics, University of Pittsburgh Medical Center, Hillman Cancer Center, and Pittsburgh Liver Research Center, Pittsburgh, PA
| | - Andrew F Stewart
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Luca Lambertini
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Donald K Scott
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adolfo Garcia-Ocaña
- Division of Endocrinology, Diabetes and Bone Diseases, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
11
|
Hu L, He F, Luo Y, Luo H, Hai L, Li Y, Zhou Z, Liu F, Dai YS. Reduced Compensatory β-Cell Proliferation in Nfatc3-Deficient Mice Fed on High-Fat Diet. Exp Clin Endocrinol Diabetes 2019; 129:651-660. [PMID: 31546271 DOI: 10.1055/a-1008-9110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND High-fat-diet induces pancreatic β-cell compensatory proliferation, and impairments in pancreatic β-cell proliferation and function can lead to defects in insulin secretion and diabetes. NFATc3 is important for HFD-induced adipose tissue inflammation. But it is unknown whether NFATc3 is required for β cell compensatory growth in mice fed with HFD. METHODS NFATc3 mRNA and protein expression levels were quantified by RT-qPCR and Western blotting, respectively, in pancreatic islets of WT mice fed on HFD for 12-20 weeks. Adenoviral-mediated overexpression of NFATc3 were conducted in Min6 cells and cultured primary mouse islets. NFATc3-/- mice and WT control mice were fed with HFD and metabolic and functional parameters were measured. RESULTS We observed that the NFATc3 expression level was reduced in the islets of high-fat-diet (HFD)-fed mice. Adenovirus-mediated overexpression of NFATc3 enhanced glucose-stimulated insulin secretion and β-cell gene expression in cultured primary mouse islets. Nfatc3-/- mice initially developed similar glucose tolerance at 2-4 weeks after HFD feeding than HFD-fed WT mice, but Nfatc3-/- mice developed improved glucose tolerance and insulin sensitivity after 8 weeks of HFD feeding compared to Nfatc3+/+fed with HFD. Furthermore, Nfatc3-/- mice on HFD exhibited decreased β-cell mass and reduced expression of genes important for β-cell proliferation and function compared to Nfatc3+/+mice on HFD. CONCLUSIONS The findings suggested that NFATc3 plays a role in maintaining the pancreatic β-cell compensatory growth and gene expression in response to obesity.
Collapse
Affiliation(s)
- Li Hu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fengli He
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hairong Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luo Hai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yabin Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Yan-Shan Dai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Bristol-Myers Squibb Company, Princeton, NJ 08540, United States
| |
Collapse
|
12
|
Ji Y, Sun S, Shrestha N, Darragh LB, Shirakawa J, Xing Y, He Y, Carboneau BA, Kim H, An D, Ma M, Oberholzer J, Soleimanpour SA, Gannon M, Liu C, Naji A, Kulkarni RN, Wang Y, Kersten S, Qi L. Toll-like receptors TLR2 and TLR4 block the replication of pancreatic β cells in diet-induced obesity. Nat Immunol 2019; 20:677-686. [PMID: 31110312 PMCID: PMC6531334 DOI: 10.1038/s41590-019-0396-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Abstract
Consumption of a high-energy Western diet triggers mild adaptive β cell proliferation to compensate for peripheral insulin resistance; however, the underlying molecular mechanism remains unclear. In the present study we show that the toll-like receptors TLR2 and TLR4 inhibited the diet-induced replication of β cells in mice and humans. The combined, but not the individual, loss of TLR2 and TLR4 increased the replication of β cells, but not that of α cells, leading to enlarged β cell area and hyperinsulinemia in diet-induced obesity. Loss of TLR2 and TLR4 increased the nuclear abundance of the cell cycle regulators cyclin D2 and Cdk4 in a manner dependent on the signaling mediator Erk. These data reveal a regulatory mechanism controlling the proliferation of β cells in diet-induced obesity and suggest that selective targeting of the TLR2/TLR4 pathways may reverse β cell failure in patients with diabetes.
Collapse
Affiliation(s)
- Yewei Ji
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Shengyi Sun
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Center for Molecular Medicine and Genetics, Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, USA
| | - Neha Shrestha
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Laurel B Darragh
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Department of Radiation Oncology, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Jun Shirakawa
- Section of 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
| | - Yuan Xing
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Yi He
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Bethany A Carboneau
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hana Kim
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- XBiotech USA, Inc., Austin, TX, USA
| | - Duo An
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Jose Oberholzer
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Scott A Soleimanpour
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maureen Gannon
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chengyang Liu
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rohit N Kulkarni
- Section of 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
| | - Yong Wang
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Sander Kersten
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Nutrition Metabolism and Genomics group, Wageningen University, Wageningen, the Netherlands
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA.
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Masuda A, Seino Y, Murase M, Hidaka S, Shibata M, Takayanagi T, Sugimura Y, Hayashi Y, Suzuki A. Short-Term High-Starch, Low-Protein Diet Induces Reversible Increase in β-cell Mass Independent of Body Weight Gain in Mice. Nutrients 2019; 11:nu11051045. [PMID: 31083314 PMCID: PMC6566232 DOI: 10.3390/nu11051045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 12/26/2022] Open
Abstract
Long-term exposure to a high starch, low-protein diet (HSTD) induces body weight gain and hyperinsulinemia concomitantly with an increase in β-cell mass (BCM) and pancreatic islets number in mice; however, the effect of short-term exposure to HSTD on BCM and islet number has not been elucidated. In the present study, we investigated changes in body weight, plasma insulin levels, BCM and islet number in mice fed HSTD for 5 weeks followed by normal chow (NC) for 2 weeks. BCM and islet number were increased in mice fed HSTD for 5 weeks compared with those in mice fed NC. On the other hand, mice fed HSTD for 5 weeks followed by NC for 2 weeks (SN) showed decreased BCM and insulin levels, compared to mice fed HSTD for 7 weeks, and no significant differences in these parameters were observed between SN and the control NC at 7 weeks. No significant difference in body weight was observed among HSTD, NC and SN fed groups. These results suggest that a high-starch diet induces an increase in BCM in a manner independent of body weight gain, and that 2 weeks of NC feeding is sufficient for the reversal of the morphological changes induced in islets by HSTD feeding.
Collapse
Affiliation(s)
- Atsushi Masuda
- Department of Endocrinology and Metabolism, Fujita Health University, Graduate School of Medicine, Toyoake 470-1192, Japan.
| | - Yusuke Seino
- Department of Endocrinology and Metabolism, Fujita Health University, Graduate School of Medicine, Toyoake 470-1192, Japan.
| | - Masatoshi Murase
- Departments of Endocrinology and Diabetes, Toyota Memorial Hospital, Toyota 471-8513, Japan.
| | - Shihomi Hidaka
- Department of Endocrinology and Metabolism, Fujita Health University, Graduate School of Medicine, Toyoake 470-1192, Japan.
| | - Megumi Shibata
- Department of Endocrinology and Metabolism, Fujita Health University, Graduate School of Medicine, Toyoake 470-1192, Japan.
| | - Takeshi Takayanagi
- Department of Endocrinology and Metabolism, Fujita Health University, Graduate School of Medicine, Toyoake 470-1192, Japan.
| | - Yoshihisa Sugimura
- Department of Endocrinology and Metabolism, Fujita Health University, Graduate School of Medicine, Toyoake 470-1192, Japan.
| | - Yoshitaka Hayashi
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 467-8601 Japan.
| | - Atsushi Suzuki
- Department of Endocrinology and Metabolism, Fujita Health University, Graduate School of Medicine, Toyoake 470-1192, Japan.
| |
Collapse
|
14
|
Murase M, Seino Y, Maekawa R, Iida A, Hosokawa K, Hayami T, Tsunekawa S, Hamada Y, Yokoi N, Seino S, Hayashi Y, Arima H. Functional adenosine triphosphate-sensitive potassium channel is required in high-carbohydrate diet-induced increase in β-cell mass. J Diabetes Investig 2019; 10:238-250. [PMID: 30084544 PMCID: PMC6400177 DOI: 10.1111/jdi.12907] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/26/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022] Open
Abstract
AIMS/INTRODUCTION A high-carbohydrate diet is known to increase insulin secretion and induce obesity. However, whether or not a high-carbohydrate diet affects β-cell mass (BCM) has been little investigated. MATERIALS AND METHODS Both wild-type (WT) mice and adenosine triphosphate-sensitive potassium channel-deficient (Kir6.2KO) mice were fed normal chow or high-starch (ST) diets for 22 weeks. BCM and the numbers of islets were analyzed by immunohistochemistry, and gene expression levels in islets were investigated by quantitative real-time reverse transcription polymerase chain reaction. MIN6-K8 β-cells were stimulated in solution containing various concentrations of glucose combined with nifedipine and glimepiride, and gene expression was analyzed. RESULTS Both WT and Kir6.2KO mice fed ST showed hyperinsulinemia and body weight gain. BCM, the number of islets and the expression levels of cyclinD2 messenger ribonucleic acid were increased in WT mice fed ST compared with those in WT mice fed normal chow. In contrast, no significant difference in BCM, the number of islets or the expression levels of cyclinD2 messenger ribonucleic acid were observed between Kir6.2KO mice fed normal chow and those fed ST. Incubation of MIN6-K8 β-cells in high-glucose media or with glimepiride increased cyclinD2 expression, whereas nifedipine attenuated a high-glucose-induced increase in cyclinD2 expression. CONCLUSIONS These results show that a high-starch diet increases BCM in an adenosine triphosphate-sensitive potassium channel-dependent manner, which is mediated through upregulation of cyclinD2 expression.
Collapse
Affiliation(s)
- Masatoshi Murase
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| | - Yusuke Seino
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| | - Ryuya Maekawa
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| | - Atsushi Iida
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| | - Kaori Hosokawa
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| | - Tomohide Hayami
- Division of Molecular and Metabolic MedicineKobe University Graduate School of MedicineKobeJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKobeJapan
- Division of DiabetesDepartment of Internal MedicineAichi Medical University School of MedicineNagakuteJapan
| | - Shin Tsunekawa
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoji Hamada
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| | - Norihide Yokoi
- Division of Molecular and Metabolic MedicineKobe University Graduate School of MedicineKobeJapan
| | - Susumu Seino
- Division of Molecular and Metabolic MedicineKobe University Graduate School of MedicineKobeJapan
| | - Yoshitaka Hayashi
- Division of Stress Adaptation and ProtectionDepartment of Genetics ResearchInstitute of Environmental MedicineNagoya UniversityNagoyaJapan
| | - Hiroshi Arima
- Departments of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
15
|
Aguayo-Mazzucato C, Bonner-Weir S. Pancreatic β Cell Regeneration as a Possible Therapy for Diabetes. Cell Metab 2018; 27:57-67. [PMID: 28889951 PMCID: PMC5762410 DOI: 10.1016/j.cmet.2017.08.007] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/05/2017] [Accepted: 08/08/2017] [Indexed: 02/08/2023]
Abstract
Diabetes is the result of having inadequate supply of functional insulin-producing β cells. Two possible approaches for replenishing the β cells are: (1) replacement by transplanting cadaveric islets or β cells derived from human embryonic stem cells/induced pluripotent stem cells and (2) induction of endogenous regeneration. This review focuses on endogenous regeneration, which can follow two pathways: enhanced replication of existing β cells and formation of new β cells from cells not expressing insulin, either by conversion from a differentiated cell type (transdifferentiation) or differentiation from progenitors (neogenesis). Exciting progress on both pathways suggest that regeneration may have therapeutic promise.
Collapse
Affiliation(s)
| | - Susan Bonner-Weir
- Joslin Diabetes Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA.
| |
Collapse
|
16
|
Tschen SI, Zeng C, Field L, Dhawan S, Bhushan A, Georgia S. Cyclin D2 is sufficient to drive β cell self-renewal and regeneration. Cell Cycle 2017; 16:2183-2191. [PMID: 28763258 PMCID: PMC5736344 DOI: 10.1080/15384101.2017.1319999] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Diabetes results from an inadequate mass of functional β cells, due to either β cell loss caused by autoimmune destruction (type I diabetes) or β cell failure in response to insulin resistance (type II diabetes). Elucidating the mechanisms that regulate β cell mass may be key to developing new techniques that foster β cell regeneration as a cellular therapy to treat diabetes. While previous studies concluded that cyclin D2 is required for postnatal β cell self-renewal in mice, it is not clear if cyclin D2 is sufficient to drive β cell self-renewal. Using transgenic mice that overexpress cyclin D2 specifically in β cells, we show that cyclin D2 overexpression increases β cell self-renewal post-weaning and results in increased β cell mass. β cells that overexpress cyclin D2 are responsive to glucose stimulation, suggesting they are functionally mature. β cells that overexpress cyclin D2 demonstrate an enhanced regenerative capacity after injury induced by streptozotocin toxicity. To understand if cyclin D2 overexpression is sufficient to drive β cell self-renewal, we generated a novel mouse model where cyclin D2 is only expressed in β cells of cyclin D2−/− mice. Transgenic overexpression of cyclin D2 in cyclin D2−/− β cells was sufficient to restore β cell mass, maintain normoglycaemia, and improve regenerative capacity when compared with cyclin D2−/− littermates. Taken together, our results indicate that cyclin D2 is sufficient to regulate β cell self-renewal and that manipulation of its expression could be used to enhance β cell regeneration.
Collapse
Affiliation(s)
- Shuen-Ing Tschen
- a Department of Medicine , University of California, Los Angeles , Los Angeles , CA
| | - Chun Zeng
- b Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center , University of California San Diego , La Jolla , CA
| | - Loren Field
- c Krannert Institute of Cardiology, and the Riley Heart Research Center, Wells Center for Pediatric Research, and Indiana University School of Medicine , Indianapolis , IN
| | - Sangeeta Dhawan
- a Department of Medicine , University of California, Los Angeles , Los Angeles , CA
| | - Anil Bhushan
- d Diabetes Center , University of California, San Francisco , San Francisco , CA
| | - Senta Georgia
- e Department of Pediatrics, Keck School of Medicine , University of Southern California; Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles , Los Angeles , California
| |
Collapse
|
17
|
Tajima K, Shirakawa J, Okuyama T, Kyohara M, Yamazaki S, Togashi Y, Terauchi Y. Effects of metformin on compensatory pancreatic β-cell hyperplasia in mice fed a high-fat diet. Am J Physiol Endocrinol Metab 2017; 313:E367-E380. [PMID: 28512156 DOI: 10.1152/ajpendo.00447.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/24/2017] [Accepted: 05/15/2017] [Indexed: 01/09/2023]
Abstract
Metformin has been widely used for the treatment of type 2 diabetes. However, the effect of metformin on pancreatic β-cells remains controversial. In this study, we investigated the impacts of treatment with metformin on pancreatic β-cells in a mouse model fed a high-fat diet (HFD), which triggers adaptive β-cell replication. An 8-wk treatment with metformin improved insulin resistance and suppressed the compensatory β-cell hyperplasia induced by HFD-feeding. In contrast, the increment in β-cell mass arising from 60 wk of HFD feeding was similar in mice treated with and those treated without metformin. Interestingly, metformin suppressed β-cell proliferation induced by 1 wk of HFD feeding without any changes in insulin resistance. Metformin directly suppressed glucose-induced β-cell proliferation in islets and INS-1 cells in accordance with a reduction in mammalian target of rapamycin phosphorylation. Taken together, metformin suppressed HFD-induced β-cell proliferation independent of the improvement of insulin resistance, partly via direct actions.
Collapse
Affiliation(s)
- Kazuki Tajima
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Jun Shirakawa
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Shunsuke Yamazaki
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
18
|
Kawamori D. Exploring the molecular mechanisms underlying α- and β-cell dysfunction in diabetes. Diabetol Int 2017; 8:248-256. [PMID: 30603330 PMCID: PMC6224887 DOI: 10.1007/s13340-017-0327-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 06/20/2017] [Indexed: 01/09/2023]
Abstract
Pancreatic islet dysfunction, including impaired insulin secretion in β cells and dysregulated glucagon secretion in α cells, is the chief pathology of diabetes. In β cells, oxidative stress, evoked by chronic hyperglycemia, was found to induce dysfunction of a critical transcription factor, PDX1, caused by its nucleocytoplasmic translocation via interactions with the insulin and JNK signaling pathways and another transcription factor, FOXO1. The significance of α-cell insulin signaling in the physiological and pathological regulation of α-cell biology was demonstrated in α-cell-specific insulin receptor knockout mice, which exhibited dysregulated glucagon secretion. Moreover, a high-glucose load directly induced excessive glucagon secretion in a glucagon-secreting cell line and isolated islets, together with impairment of insulin signaling. These findings indicate that disordered insulin signaling is central to the pathophysiology of islet dysfunction in both α and β cells. On the other hand, certain beneficial effects of GLP-1 on dysfunctional α and β cells indicate that it has therapeutic potential for diabetes patients who exhibit insulin resistance in islets. These studies, involving basic medical research approaches, have-at least in part-clarified the molecular mechanisms underlying α- and β-cell dysfunction in diabetes, and offer important clues that should aid the development of future therapeutic approaches to the disease.
Collapse
Affiliation(s)
- Dan Kawamori
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
19
|
Kawamori D, Shirakawa J, Liew CW, Hu J, Morioka T, Duttaroy A, Burkey B, Kulkarni RN. GLP-1 signalling compensates for impaired insulin signalling in regulating beta cell proliferation in βIRKO mice. Diabetologia 2017; 60:1442-1453. [PMID: 28526921 PMCID: PMC5508991 DOI: 10.1007/s00125-017-4303-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/18/2017] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS We aimed to investigate potential interactions between insulin and glucagon-like peptide (GLP)-1 signalling pathways in the regulation of beta cell-cycle dynamics in vivo, in the context of the therapeutic potential of GLP-1 to modulate impaired beta cell function. METHODS Beta cell-specific insulin receptor knockout (βIRKO) mice, which exhibit beta cell dysfunction and an age-dependent decrease in beta cell mass, were treated with the dipeptidyl peptidase-4 inhibitor vildagliptin. Following this, glucose homeostasis and beta cell proliferation were evaluated and underlying molecular mechanisms were investigated. RESULTS The sustained elevation in circulating GLP-1 levels, caused by treatment of the knockout mice with vildagliptin for 6 weeks, significantly improved glucose tolerance secondary to enhanced insulin secretion and proliferation of beta cells. Treating βIRKO beta cell lines with the GLP-1 analogue, exendin-4, promoted Akt phosphorylation and protein expression of cyclins A, D1 and E two- to threefold, in addition to cyclin D2. Pancreases from the vildagliptin-treated βIRKO mice exhibited increased cyclin D1 expression, while cyclin D2 expression was impaired. CONCLUSIONS/INTERPRETATION Activation of GLP-1 signalling compensates for impaired growth factor (insulin) signalling and enhances expression of cyclins to promote beta cell proliferation. Together, these data indicate the potential of GLP-1-related therapies to enhance beta cell proliferation and promote beneficial outcomes in models with dysfunctional beta cells.
Collapse
Affiliation(s)
- Dan Kawamori
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Medical Education Center, Faculty of Medicine, Osaka University, Osaka, Japan
| | - Jun Shirakawa
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
| | - Chong Wee Liew
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jiang Hu
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
| | - Tomoaki Morioka
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
- Department of Metabolism, Endocrinology and Molecular Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Alokesh Duttaroy
- Cardiovascular & Metabolic Diseases, Novartis Institutes for Biomedical Research, Inc., Cambridge, MA, USA
| | | | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
20
|
Javeed N, Matveyenko AV. β cell self-renewal: Cyclin D2 to the rescue. Cell Cycle 2017; 16:1326-1327. [PMID: 28594265 DOI: 10.1080/15384101.2017.1337988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Naureen Javeed
- a Department of Physiology and Biomedical Engineering , Mayo Clinic School of Medicine, Mayo Clinic, Rochester , MN , USA
| | - Aleksey V Matveyenko
- a Department of Physiology and Biomedical Engineering , Mayo Clinic School of Medicine, Mayo Clinic, Rochester , MN , USA
| |
Collapse
|
21
|
Tajima K, Shirakawa J, Togashi Y, Yamazaki S, Okuyama T, Kyohara M, Konishi H, Terauchi Y. Metabolic recovery of lipodystrophy, liver steatosis, and pancreatic β cell proliferation after the withdrawal of OSI-906. Sci Rep 2017. [PMID: 28646158 PMCID: PMC5482874 DOI: 10.1038/s41598-017-04304-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Growth factor signaling via insulin receptor (IR) and IGF-1 receptor (IGF1R) plays several important roles in the pathogenesis of metabolic syndrome and diabetes. OSI-906 (linsitinib), an anti-tumor drug, is an orally bioavailable dual inhibitor of IR and IGF1R. To investigate the recovery from metabolic changes induced by the acute inhibition of IR and IGF1R in adult mice, mice were treated with OSI-906 or a vehicle for 7 days and the results were analyzed on the last day of injection (Day 7) or after 7 or 21 days of withdrawal (Day 14 or Day 28). On day 7, the visceral white fat mass was significantly reduced in mice treated with OSI-906 accompanied by a reduced expression of leptin and an increased expression of the lipolysis-related genes Lpl and Atgl. Interestingly, the lipoatrophy and the observed changes in gene expression were completely reversed on day 14. Similarly, liver steatosis and β cell proliferation were transiently observed on day 7 but had disappeared by day 14. Taken together, these results suggest that this model for the acute inhibition of systemic IR/IGF1R signaling may be useful for investigating the recovery from metabolic disorders induced by impaired growth factor signaling.
Collapse
Affiliation(s)
- Kazuki Tajima
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Jun Shirakawa
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan.
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Shunsuke Yamazaki
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Hiromi Konishi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan.
| |
Collapse
|
22
|
Wen X, Yang Y. Emerging roles of GLIS3 in neonatal diabetes, type 1 and type 2 diabetes. J Mol Endocrinol 2017; 58:R73-R85. [PMID: 27899417 DOI: 10.1530/jme-16-0232] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 11/25/2016] [Indexed: 12/26/2022]
Abstract
GLI-similar 3 (GLIS3), a member of the Krüppel-like zinc finger protein subfamily, is predominantly expressed in the pancreas, thyroid and kidney. Glis3 mRNA can be initially detected in mouse pancreas at embryonic day 11.5 and is largely restricted to β cells, pancreatic polypeptide-expressing cells, as well as ductal cells at later stage of pancreas development. Mutations in GLIS3 cause a neonatal diabetes syndrome, characterized by neonatal diabetes, congenital hypothyroidism and polycystic kidney. Importantly, genome-wide association studies showed that variations of GLIS3 are strongly associated with both type 1 diabetes (T1D) and type 2 diabetes (T2D) in multiple populations. GLIS3 cooperates with pancreatic and duodenal homeobox 1 (PDX1), v-maf musculoaponeurotic fibrosarcoma oncogene family, protein A (MAFA), as well as neurogenic differentiation 1 (NEUROD1) and potently controls insulin gene transcription. GLIS3 also plays a role in β cell survival and likely in insulin secretion. Any perturbation of these functions may underlie all three forms of diabetes. GLIS3, synergistically with hepatocyte nuclear factor 6 (HNF6) and forkhead box A2 (FOXA2), controls fetal islet differentiation via transactivating neurogenin 3 (NGN3) and impairment of this function leads to neonatal diabetes. In addition, GLIS3 is also required for the compensatory β cell proliferation and mass expansion in response to insulin resistance, which if disrupted may predispose to T2D. The increasing understanding of the mechanisms of GLIS3 in β cell development, survival and function maintenance will provide new insights into disease pathogenesis and potential therapeutic target identification to combat diabetes.
Collapse
Affiliation(s)
- Xianjie Wen
- Division of EndocrinologyDepartment of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
- Department of AnesthesiologyThe First People's Hospital of Foshan & Foshan Hospital of Sun Yat-sen University, Guangdong, China
| | - Yisheng Yang
- Division of EndocrinologyDepartment of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
23
|
Kong Y, Sharma RB, Nwosu BU, Alonso LC. Islet biology, the CDKN2A/B locus and type 2 diabetes risk. Diabetologia 2016; 59:1579-93. [PMID: 27155872 PMCID: PMC4930689 DOI: 10.1007/s00125-016-3967-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes, fuelled by the obesity epidemic, is an escalating worldwide cause of personal hardship and public cost. Diabetes incidence increases with age, and many studies link the classic senescence and ageing protein p16(INK4A) to diabetes pathophysiology via pancreatic islet biology. Genome-wide association studies (GWASs) have unequivocally linked the CDKN2A/B locus, which encodes p16 inhibitor of cyclin-dependent kinase (p16(INK4A)) and three other gene products, p14 alternate reading frame (p14(ARF)), p15(INK4B) and antisense non-coding RNA in the INK4 locus (ANRIL), with human diabetes risk. However, the mechanism by which the CDKN2A/B locus influences diabetes risk remains uncertain. Here, we weigh the evidence that CDKN2A/B polymorphisms impact metabolic health via islet biology vs effects in other tissues. Structured in a bedside-to-bench-to-bedside approach, we begin with a summary of the evidence that the CDKN2A/B locus impacts diabetes risk and a brief review of the basic biology of CDKN2A/B gene products. The main emphasis of this work is an in-depth look at the nuanced roles that CDKN2A/B gene products and related proteins play in the regulation of beta cell mass, proliferation and insulin secretory function, as well as roles in other metabolic tissues. We finish with a synthesis of basic biology and clinical observations, incorporating human physiology data. We conclude that it is likely that the CDKN2A/B locus influences diabetes risk through both islet and non-islet mechanisms.
Collapse
Affiliation(s)
- Yahui Kong
- AS7-2047, Division of Diabetes, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Rohit B Sharma
- AS7-2047, Division of Diabetes, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Benjamin U Nwosu
- Division of Endocrinology, Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA
| | - Laura C Alonso
- AS7-2047, Division of Diabetes, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
24
|
Dirice E, Walpita D, Vetere A, Meier BC, Kahraman S, Hu J, Dančík V, Burns SM, Gilbert TJ, Olson DE, Clemons PA, Kulkarni RN, Wagner BK. Inhibition of DYRK1A Stimulates Human β-Cell Proliferation. Diabetes 2016; 65:1660-71. [PMID: 26953159 PMCID: PMC4878416 DOI: 10.2337/db15-1127] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 02/22/2016] [Indexed: 12/15/2022]
Abstract
Restoring functional β-cell mass is an important therapeutic goal for both type 1 and type 2 diabetes (1). While proliferation of existing β-cells is the primary means of β-cell replacement in rodents (2), it is unclear whether a similar principle applies to humans, as human β-cells are remarkably resistant to stimulation of division (3,4). Here, we show that 5-iodotubercidin (5-IT), an annotated adenosine kinase inhibitor previously reported to increase proliferation in rodent and porcine islets (5), strongly and selectively increases human β-cell proliferation in vitro and in vivo. Remarkably, 5-IT also increased glucose-dependent insulin secretion after prolonged treatment. Kinome profiling revealed 5-IT to be a potent and selective inhibitor of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) and cell division cycle-like kinase families. Induction of β-cell proliferation by either 5-IT or harmine, another natural product DYRK1A inhibitor, was suppressed by coincubation with the calcineurin inhibitor FK506, suggesting involvement of DYRK1A and nuclear factor of activated T cells signaling. Gene expression profiling in whole islets treated with 5-IT revealed induction of proliferation- and cell cycle-related genes, suggesting that true proliferation is induced by 5-IT. Furthermore, 5-IT promotes β-cell proliferation in human islets grafted under the kidney capsule of NOD-scid IL2Rg(null) mice. These results point to inhibition of DYRK1A as a therapeutic strategy to increase human β-cell proliferation.
Collapse
Affiliation(s)
- Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Deepika Walpita
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Amedeo Vetere
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Bennett C Meier
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
| | - Sevim Kahraman
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Vlado Dančík
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Sean M Burns
- Chemical Biology Program, Harvard Medical School, Boston, MA Diabetes Unit, Departments of Medicine and Molecular Biology, Massachusetts General Hospital, Boston, MA
| | - Tamara J Gilbert
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - David E Olson
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | - Paul A Clemons
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Bridget K Wagner
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| |
Collapse
|
25
|
Yang Y, Chan L. Monogenic Diabetes: What It Teaches Us on the Common Forms of Type 1 and Type 2 Diabetes. Endocr Rev 2016; 37:190-222. [PMID: 27035557 PMCID: PMC4890265 DOI: 10.1210/er.2015-1116] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To date, more than 30 genes have been linked to monogenic diabetes. Candidate gene and genome-wide association studies have identified > 50 susceptibility loci for common type 1 diabetes (T1D) and approximately 100 susceptibility loci for type 2 diabetes (T2D). About 1-5% of all cases of diabetes result from single-gene mutations and are called monogenic diabetes. Here, we review the pathophysiological basis of the role of monogenic diabetes genes that have also been found to be associated with common T1D and/or T2D. Variants of approximately one-third of monogenic diabetes genes are associated with T2D, but not T1D. Two of the T2D-associated monogenic diabetes genes-potassium inward-rectifying channel, subfamily J, member 11 (KCNJ11), which controls glucose-stimulated insulin secretion in the β-cell; and peroxisome proliferator-activated receptor γ (PPARG), which impacts multiple tissue targets in relation to inflammation and insulin sensitivity-have been developed as major antidiabetic drug targets. Another monogenic diabetes gene, the preproinsulin gene (INS), is unique in that INS mutations can cause hyperinsulinemia, hyperproinsulinemia, neonatal diabetes mellitus, one type of maturity-onset diabetes of the young (MODY10), and autoantibody-negative T1D. Dominant heterozygous INS mutations are the second most common cause of permanent neonatal diabetes. Moreover, INS gene variants are strongly associated with common T1D (type 1a), but inconsistently with T2D. Variants of the monogenic diabetes gene Gli-similar 3 (GLIS3) are associated with both T1D and T2D. GLIS3 is a key transcription factor in insulin production and β-cell differentiation during embryonic development, which perturbation forms the basis of monogenic diabetes as well as its association with T1D. GLIS3 is also required for compensatory β-cell proliferation in adults; impairment of this function predisposes to T2D. Thus, monogenic forms of diabetes are invaluable "human models" that have contributed to our understanding of the pathophysiological basis of common T1D and T2D.
Collapse
Affiliation(s)
- Yisheng Yang
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Lawrence Chan
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
26
|
Ueberberg S, Tannapfel A, Schenker P, Viebahn R, Uhl W, Schneider S, Meier JJ. Differential expression of cell-cycle regulators in human beta-cells derived from insulinoma tissue. Metabolism 2016; 65:736-746. [PMID: 27085780 DOI: 10.1016/j.metabol.2016.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/09/2016] [Accepted: 02/17/2016] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The low frequency of beta-cell replication in the adult human pancreas limits beta-cell regeneration. A better understanding of the regulation of human beta-cell proliferation is crucial to develop therapeutic strategies aiming to enhance beta-cell mass. METHODS To identify factors that control beta-cell proliferation, cell-cycle regulation was examined in human insulinomas as a model of increased beta-cell proliferation (n=11) and healthy pancreatic tissue from patients with benign pancreatic tumors (n=9). Tissue sections were co-stained for insulin and cell-cycle proteins. Transcript levels of selected cell-cycle factors in beta-cells were determined by qRT-PCR after performing laser-capture microdissection. RESULTS The frequency of beta-cell replication was 3.74±0.92% in the insulinomas and 0.11±0.04% in controls (p=0.0016). p21 expression was higher in insulinomas (p=0.0058), and Rb expression was higher by trend (p=0.085), whereas p16 (p<0.0001), Cyclin C (p<0.0001), and p57 (p=0.018) expression levels were lower. The abundance of Cyclin D3 (p=0.62) and p27 (p=0.68) was not different between the groups. The reduced expression of p16 (p<0.0001) and p57 (p=0.012) in insulinomas and the unchanged expression of Cyclin D3 (p=0.77) and p27 (p=0.55) were confirmed using qRT-PCR. CONCLUSIONS The expression of certain cell-cycle factors in beta-cells derived from insulinomas and healthy adults differs markedly. Targeting such differentially regulated cell-cycle proteins may evolve as a future strategy to enhance beta-cell regeneration.
Collapse
Affiliation(s)
- Sandra Ueberberg
- Diabetes Division, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, Bochum 44791, Germany
| | - Andrea Tannapfel
- Department of Pathology, Ruhr-University Bochum, Bürkle de la Camp-Platz 1, Bochum 44789, Germany
| | - Peter Schenker
- Department of Surgery, Knappschaftskrankenhaus Bochum, Ruhr-University Bochum, In der Schornau 23-25, Bochum 44892, Germany
| | - Richard Viebahn
- Department of Surgery, Knappschaftskrankenhaus Bochum, Ruhr-University Bochum, In der Schornau 23-25, Bochum 44892, Germany
| | - Waldemar Uhl
- Department of Surgery, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, Bochum 44791, Germany
| | - Stephan Schneider
- Department of Medicine II, St. Vinzenz Hospital, Merheimer Str. 221-223, Cologne 50733, Germany
| | - Juris J Meier
- Diabetes Division, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, Bochum 44791, Germany.
| |
Collapse
|
27
|
Lakshmipathi J, Alvarez-Perez JC, Rosselot C, Casinelli GP, Stamateris RE, Rausell-Palamos F, O'Donnell CP, Vasavada RC, Scott DK, Alonso LC, Garcia-Ocaña A. PKCζ Is Essential for Pancreatic β-Cell Replication During Insulin Resistance by Regulating mTOR and Cyclin-D2. Diabetes 2016; 65:1283-96. [PMID: 26868297 PMCID: PMC4839210 DOI: 10.2337/db15-1398] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/06/2016] [Indexed: 12/23/2022]
Abstract
Adaptive β-cell replication occurs in response to increased metabolic demand during insulin resistance. The intracellular mediators of this compensatory response are poorly defined and their identification could provide significant targets for β-cell regeneration therapies. Here we show that glucose and insulin in vitro and insulin resistance in vivo activate protein kinase C ζ (PKCζ) in pancreatic islets and β-cells. PKCζ is required for glucose- and glucokinase activator-induced proliferation of rodent and human β-cells in vitro. Furthermore, either kinase-dead PKCζ expression (KD-PKCζ) or disruption of PKCζ in mouse β-cells blocks compensatory β-cell replication when acute hyperglycemia/hyperinsulinemia is induced. Importantly, KD-PKCζ inhibits insulin resistance-mediated mammalian target of rapamycin (mTOR) activation and cyclin-D2 upregulation independent of Akt activation. In summary, PKCζ activation is key for early compensatory β-cell replication in insulin resistance by regulating the downstream signals mTOR and cyclin-D2. This suggests that alterations in PKCζ expression or activity might contribute to inadequate β-cell mass expansion and β-cell failure leading to type 2 diabetes.
Collapse
Affiliation(s)
- Jayalakshmi Lakshmipathi
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Juan Carlos Alvarez-Perez
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Carolina Rosselot
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gabriella P Casinelli
- Division of Pediatric Hematology/Oncology and Blood and Marrow Transplantation, Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Rachel E Stamateris
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Francisco Rausell-Palamos
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Christopher P O'Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Laura C Alonso
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Adolfo Garcia-Ocaña
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
28
|
Stamateris RE, Sharma RB, Kong Y, Ebrahimpour P, Panday D, Ranganath P, Zou B, Levitt H, Parambil NA, O'Donnell CP, García-Ocaña A, Alonso LC. Glucose Induces Mouse β-Cell Proliferation via IRS2, MTOR, and Cyclin D2 but Not the Insulin Receptor. Diabetes 2016; 65:981-95. [PMID: 26740601 PMCID: PMC5314707 DOI: 10.2337/db15-0529] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022]
Abstract
An important goal in diabetes research is to understand the processes that trigger endogenous β-cell proliferation. Hyperglycemia induces β-cell replication, but the mechanism remains debated. A prime candidate is insulin, which acts locally through the insulin receptor. Having previously developed an in vivo mouse hyperglycemia model, we tested whether glucose induces β-cell proliferation through insulin signaling. By using mice lacking insulin signaling intermediate insulin receptor substrate 2 (IRS2), we confirmed that hyperglycemia-induced β-cell proliferation requires IRS2 both in vivo and ex vivo. Of note, insulin receptor activation was not required for glucose-induced proliferation, and insulin itself was not sufficient to drive replication. Glucose and insulin caused similar acute signaling in mouse islets, but chronic signaling differed markedly, with mammalian target of rapamycin (MTOR) and extracellular signal-related kinase (ERK) activation by glucose and AKT activation by insulin. MTOR but not ERK activation was required for glucose-induced proliferation. Cyclin D2 was necessary for glucose-induced β-cell proliferation. Cyclin D2 expression was reduced when either IRS2 or MTOR signaling was lost, and restoring cyclin D2 expression rescued the proliferation defect. Human islets shared many of these regulatory pathways. Taken together, these results support a model in which IRS2, MTOR, and cyclin D2, but not the insulin receptor, mediate glucose-induced proliferation.
Collapse
Affiliation(s)
- Rachel E Stamateris
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Rohit B Sharma
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Yahui Kong
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Pantea Ebrahimpour
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Deepika Panday
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Pavana Ranganath
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Baobo Zou
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Helena Levitt
- Division of Endocrinology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Christopher P O'Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Disease, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Laura C Alonso
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
29
|
Kim J, Lee KJ, Kim JS, Rho JG, Shin JJ, Song WK, Lee EK, Egan JM, Kim W. Cannabinoids Regulate Bcl-2 and Cyclin D2 Expression in Pancreatic β Cells. PLoS One 2016; 11:e0150981. [PMID: 26967640 PMCID: PMC4788443 DOI: 10.1371/journal.pone.0150981] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/22/2016] [Indexed: 02/07/2023] Open
Abstract
Recent reports have shown that cannabinoid 1 receptors (CB1Rs) are expressed in pancreatic β cells, where they induce cell death and cell cycle arrest by directly inhibiting insulin receptor activation. Here, we report that CB1Rs regulate the expression of the anti-apoptotic protein Bcl-2 and cell cycle regulator cyclin D2 in pancreatic β cells. Treatment of MIN6 and βTC6 cells with a synthetic CB1R agonist, WIN55,212–2, led to a decrease in the expression of Bcl-2 and cyclin D2, in turn inducing cell cycle arrest in G0/G1 phase and caspase-3-dependent apoptosis. Additionally, genetic deletion and pharmacological blockade of CB1Rs after injury in mice led to increased levels of Bcl-2 and cyclin D2 in pancreatic β cells. These findings provide evidence for the involvement of Bcl-2 and cyclin D2 mediated by CB1Rs in the regulation of β-cell survival and growth, and will serve as a basis for developing new therapeutic interventions to enhance β-cell function and growth in diabetes.
Collapse
Affiliation(s)
- Jihye Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Kyung Jin Lee
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Korea
| | - Jung Seok Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Jun Gi Rho
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Jung Jae Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Woo Keun Song
- Department of Life Science, Bio Imaging and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, United States of America
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| |
Collapse
|
30
|
Zhang T, Kim DH, Xiao X, Lee S, Gong Z, Muzumdar R, Calabuig-Navarro V, Yamauchi J, Harashima H, Wang R, Bottino R, Alvarez-Perez JC, Garcia-Ocaña A, Gittes G, Dong HH. FoxO1 Plays an Important Role in Regulating β-Cell Compensation for Insulin Resistance in Male Mice. Endocrinology 2016; 157:1055-70. [PMID: 26727107 PMCID: PMC4769368 DOI: 10.1210/en.2015-1852] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
β-Cell compensation is an essential mechanism by which β-cells increase insulin secretion for overcoming insulin resistance to maintain euglycemia in obesity. Failure of β-cells to compensate for insulin resistance contributes to insulin insufficiency and overt diabetes. To understand the mechanism of β-cell compensation, we characterized the role of forkhead box O1 (FoxO1) in β-cell compensation in mice under physiological and pathological conditions. FoxO1 is a key transcription factor that serves as a nutrient sensor for integrating insulin signaling to cell metabolism, growth, and proliferation. We showed that FoxO1 improved β-cell compensation via 3 distinct mechanisms by increasing β-cell mass, enhancing β-cell glucose sensing, and augmenting β-cell antioxidative function. These effects accounted for increased glucose-stimulated insulin secretion and enhanced glucose tolerance in β-cell-specific FoxO1-transgenic mice. When fed a high-fat diet, β-cell-specific FoxO1-transgenic mice were protected from developing fat-induced glucose disorder. This effect was attributable to increased β-cell mass and function. Furthermore, we showed that FoxO1 activity was up-regulated in islets, correlating with the induction of physiological β-cell compensation in high-fat-induced obese C57BL/6J mice. These data characterize FoxO1 as a pivotal factor for orchestrating physiological adaptation of β-cell mass and function to overnutrition and obesity.
Collapse
Affiliation(s)
- Ting Zhang
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Dae Hyun Kim
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Xiangwei Xiao
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Sojin Lee
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Zhenwei Gong
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Radhika Muzumdar
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Virtu Calabuig-Navarro
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Jun Yamauchi
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Hideyoshi Harashima
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Rennian Wang
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Rita Bottino
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Juan Carlos Alvarez-Perez
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Adolfo Garcia-Ocaña
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - George Gittes
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - H Henry Dong
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| |
Collapse
|
31
|
Chen H, Kleinberger JW, Takane KK, Salim F, Fiaschi-Taesch N, Pappas K, Parsons R, Jiang J, Zhang Y, Liu H, Wang P, Bender AS, Frank SJ, Stewart AF. Augmented Stat5 Signaling Bypasses Multiple Impediments to Lactogen-Mediated Proliferation in Human β-Cells. Diabetes 2015; 64:3784-97. [PMID: 26159175 PMCID: PMC4613973 DOI: 10.2337/db15-0083] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/30/2015] [Indexed: 12/20/2022]
Abstract
Pregnancy in rodents is associated with a two- to threefold increase in β-cell mass, which is attributable to large increases in β-cell proliferation, complimented by increases in β-cell size, survival, and function and mediated mainly by the lactogenic hormones prolactin (PRL) and placental lactogens. In humans, however, β-cell mass does not increase as dramatically during pregnancy, and PRL fails to activate proliferation in human islets in vitro. To determine why, we explored the human PRL-prolactin receptor (hPRLR)-Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5)-cyclin-cdk signaling cascade in human β-cells. Surprisingly, adult human β-cells express little or no PRLR. As expected, restoration of the hPRLR in human β-cells rescued JAK2-STAT5 signaling in response to PRL. However, rescuing hPRLR-STAT5 signaling nevertheless failed to confer proliferative ability on adult human β-cells in response to PRL. Surprisingly, mouse (but not human) Stat5a overexpression led to upregulation of cyclins D1-3 and cdk4, as well as their nuclear translocation, all of which are associated with β-cell cycle entry. Collectively, the findings show that human β-cells fail to proliferate in response to PRL for multiple reasons, one of which is a paucity of functional PRL receptors, and that murine Stat5 overexpression is able to bypass these impediments.
Collapse
Affiliation(s)
- Hainan Chen
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Karen K Takane
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Fatimah Salim
- Duquesne University School of Nursing, Pittsburgh, PA
| | - Nathalie Fiaschi-Taesch
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kyrie Pappas
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ramon Parsons
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jing Jiang
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| | - Yue Zhang
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| | - Hongtao Liu
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Peng Wang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Aaron S Bender
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stuart J Frank
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL Endocrinology Section, Birmingham VA Medical Center, Birmingham, AL
| | - Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
32
|
Alonso-Magdalena P, García-Arévalo M, Quesada I, Nadal Á. Bisphenol-A treatment during pregnancy in mice: a new window of susceptibility for the development of diabetes in mothers later in life. Endocrinology 2015; 156:1659-70. [PMID: 25830705 DOI: 10.1210/en.2014-1952] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Evidence now exists supporting the hypothesis that endocrine-disrupting chemicals (EDCs) can harmfully impact glucose metabolism. Thus, EDCs are beginning to be considered important contributors to the increased incidence of diabetes, obesity, or both. The possible effect of exposure to EDCs during pregnancy on glucose homeostasis in mothers later in life is presently unknown. Here we show that several months after delivery, mothers treated with the widespread EDC bisphenol-A (BPA) during gestation, at environmentally relevant doses, exhibit profound glucose intolerance and altered insulin sensitivity as well as increased body weight. These mice presented a decreased insulin secretion both in vivo and in vitro together with reduced pancreatic β-cell mass. The proliferation capacity was decreased in association with a diminished expression of the cell cycle activators: cyclin D2 and cyclin-dependent kinase-4. In addition, the rate of β-cells apoptosis was increased as well as the expression of the cell cycle inhibitors p16 and p53. Conversely, no effects on glucose metabolism or insulin sensitivity were observed when female nonpregnant mice were treated with BPA at the same doses. Taken together, these findings reveal that BPA exposure during gestation has harmful long-term implications in glucose metabolism for the mother. This finding highlights a new window of susceptibility for EDC exposure that may be important for the development of type 2 diabetes.
Collapse
Affiliation(s)
- Paloma Alonso-Magdalena
- Departamento de Biología Aplicada (P.A.-M., I.Q.), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (P.A.-M., M.G.-A., I.Q., A.N.), Universidad Miguel Hernández de Elche, Elche 03202, Alicante, Spain
| | | | | | | |
Collapse
|
33
|
Jacovetti C, Jimenez V, Ayuso E, Laybutt R, Peyot ML, Prentki M, Bosch F, Regazzi R. Contribution of Intronic miR-338-3p and Its Hosting Gene AATK to Compensatory β-Cell Mass Expansion. Mol Endocrinol 2015; 29:693-702. [PMID: 25751313 DOI: 10.1210/me.2014-1299] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The elucidation of the mechanisms directing β-cell mass regeneration and maintenance is of interest, because the deficit of β-cell mass contributes to diabetes onset and progression. We previously found that the level of the microRNA (miRNA) miR-338-3p is decreased in pancreatic islets from rodent models displaying insulin resistance and compensatory β-cell mass expansion, including pregnant rats, diet-induced obese mice, and db/db mice. Transfection of rat islet cells with oligonucleotides that specifically block miR-338-3p activity increased the fraction of proliferating β-cells in vitro and promoted survival under proapoptotic conditions without affecting the capacity of β-cells to release insulin in response to glucose. Here, we evaluated the role of miR-338-3p in vivo by injecting mice with an adeno-associated viral vector permitting specific sequestration of this miRNA in β-cells. We found that the adeno-associated viral construct increased the fraction of proliferating β-cells confirming the data obtained in vitro. miR-338-3p is generated from an intron of the gene coding for apoptosis-associated tyrosine kinase (AATK). Similarly to miR-338-3p, we found that AATK is down-regulated in rat and human islets and INS832/13 β-cells in the presence of the cAMP-raising agents exendin-4, estradiol, and a G-protein-coupled Receptor 30 agonist. Moreover, AATK expression is reduced in islets of insulin resistant animal models and selective silencing of AATK in INS832/13 cells by RNA interference promoted β-cell proliferation. The results point to a coordinated reduction of miR-338-3p and AATK under insulin resistance conditions and provide evidence for a cooperative action of the miRNA and its hosting gene in compensatory β-cell mass expansion.
Collapse
Affiliation(s)
- Cécile Jacovetti
- Department of Fundamental Neurosciences (C.J., R.R.), University of Lausanne, 1005 Lausanne, Switzerland; Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology (V.J., E.A., F.B.), School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain; Diabetes and Obesity Research Program (R.L.), Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010 New South Wales, Australia; and Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal (M.-L.P., M.P.), and Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, Quebec, H2X 0A9 Canada
| | | | | | | | | | | | | | | |
Collapse
|
34
|
An integrated cell purification and genomics strategy reveals multiple regulators of pancreas development. PLoS Genet 2014; 10:e1004645. [PMID: 25330008 PMCID: PMC4199491 DOI: 10.1371/journal.pgen.1004645] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 08/02/2014] [Indexed: 12/15/2022] Open
Abstract
The regulatory logic underlying global transcriptional programs controlling development of visceral organs like the pancreas remains undiscovered. Here, we profiled gene expression in 12 purified populations of fetal and adult pancreatic epithelial cells representing crucial progenitor cell subsets, and their endocrine or exocrine progeny. Using probabilistic models to decode the general programs organizing gene expression, we identified co-expressed gene sets in cell subsets that revealed patterns and processes governing progenitor cell development, lineage specification, and endocrine cell maturation. Purification of Neurog3 mutant cells and module network analysis linked established regulators such as Neurog3 to unrecognized gene targets and roles in pancreas development. Iterative module network analysis nominated and prioritized transcriptional regulators, including diabetes risk genes. Functional validation of a subset of candidate regulators with corresponding mutant mice revealed that the transcription factors Etv1, Prdm16, Runx1t1 and Bcl11a are essential for pancreas development. Our integrated approach provides a unique framework for identifying regulatory genes and functional gene sets underlying pancreas development and associated diseases such as diabetes mellitus. Discovery of specific pancreas developmental regulators has accelerated in recent years. In contrast, the global regulatory programs controlling pancreas development are poorly understood compared to other organs or tissues like heart or blood. Decoding this regulatory logic may accelerate development of replacement organs from renewable sources like stem cells, but this goal requires identification of regulators and assessment of their functions on a global scale. To address this important challenge for pancreas biology, we combined purification of normal and mutant cells with genome-scale methods to generate and analyze expression profiles from developing pancreas cells. Our work revealed regulatory gene sets governing development of pancreas progenitor cells and their progeny. Our integrative approach nominated multiple pancreas developmental regulators, including suspected risk genes for human diabetes, which we validated by phenotyping mutant mice on a scale not previously reported. Selection of these candidate regulators was unbiased; thus it is remarkable that all were essential for pancreatic islet development. Thus, our studies provide a new heuristic resource for identifying genetic functions underlying pancreas development and diseases like diabetes mellitus.
Collapse
|
35
|
MafA is required for postnatal proliferation of pancreatic β-cells. PLoS One 2014; 9:e104184. [PMID: 25126749 PMCID: PMC4134197 DOI: 10.1371/journal.pone.0104184] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 06/16/2014] [Indexed: 12/05/2022] Open
Abstract
The postnatal proliferation and maturation of insulin-secreting pancreatic β-cells are critical for glucose metabolism and disease development in adults. Elucidation of the molecular mechanisms underlying these events will be beneficial to direct the differentiation of stem cells into functional β-cells. Maturation of β-cells is accompanied by increased expression of MafA, an insulin gene transcription factor. Transcriptome analysis of MafA knockout islets revealed MafA is required for the expression of several molecules critical for β-cell function, including Glut2, ZnT8, Granuphilin, Vdr, Pcsk1 and Urocortin 3, as well as Prolactin receptor (Prlr) and its downstream target Cyclin D2 (Ccnd2). Inhibition of MafA expression in mouse islets or β-cell lines resulted in reduced expression of Prlr and Ccnd2, and MafA transactivated the Prlr promoter. Stimulation of β-cells by prolactin resulted in the phosphorylation and translocation of Stat5B and an increased nuclear pool of Ccnd2 via Prlr and Jak2. Consistent with these results, the loss of MafA resulted in impaired proliferation of β-cells at 4 weeks of age. These results suggest that MafA regulates the postnatal proliferation of β-cells via prolactin signaling.
Collapse
|
36
|
Cyclin D3 promotes pancreatic β-cell fitness and viability in a cell cycle-independent manner and is targeted in autoimmune diabetes. Proc Natl Acad Sci U S A 2014; 111:E3405-14. [PMID: 25092329 DOI: 10.1073/pnas.1323236111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is an autoimmune condition caused by the lymphocyte-mediated destruction of the insulin-producing β cells in pancreatic islets. We aimed to identify final molecular entities targeted by the autoimmune assault on pancreatic β cells that are causally related to β cell viability. Here, we show that cyclin D3 is targeted by the autoimmune attack on pancreatic β cells in vivo. Cyclin D3 is down-regulated in a dose-dependent manner in β cells by leukocyte infiltration into the islets of the nonobese diabetic (NOD) type 1 diabetes-prone mouse model. Furthermore, we established a direct in vivo causal link between cyclin D3 expression levels and β-cell fitness and viability in the NOD mice. We found that changes in cyclin D3 expression levels in vivo altered the β-cell apoptosis rates, β-cell area homeostasis, and β-cell sensitivity to glucose without affecting β-cell proliferation in the NOD mice. Cyclin D3-deficient NOD mice exhibited exacerbated diabetes and impaired glucose responsiveness; conversely, transgenic NOD mice overexpressing cyclin D3 in β cells exhibited mild diabetes and improved glucose responsiveness. Overexpression of cyclin D3 in β cells of cyclin D3-deficient mice rescued them from the exacerbated diabetes observed in transgene-negative littermates. Moreover, cyclin D3 overexpression protected the NOD-derived insulinoma NIT-1 cell line from cytokine-induced apoptosis. Here, for the first time to our knowledge, cyclin D3 is identified as a key molecule targeted by autoimmunity that plays a nonredundant, protective, and cell cycle-independent role in β cells against inflammation-induced apoptosis and confers metabolic fitness to these cells.
Collapse
|
37
|
Thomsen SK, Gloyn AL. The pancreatic β cell: recent insights from human genetics. Trends Endocrinol Metab 2014; 25:425-34. [PMID: 24986330 PMCID: PMC4229643 DOI: 10.1016/j.tem.2014.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus is a metabolic disease characterised by relative or absolute pancreatic β cell dysfunction. Genetic variants implicated in disease risk can be identified by studying affected individuals. To understand the mechanisms driving genetic associations, variants must be translated through causative transcripts to biological insights. Studies into the genetic basis of Mendelian forms of diabetes have successfully identified genes involved in both β cell function and pancreatic development. For type 2 diabetes (T2D), genome-wide association studies (GWASs) are uncovering an ever-increasing number of susceptibility variants that exert their effect through β cell dysfunction, but translation to mechanistic understanding has in most cases been slow. Improved annotations of the islet genome and advances in whole-genome and -exome sequencing (WHS and WES) have facilitated recent progress.
Collapse
Affiliation(s)
- Soren K Thomsen
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Headington, OX3 7LE, UK
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Headington, OX3 7LE, UK; Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, Churchill Hospital, Headington, OX3 7LE, UK.
| |
Collapse
|
38
|
Togashi Y, Shirakawa J, Orime K, Kaji M, Sakamoto E, Tajima K, Inoue H, Nakamura A, Tochino Y, Goshima Y, Shimomura I, Terauchi Y. β-Cell proliferation after a partial pancreatectomy is independent of IRS-2 in mice. Endocrinology 2014; 155:1643-52. [PMID: 24517226 DOI: 10.1210/en.2013-1796] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The glucokinase-induced up-regulation of insulin receptor substrate 2 (IRS-2) plays an important role in β-cell adaptive proliferation in response to high-fat diet-induced insulin resistance. This study aimed to investigate the role of IRS-2 in the proliferation of β-cells after a 60% partial pancreatectomy. IRS-2-deficient (IRS-2(-/-)) mice or wild-type mice were subjected to a pancreatectomy (60% partial pancreatectomy) or a sham operation (Sham). The β-cell proliferation and gene expression profiles of the islets were then assessed. Gene expression in islets from pancreatectomized and Sham C57BL/6J male mice was analyzed using a cDNA microarray analysis. To compare with β-cell proliferation induced by a high-fat diet, Gck(+/-) mice subjected to a pancreatectomy were also analyzed. The IRS-2(-/-) mice exhibited β-cell expansion and a significant increase in β-cell proliferation after the pancreatectomy, compared with the Sham group. Although glucose-stimulated insulin secretion from islets was not impaired, IRS-2(-/-) mice manifested severe hyperglycemia after the pancreatectomy. The expression levels of Aurora kinase B, Cyclin A, and Cyclin B1 in the pancreatectomized islets were also enhanced in the IRS-2(-/-) mice. A gene set enrichment analysis suggested an association between the genes that were up-regulated in the pancreatectomized islets and those involved in M phase progression in the cell cycle. β-Cell proliferation after a pancreatectomy was observed even in the Gck(+/-) mice. In conclusion, IRS-2 was not required for β-cell proliferation but might be needed for functional β-cell mass, after a pancreatectomy. A partial pancreatectomy in mice may be an attractive model for the development of new strategy for exploring the unique nature of β-cell proliferation.
Collapse
Affiliation(s)
- Yu Togashi
- Department of Endocrinology and Metabolism (Y.Tog., J.S., K.O., M.K., E.S., K.T., H.I., A.N., Y.Te.) and Molecular Pharmacology and Neurobiology (Y.G., J.S.), Graduate School of Medicine Yokohama-City University, Yokohama 236-0004, Japan; and Department of Metabolic Medicine (Y.Toc., I.S.), Graduate School of Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Tsai MJ, Yang-Yen HF, Chiang MK, Wang MJ, Wu SS, Chen SH. TCTP is essential for β-cell proliferation and mass expansion during development and β-cell adaptation in response to insulin resistance. Endocrinology 2014; 155:392-404. [PMID: 24248465 DOI: 10.1210/en.2013-1663] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The perinatal period is critical for β-cell mass establishment, which is characterized by a transient burst in proliferation to increase β-cell mass in response to the need for glucose homeostasis throughout life. In adulthood, the ability of β-cells to grow, proliferate, and expand their mass is also characteristic of pathological states of insulin resistance. Translationally controlled tumor-associated protein (TCTP), an evolutionarily highly conserved protein that is implicated in cell growth and proliferation, has been identified as a novel glucose-regulated survival-supporting protein in pancreatic β-cells. In this study, the enhanced β-cell proliferation detected both during the perinatal developmental period and in insulin-resistant states in high-fat diet-fed mice was found to parallel the expression of TCTP in pancreatic β-cells. Specific knockout of TCTP in β-cells led to increased expression of total and nuclear Forkhead box protein O1 and tumor suppressor protein 53, and decreased expression of p70S6 kinase phosphorylation and cyclin D2 and cyclin-dependent kinase 2. This resulted in decreased β-cell proliferation and growth, reduced β-cell mass, and insulin secretion. Together, these effects led to hyperglycemia. These observations suggest that TCTP is essential for β-cell mass expansion during development and β-cell adaptation in response to insulin resistance.
Collapse
Affiliation(s)
- Ming-Jen Tsai
- PhD Program in Pharmacology and Toxicology (M.J.-T., S.-H.C.) and Department of Pharmacology (S.-S.W., S.-H.C.), School of Medicine, Tzu Chi University, Hualien 970, Taiwan; Departments of Emergency Medicine (M.J.-T.) and Medical Research (M.-J.W.), Neuro-Medical Scientific Center, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan; Institutes of Molecular Biology (H.-F.Y.-Y.), Academia Sinica, Taipei 115, Taiwan; and Department of Life Science and Institute of Molecular Biology (M.-K.C.), National Chung-Cheng University, Chia-Yi 621, Taiwan
| | | | | | | | | | | |
Collapse
|
40
|
Taneera J, Fadista J, Ahlqvist E, Zhang M, Wierup N, Renström E, Groop L. Expression profiling of cell cycle genes in human pancreatic islets with and without type 2 diabetes. Mol Cell Endocrinol 2013; 375:35-42. [PMID: 23707792 DOI: 10.1016/j.mce.2013.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/30/2013] [Accepted: 05/01/2013] [Indexed: 02/07/2023]
Abstract
Microarray gene expression data were used to analyze the expression pattern of cyclin, cyclin-dependent kinase (CDKs) and cyclin-dependent kinase inhibitor (CDKIs) genes from human pancreatic islets with and without type 2 diabetes (T2D). Of the cyclin genes, CCNI was the most expressed. Data obtained from microarray and qRT-PCR showed higher expression of CCND1 in diabetic islets. Among the CDKs, CDK4, CDK8 and CDK9 were highly expressed, while CDK1 was expressed at low level. High expression of CDK18 was observed in diabetic islets. Of the CDKIs, CDKN1A expression was higher in diabetic islets in both microarray and qRT-PCR. Expression of CDKN1A, CDKN2A, CCNI2, CDK3 and CDK16 was correlated with age. Finally, eight SNPs in these genes were associated with T2D in the DIAGRAM database. Our data provide a comprehensive expression pattern of cell cycle genes in human islets. More human studies are required to confirm and reproduce animal studies.
Collapse
Affiliation(s)
- Jalal Taneera
- Lund University Diabetes Center, Department of Clinical Sciences, Diabetes & Endocrinology, Skåne University Hospital, Lund University, Malmö 20502, Sweden.
| | | | | | | | | | | | | |
Collapse
|
41
|
Stamateris RE, Sharma RB, Hollern DA, Alonso LC. Adaptive β-cell proliferation increases early in high-fat feeding in mice, concurrent with metabolic changes, with induction of islet cyclin D2 expression. Am J Physiol Endocrinol Metab 2013; 305:E149-59. [PMID: 23673159 PMCID: PMC3725565 DOI: 10.1152/ajpendo.00040.2013] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 05/10/2013] [Indexed: 01/09/2023]
Abstract
Type 2 diabetes (T2D) is caused by relative insulin deficiency, due in part to reduced β-cell mass (11, 62). Therapies aimed at expanding β-cell mass may be useful to treat T2D (14). Although feeding rodents a high-fat diet (HFD) for an extended period (3-6 mo) increases β-cell mass by inducing β-cell proliferation (16, 20, 53, 54), evidence suggests that adult human β-cells may not meaningfully proliferate in response to obesity. The timing and identity of the earliest initiators of the rodent compensatory growth response, possible therapeutic targets to drive proliferation in refractory human β-cells, are not known. To develop a model to identify early drivers of β-cell proliferation, we studied mice during the first week of HFD exposure, determining the onset of proliferation in the context of diet-related physiological changes. Within the first week of HFD, mice consumed more kilocalories, gained weight and fat mass, and developed hyperglycemia, hyperinsulinemia, and glucose intolerance due to impaired insulin secretion. The β-cell proliferative response also began within the first week of HFD feeding. Intriguingly, β-cell proliferation increased before insulin resistance was detected. Cyclin D2 protein expression was increased in islets by day 7, suggesting it may be an early effector driving compensatory β-cell proliferation in mice. This study defines the time frame and physiology to identify novel upstream regulatory signals driving mouse β-cell mass expansion, in order to explore their efficacy, or reasons for inefficacy, in initiating human β-cell proliferation.
Collapse
Affiliation(s)
- Rachel E Stamateris
- Division of Diabetes, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | |
Collapse
|
42
|
Suzuki T, Dai P, Hatakeyama T, Harada Y, Tanaka H, Yoshimura N, Takamatsu T. TGF-β Signaling Regulates Pancreatic β-Cell Proliferation through Control of Cell Cycle Regulator p27 Expression. Acta Histochem Cytochem 2013; 46:51-8. [PMID: 23720603 PMCID: PMC3661777 DOI: 10.1267/ahc.12035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 01/31/2013] [Indexed: 02/06/2023] Open
Abstract
Proliferation of pancreatic β-cells is an important mechanism underlying β-cell mass adaptation to metabolic demands. Increasing β-cell mass by regeneration may ameliorate or correct both type 1 and type 2 diabetes, which both result from inadequate production of insulin by β-cells of the pancreatic islet. Transforming growth factor β (TGF-β) signaling is essential for fetal development and growth of pancreatic islets. In this study, we exposed HIT-T15, a clonal pancreatic β-cell line, to TGF-β signaling. We found that inhibition of TGF-β signaling promotes proliferation of the cells significantly, while TGF-β signaling stimulation inhibits proliferation of the cells remarkably. We confirmed that this proliferative regulation by TGF-β signaling is due to the changed expression of the cell cycle regulator p27. Furthermore, we demonstrated that there is no observed effect on transcriptional activity of p27 by TGF-β signaling. Our data show that TGF-β signaling mediates the cell-cycle progression of pancreatic β-cells by regulating the nuclear localization of CDK inhibitor, p27. Inhibition of TGF-β signaling reduces the nuclear accumulation of p27, and as a result this inhibition promotes proliferation of β-cells.
Collapse
Affiliation(s)
- Tomoyuki Suzuki
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
- Department of Transplantation and Regenerative Surgery, Kyoto Prefectural University of Medicine
| | - Ping Dai
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| | - Tomoya Hatakeyama
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
- Division of Digestive Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| | - Norio Yoshimura
- Department of Transplantation and Regenerative Surgery, Kyoto Prefectural University of Medicine
| | - Tetsuro Takamatsu
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| |
Collapse
|
43
|
López-Acosta JF, Moreno-Amador JL, Jiménez-Palomares M, Díaz-Marrero AR, Cueto M, Perdomo G, Cózar-Castellano I. Epoxypukalide induces proliferation and protects against cytokine-mediated apoptosis in primary cultures of pancreatic β-cells. PLoS One 2013; 8:e52862. [PMID: 23300997 PMCID: PMC3534672 DOI: 10.1371/journal.pone.0052862] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 11/23/2012] [Indexed: 01/09/2023] Open
Abstract
There is an urgency to find new treatments for the devastating epidemic of diabetes. Pancreatic β-cells viability and function are impaired in the two most common forms of diabetes, type 1 and type 2. Regeneration of pancreatic β-cells has been proposed as a potential therapy for diabetes. In a preliminary study, we screened a collection of marine products for β-cell proliferation. One unique compound (epoxypukalide) showed capability to induce β-cell replication in the cell line INS1 832/13 and in primary rat cell cultures. Epoxypukalide was used to study β-cell proliferation by [3H]thymidine incorporation and BrdU incorporation followed by BrdU/insulin staining in primary cultures of rat islets. AKT and ERK1/2 signalling pathways were analyzed. Cell cycle activators, cyclin D2 and cyclin E, were detected by western-blot. Apoptosis was studied by TUNEL and cleaved caspase 3. β-cell function was measured by glucose-stimulated insulin secretion. Epoxypukalide induced 2.5-fold increase in β-cell proliferation; this effect was mediated by activation of ERK1/2 signalling pathway and upregulation of the cell cycle activators, cyclin D2 and cyclin E. Interestingly, epoxypukalide showed protection from basal (40% lower versus control) and cytokine-induced apoptosis (80% lower versus control). Finally, epoxypukalide did not impair β-cell function when measured by glucose-stimulated insulin secretion. In conclusion, epoxypukalide induces β-cell proliferation and protects against basal and cytokine-mediated β-cell death in primary cultures of rat islets. These findings may be translated into new treatments for diabetes.
Collapse
Affiliation(s)
- José Francisco López-Acosta
- Instituto de Genética y Biología Molecular (IBGM)-Universidad de Valladolid, Valladolid, Spain
- Unidad de Investigación, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | | | | | - Ana R. Díaz-Marrero
- Instituto de Productos Naturales y Agrobiología del CSIC, La Laguna, Tenerife, Spain
| | - Mercedes Cueto
- Instituto de Productos Naturales y Agrobiología del CSIC, La Laguna, Tenerife, Spain
| | - Germán Perdomo
- Unidad de Investigación, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Irene Cózar-Castellano
- Instituto de Genética y Biología Molecular (IBGM)-Universidad de Valladolid, Valladolid, Spain
- Unidad de Investigación, Hospital Universitario Puerta del Mar, Cádiz, Spain
- * E-mail:
| |
Collapse
|
44
|
Orime K, Shirakawa J, Togashi Y, Tajima K, Inoue H, Ito Y, Sato K, Nakamura A, Aoki K, Goshima Y, Terauchi Y. Trefoil factor 2 promotes cell proliferation in pancreatic β-cells through CXCR-4-mediated ERK1/2 phosphorylation. Endocrinology 2013. [PMID: 23183167 DOI: 10.1210/en.2012-1814] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Decreased β-cell mass is a hallmark of type 2 diabetes, and therapeutic approaches to increase the pancreatic β-cell mass have been expected. In recent years, gastrointestinal incretin peptides have been shown to exert a cell-proliferative effect in pancreatic β-cells. Trefoil factor 2 (TFF2), which is predominantly expressed in the surface epithelium of the stomach, plays a role in antiapoptosis, migration, and proliferation. The TFF family is expressed in pancreatic β-cells, whereas the role of TFF2 in pancreatic β-cells has been obscure. In this study, we investigated the mechanism by which TFF2 enhances pancreatic β-cell proliferation. The effects of TFF2 on cell proliferation were evaluated in INS-1 cells, MIN6 cells, and mouse islets using an adenovirus vector containing TFF2 or a recombinant TFF2 peptide. The forced expression of TFF2 led to an increase in bromodeoxyuridine (BrdU) incorporation in both INS-1 cells and islets, without any alteration in insulin secretion. TFF2 significantly increased the mRNA expression of cyclin A2, D1, D2, D3, and E1 in islets. TFF2 peptide increased ERK1/2 phosphorylation and BrdU incorporation in MIN6 cells. A MAPK kinase inhibitor (U0126) abrogated the TFF2 peptide-mediated proliferation of MIN6 cells. A CX-chemokine receptor-4 antagonist also prevented the TFF2 peptide-mediated increase in ERK1/2 phosphorylation and BrdU incorporation in MIN6 cells. These results indicated that TFF2 is involved in β-cell proliferation at least partially via CX-chemokine receptor-4-mediated ERK1/2 phosphorylation, suggesting TFF2 may be a novel target for inducing β-cell proliferation.
Collapse
Affiliation(s)
- Kazuki Orime
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yang Y, Chang BHJ, Chan L. Sustained expression of the transcription factor GLIS3 is required for normal beta cell function in adults. EMBO Mol Med 2012. [PMID: 23197416 PMCID: PMC3569656 DOI: 10.1002/emmm.201201398] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Genome-wide association studies identified GLIS3 as a susceptibility locus for type 1 and type 2 diabetes. Global Glis3 deficiency in mice leads to congenital diabetes and neonatal lethality. In this study, we explore the role of Glis3 in adulthood using Glis3+/− and conditional knockout animals. We challenged Glis3+/− mice with high fat diet for 20 weeks and found that they developed diabetes because of impaired beta cell mass expansion. GLIS3 controls beta cell proliferation in response to high-fat feeding at least partly by regulating Ccnd2 transcription. To determine if sustained Glis3 expression is essential to normal beta cell function, we generated Glis3fl/fl/Pdx1CreERT+ animal by intercrossing Glis3fl/fl mice with Pdx1CreERT+ mice and used tamoxifen (TAM) to induce Glis3 deletion in adults. Adult Glis3fl/fl/Pdx1CreERT+ mice are euglycaemic. TAM-mediated beta cell-specific inactivation of Glis3 in adult mice downregulates insulin expression, leading to hyperglycaemia and subsequently enhanced beta cell apoptosis. We conclude that normal Glis3 expression is required for pancreatic beta cell function and mass maintenance during adulthood, which impairment leads to diabetes in adults.
Collapse
Affiliation(s)
- Yisheng Yang
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Diabetes and Endocrinology Research Center, Baylor College of Medicine, Houston, TX, USA
| | | | | |
Collapse
|
46
|
Goldfine AB, Kulkarni RN. Modulation of β-cell function: a translational journey from the bench to the bedside. Diabetes Obes Metab 2012; 14 Suppl 3:152-60. [PMID: 22928576 DOI: 10.1111/j.1463-1326.2012.01647.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Both decreased insulin secretion and action contribute to the pathogenesis of type 2 diabetes (T2D) in humans. The insulin receptor and insulin signalling proteins are present in the rodent and human β-cell and modulate cell growth and function. Insulin receptors and insulin signalling proteins in β-cells are critical for compensatory islet growth in response to insulin resistance. Rodents with tissue-specific knockout of the insulin receptor in the β-cell (βIRKO) show reduced first-phase glucose-stimulated insulin secretion (GSIS) and with aging develop glucose intolerance and diabetes, phenotypically similar to the process seen in human T2D. Expression of multiple insulin signalling proteins is reduced in islets of patients with T2D. Insulin potentiates GSIS in isolated human β-cells. Recent studies in humans in vivo show that pre-exposure to insulin increases GSIS, and this effect is diminished in persons with insulin resistance or T2D. β-Cell function correlates to whole-body insulin sensitivity. Together, these findings suggest that pancreatic β-cell dysfunction could be caused by a defect in insulin signalling within β-cell, and β-cell insulin resistance may lead to a loss of β-cell function and/or mass, contributing to the pathophysiology of T2D.
Collapse
Affiliation(s)
- A B Goldfine
- Section of Clinical Research, Joslin Diabetes Center, Boston, MA 02215, USA.
| | | |
Collapse
|
47
|
Kulkarni RN, Mizrachi EB, Ocana AG, Stewart AF. Human β-cell proliferation and intracellular signaling: driving in the dark without a road map. Diabetes 2012; 61:2205-13. [PMID: 22751699 PMCID: PMC3425429 DOI: 10.2337/db12-0018] [Citation(s) in RCA: 190] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A major goal in diabetes research is to find ways to enhance the mass and function of insulin secreting β-cells in the endocrine pancreas to prevent and/or delay the onset or even reverse overt diabetes. In this Perspectives in Diabetes article, we highlight the contrast between the relatively large body of information that is available in regard to signaling pathways, proteins, and mechanisms that together provide a road map for efforts to regenerate β-cells in rodents versus the scant information in human β-cells. To reverse the state of ignorance regarding human β-cell signaling, we suggest a series of questions for consideration by the scientific community to construct a human β-cell proliferation road map. The hope is that the knowledge from the new studies will allow the community to move faster towards developing therapeutic approaches to enhance human β-cell mass in the long-term goal of preventing and/or curing type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Rohit N. Kulkarni
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Corresponding authors: Rohit N. Kulkarni, , and Andrew F. Stewart,
| | - Ernesto-Bernal Mizrachi
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Adolfo Garcia Ocana
- Division of Endocrinology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Andrew F. Stewart
- Division of Endocrinology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Corresponding authors: Rohit N. Kulkarni, , and Andrew F. Stewart,
| |
Collapse
|
48
|
Pascoe J, Hollern D, Stamateris R, Abbasi M, Romano LC, Zou B, O’Donnell CP, Garcia-Ocana A, Alonso LC. Free fatty acids block glucose-induced β-cell proliferation in mice by inducing cell cycle inhibitors p16 and p18. Diabetes 2012; 61:632-41. [PMID: 22338094 PMCID: PMC3282818 DOI: 10.2337/db11-0991] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pancreatic β-cell proliferation is infrequent in adult humans and is not increased in type 2 diabetes despite obesity and insulin resistance, suggesting the existence of inhibitory factors. Free fatty acids (FFAs) may influence proliferation. In order to test whether FFAs restrict β-cell proliferation in vivo, mice were intravenously infused with saline, Liposyn II, glucose, or both, continuously for 4 days. Lipid infusion did not alter basal β-cell proliferation, but blocked glucose-stimulated proliferation, without inducing excess β-cell death. In vitro exposure to FFAs inhibited proliferation in both primary mouse β-cells and in rat insulinoma (INS-1) cells, indicating a direct effect on β-cells. Two of the fatty acids present in Liposyn II, linoleic acid and palmitic acid, both reduced proliferation. FFAs did not interfere with cyclin D2 induction or nuclear localization by glucose, but increased expression of inhibitor of cyclin dependent kinase 4 (INK4) family cell cycle inhibitors p16 and p18. Knockdown of either p16 or p18 rescued the antiproliferative effect of FFAs. These data provide evidence for a novel antiproliferative form of β-cell glucolipotoxicity: FFAs restrain glucose-stimulated β-cell proliferation in vivo and in vitro through cell cycle inhibitors p16 and p18. If FFAs reduce proliferation induced by obesity and insulin resistance, targeting this pathway may lead to new treatment approaches to prevent diabetes.
Collapse
Affiliation(s)
- Jordan Pascoe
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas Hollern
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rachel Stamateris
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Munira Abbasi
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lia C. Romano
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Baobo Zou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher P. O’Donnell
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adolfo Garcia-Ocana
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Laura C. Alonso
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
- Corresponding author: Laura C. Alonso,
| |
Collapse
|
49
|
Wang Y, Liu Y, Wang H, Li C, Qi P, Bao J. Agaricus bisporus lectins mediates islet β-cell proliferation through regulation of cell cycle proteins. Exp Biol Med (Maywood) 2012; 237:287-96. [DOI: 10.1258/ebm.2011.011251] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This study was designed to determine the therapeutic effect of Agaricus bisporus lectins (ABL) by the regeneration of β-cells in mice following 70% partial pancreatectomy (PPx), and to explore the mechanisms of ABL-induced β-cell proliferation. Adult C57BL/6J mice were subjected to a 70% PPx operation or a sham operation, and mice received 10 mg/kg body weight of ABL or saline immediately after surgery. Blood glucose concentrations and insulin secretion levels were measured. To determine the growth rates of β-cells and duct cells, immunohistological analysis of pancreatic tissues was performed. Key cell cycle proteins and β-cell specific genes were measured by realtime polymerase chain reaction, Western blotting and immunohistological staining. In this study, a significant decrease in blood glucose concentrations, increase in glucose tolerance and expanded β-cell mass were observed in the ABL-treated mice. At the same time, after ABL treatment, increased β-cell proliferation rates were observed. Further studies on the expression of cyclin D1, cyclin D2 and Cdk4 demonstrated that these genes were significantly up-regulated in the ABL-treated mice. Meanwhile, Cdk4 activity was also enhanced. Moreover, the expression of PDX-1 (pancreatic and duodenal homeobox 1), Ngn3 (neurogenin 3), insulin, GLUT-1 (glucose transporter 1) and glucokinase was also increased in the ABL-treated mice. These findings demonstrate that ABL administration could partially reverse the impaired β-cell growth potential by regulating cell cycle proteins. Induction of islet β-cell proliferation by ABL suggests the therapeutic potential in preventing and/or treating diabetes.
Collapse
Affiliation(s)
- Yi Wang
- School of Life Science, Sichuan University, Chengdu, Sichuan 610064
- Institute of Organ Transplantation, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072
| | - Yuande Liu
- 91388 Military Hospital, Guangdong, Zhanjiang 524022, China
| | - Hailian Wang
- Institute of Organ Transplantation, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072
| | - Chunyang Li
- School of Life Science, Sichuan University, Chengdu, Sichuan 610064
| | - Ping Qi
- Institute of Organ Transplantation, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072
| | - Jinku Bao
- School of Life Science, Sichuan University, Chengdu, Sichuan 610064
| |
Collapse
|
50
|
Velazquez-Garcia S, Valle S, Rosa TC, Takane KK, Demirci C, Alvarez-Perez JC, Mellado-Gil JM, Ernst S, Scott DK, Vasavada RC, Alonso LC, Garcia-Ocaña A. Activation of protein kinase C-ζ in pancreatic β-cells in vivo improves glucose tolerance and induces β-cell expansion via mTOR activation. Diabetes 2011; 60:2546-59. [PMID: 21911744 PMCID: PMC3178296 DOI: 10.2337/db10-1783] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE PKC-ζ activation is a key signaling event for growth factor-induced β-cell replication in vitro. However, the effect of direct PKC-ζ activation in the β-cell in vivo is unknown. In this study, we examined the effects of PKC-ζ activation in β-cell expansion and function in vivo in mice and the mechanisms associated with these effects. RESEARCH DESIGN AND METHODS We characterized glucose homeostasis and β-cell phenotype of transgenic (TG) mice with constitutive activation of PKC-ζ in the β-cell. We also analyzed the expression and regulation of signaling pathways, G1/S cell cycle molecules, and β-cell functional markers in TG and wild-type mouse islets. RESULTS TG mice displayed increased plasma insulin, improved glucose tolerance, and enhanced insulin secretion with concomitant upregulation of islet insulin and glucokinase expression. In addition, TG mice displayed increased β-cell proliferation, size, and mass compared with wild-type littermates. The increase in β-cell proliferation was associated with upregulation of cyclins D1, D2, D3, and A and downregulation of p21. Phosphorylation of D-cyclins, known to initiate their rapid degradation, was reduced in TG mouse islets. Phosphorylation/inactivation of GSK-3β and phosphorylation/activation of mTOR, critical regulators of D-cyclin expression and β-cell proliferation, were enhanced in TG mouse islets, without changes in Akt phosphorylation status. Rapamycin treatment in vivo eliminated the increases in β-cell proliferation, size, and mass; the upregulation of cyclins Ds and A in TG mice; and the improvement in glucose tolerance-identifying mTOR as a novel downstream mediator of PKC-ζ-induced β-cell replication and expansion in vivo. CONCLUSIONS PKC:-ζ, through mTOR activation, modifies the expression pattern of β-cell cycle molecules leading to increased β-cell replication and mass with a concomitant enhancement in β-cell function. Approaches to enhance PKC-ζ activity may be of value as a therapeutic strategy for the treatment of diabetes.
Collapse
Affiliation(s)
- Silvia Velazquez-Garcia
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shelley Valle
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Taylor C. Rosa
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Karen K. Takane
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Cem Demirci
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Juan C. Alvarez-Perez
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jose M. Mellado-Gil
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sara Ernst
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Donald K. Scott
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rupangi C. Vasavada
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Laura C. Alonso
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adolfo Garcia-Ocaña
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
- Corresponding author: Adolfo Garcia-Ocaña,
| |
Collapse
|