1
|
Kadhim AZ, Vanderkruk B, Mar S, Dan M, Zosel K, Xu EE, Spencer RJ, Sasaki S, Cheng X, Sproul SLJ, Speckmann T, Nian C, Cullen R, Shi R, Luciani DS, Hoffman BG, Taubert S, Lynn FC. Transcriptional coactivator MED15 is required for beta cell maturation. Nat Commun 2024; 15:8711. [PMID: 39379383 PMCID: PMC11461855 DOI: 10.1038/s41467-024-52801-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/23/2024] [Indexed: 10/10/2024] Open
Abstract
Mediator, a co-regulator complex required for RNA Polymerase II activity, interacts with tissue-specific transcription factors to regulate development and maintain homeostasis. We observe reduced Mediator subunit MED15 expression in endocrine hormone-producing pancreatic islets isolated from people living with type 2 diabetes and sought to understand how MED15 and Mediator control gene expression programs important for the function of insulin-producing β-cells. Here we show that Med15 is expressed during mouse β-cell development and maturation. Knockout of Med15 in mouse β-cells causes defects in β-cell maturation without affecting β-cell mass or insulin expression. ChIP-seq and co-immunoprecipitation analyses found that Med15 binds β-cell transcription factors Nkx6-1 and NeuroD1 to regulate key β-cell maturation genes. In support of a conserved role during human development, human embryonic stem cell-derived β-like cells, genetically engineered to express high levels of MED15, express increased levels of maturation markers. We provide evidence of a conserved role for Mediator in β-cell maturation and demonstrate an additional layer of control that tunes β-cell transcription factor function.
Collapse
Affiliation(s)
- Alex Z Kadhim
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Ben Vanderkruk
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Samantha Mar
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Meixia Dan
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Katarina Zosel
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Eric E Xu
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Rachel J Spencer
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Shugo Sasaki
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Xuanjin Cheng
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Shannon L J Sproul
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Thilo Speckmann
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Cuilan Nian
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Robyn Cullen
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Rocky Shi
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Dan S Luciani
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Bradford G Hoffman
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Stefan Taubert
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada.
| | - Francis C Lynn
- Diabetes Research Program, BC Children's Hospital Research Institute, Vancouver, Canada.
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
2
|
Azad A, Altunbas HA, Manguoglu AE. From islet transplantation to beta-cell regeneration: an update on beta-cell-based therapeutic approaches in type 1 diabetes. Expert Rev Endocrinol Metab 2024; 19:217-227. [PMID: 38693782 DOI: 10.1080/17446651.2024.2347263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 03/06/2024] [Indexed: 05/03/2024]
Abstract
INTRODUCTION Type 1 diabetes (T1D) mellitus is an autoimmune disease in which immune cells, predominantly effector T cells, destroy insulin-secreting beta-cells. Beta-cell destruction led to various consequences ranging from retinopathy and nephropathy to neuropathy. Different strategies have been developed to achieve normoglycemia, including exogenous glucose compensation, whole pancreas transplantation, islet transplantation, and beta-cell replacement. AREAS COVERED The last two decades of experience have shown that indigenous glucose compensation through beta-cell regeneration and protection is a peerless method for T1D therapy. Tremendous studies have tried to find an unlimited source for beta-cell regeneration, on the one hand, and beta-cell protection against immune attack, on the other hand. Recent advances in stem cell technology, gene editing methods, and immune modulation approaches provide a unique opportunity for both beta-cell regeneration and protection. EXPERT OPINION Pluripotent stem cell differentiation into the beta-cell is considered an unlimited source for beta-cell regeneration. Devising engineered pancreas-specific regulatory T cells using Chimeric Antigen Receptor (CAR) technology potentiates an effective immune tolerance induction for beta-cell protection. Beta-cell regeneration using pluripotent stem cells and beta-cell protection using pancreas-specific engineered regulatory T cells promises to develop a curative protocol in T1D.
Collapse
Affiliation(s)
- Asef Azad
- Department of Medical Biology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Hasan Ali Altunbas
- Department of Endocrinology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ayse Esra Manguoglu
- Department of Medical Biology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
3
|
Cheng Z, Guo D, Ruzi A, Pan T, You K, Chen Y, Huang X, Zhang J, Yang F, Niu L, Xu K, Li YX. Modeling MEN1 with Patient-Origin iPSCs Reveals GLP-1R Mediated Hypersecretion of Insulin. Cells 2022; 11:2387. [PMID: 35954231 PMCID: PMC9368616 DOI: 10.3390/cells11152387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 12/10/2022] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is an inherited disease caused by mutations in the MEN1 gene encoding a nuclear protein menin. Among those different endocrine tumors of MEN1, the pancreatic neuroendocrine tumors (PNETs) are life-threatening and frequently implicated. Since there are uncertainties in genotype and phenotype relationship and there are species differences between humans and mice, it is worth it to replenish the mice model with human cell resources. Here, we tested whether the patient-origin induced pluripotent stem cell (iPSC) lines could phenocopy some defects of MEN1. In vitro β-cell differentiation revealed that the percentage of insulin-positive cells and insulin secretion were increased by at least two-fold in MEN1-iPSC derived cells, which was mainly resulted from significantly higher proliferative activities in the pancreatic progenitor stage (Day 7-13). This scenario was paralleled with increased expressions of prohormone convertase1/3 (PC1/3), glucagon-like peptide-1 (GLP-1), GLP-1R, and factors in the phosphatidylinositol 3-kinase (PI3K)/AKT signal pathway, and the GLP-1R was mainly expressed in β-like cells. Blockages of either GLP-1R or PI3K significantly reduced the percentages of insulin-positive cells and hypersecretion of insulin in MEN1-derived cells. Furthermore, in transplantation of different stages of MEN1-derived cells into immune-deficient mice, only those β-like cells produced tumors that mimicked the features of the PNETs from the original patient. To the best of our knowledge, this was the first case using patient-origin iPSCs modeling most phenotypes of MEN1, and the results suggested that GLP-1R may be a potential therapeutic target for MEN1-related hyperinsulinemia.
Collapse
Affiliation(s)
- Ziqi Cheng
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Dongsheng Guo
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
| | - Aynisahan Ruzi
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
| | - Tingcai Pan
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
| | - Kai You
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
| | - Yan Chen
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xinping Huang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Jiaye Zhang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
| | - Fan Yang
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China;
| | - Lizhi Niu
- Guangzhou Fuda Cancer Hospital, Guangzhou 510305, China; (L.N.); (K.X.)
| | - Kecheng Xu
- Guangzhou Fuda Cancer Hospital, Guangzhou 510305, China; (L.N.); (K.X.)
| | - Yin-Xiong Li
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (Z.C.); (D.G.); (A.R.); (T.P.); (K.Y.); (Y.C.); (X.H.); (J.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- State Key Laboratory of Respiratory Disease, Guangzhou 510000, China
- China-New Zealand Joint Laboratory of Biomedicine and Health, Guangzhou 510530, China
| |
Collapse
|
4
|
Evans RM, Wei Z. Interorgan crosstalk in pancreatic islet function and pathology. FEBS Lett 2022; 596:607-619. [PMID: 35014695 DOI: 10.1002/1873-3468.14282] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022]
Abstract
Pancreatic β cells secrete insulin in response to glucose, a process that is regulated at multiple levels, including a network of input signals from other organ systems. Impaired islet function contributes to the pathogenesis of type 2 diabetes mellitus (T2DM), and targeting inter-organ communications, such as GLP-1 signalling, to enhance β-cell function has been proven to be a successful therapeutic strategy in the last decade. In this review, we will discuss recent advances in inter-organ communication from the metabolic, immune and neural system to pancreatic islets, their biological implication in normal pancreas endocrine function and their role in the (mal)adaptive responses of islet to nutrition-induced stress.
Collapse
Affiliation(s)
- Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zong Wei
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, USA
| |
Collapse
|
5
|
Barella LF, Rossi M, Pydi SP, Meister J, Jain S, Cui Y, Gavrilova O, Fulgenzi G, Tessarollo L, Wess J. β-Arrestin-1 is required for adaptive β-cell mass expansion during obesity. Nat Commun 2021; 12:3385. [PMID: 34099679 PMCID: PMC8184739 DOI: 10.1038/s41467-021-23656-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/11/2021] [Indexed: 01/14/2023] Open
Abstract
Obesity is the key driver of peripheral insulin resistance, one of the key features of type 2 diabetes (T2D). In insulin-resistant individuals, the expansion of beta-cell mass is able to delay or even prevent the onset of overt T2D. Here, we report that beta-arrestin-1 (barr1), an intracellular protein known to regulate signaling through G protein-coupled receptors, is essential for beta-cell replication and function in insulin-resistant mice maintained on an obesogenic diet. Specifically, insulin-resistant beta-cell-specific barr1 knockout mice display marked reductions in beta-cell mass and the rate of beta-cell proliferation, associated with pronounced impairments in glucose homeostasis. Mechanistic studies suggest that the observed metabolic deficits are due to reduced Pdx1 expression levels caused by beta-cell barr1 deficiency. These findings indicate that strategies aimed at enhancing barr1 activity and/or expression in beta-cells may prove useful to restore proper glucose homeostasis in T2D.
Collapse
Affiliation(s)
- Luiz F Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.
| | - Mario Rossi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Bethesda, MD, USA
| | - Gianluca Fulgenzi
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.
| |
Collapse
|
6
|
Jiang Z, Yang M, Jin J, Song Z, Li C, Zhu Y, Tang Y, Ni C. miR-124-3p Down-Regulation Influences Pancreatic-β-Cell Function by Targeting Secreted Frizzled-Related Protein 5 (SFRP5) in Diabetes Mellitus. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Diabetes mellitus (DM) is a complex metabolic disease characterized by hyperglycemia, insulin resistance and pancreatic β-cell dysfunction. There are evidences showed that microRNAs (miRNAs) play important roles in DM. The purpose of our study was to determine the role of miR-124-3p
in DM. Quantitative reverse transcription PCR (qRT-PCR) was applied to measure the level of miR- 124-3p in peripheral blood from healthy control patients and DM patients. Then we explored the effects of miR-124-3p inhibitor on the secretion of insulin of pancreatic β-cells. Moreover,
we determined the effects of miR-124-3p inhibitor on the apoptosis and viability of pancreatic β-cells through flow cytometry and MTT assay. And we also used western blotting to detect the protein expression of cleaved-caspase3/pro-caspase3, and the activity of caspase3 was detected.
In addition, we confirmed the direct target of miR-124-3p using Dual luciferase reporter assay. Our data showed that in the blood of DM patients, SFRP5 was significantly reduced, while miR-124-3p was increased significantly. Furthermore, we found that down-regulation of miR-124-3p increased
total insulin content in INS-1 cells, enhanced insulin secretion in INS-1 cells. Furthermore, we revealed that miR-124-3p inhibitor enhanced INS-1 cell viability, decreased apoptosis of INS-1 cells, increased pro-caspase3 expression, decreased cleaved-caspase3 expression and caspase3 activity.
In addition, we proved SFRP5 was a direct target of miR-124-3p in pancreatic β-cells. Moreover, SFRP5-siRNA reversed all the effects of miR-124-3p knockdown on pancreatic β-cells.
Collapse
Affiliation(s)
- Zhenhuan Jiang
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134,
China
| | - Min Yang
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Jianming Jin
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Zhenqiang Song
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Chenguang Li
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Yanjuan Zhu
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Yunzhao Tang
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Changlin Ni
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| |
Collapse
|
7
|
Tatsuoka H, Sakamoto S, Yabe D, Kabai R, Kato U, Okumura T, Botagarova A, Tokumoto S, Usui R, Ogura M, Nagashima K, Mukai E, Fujitani Y, Watanabe A, Inagaki N. Single-Cell Transcriptome Analysis Dissects the Replicating Process of Pancreatic Beta Cells in Partial Pancreatectomy Model. iScience 2020; 23:101774. [PMID: 33294783 PMCID: PMC7689163 DOI: 10.1016/j.isci.2020.101774] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/16/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022] Open
Abstract
Heterogeneity of gene expression and rarity of replication hamper molecular analysis of β-cell mass restoration in adult pancreas. Here, we show transcriptional dynamics in β-cell replication process by single-cell RNA sequencing of murine pancreas with or without partial pancreatectomy. We observed heterogeneity of Ins1-expressing β-cells and identified the one cluster as replicating β-cells with high expression of cell proliferation markers Pcna and Mki67. We also recapitulated cell cycle transition accompanied with switching expression of cyclins and E2F transcription factors. Both transient activation of endoplasmic reticulum stress responders like Atf6 and Hspa5 and elevated expression of tumor suppressors like Trp53, Rb1, and Brca1 and DNA damage responders like Atm, Atr, Rad51, Chek1, and Chek2 during the transition to replication associated fine balance of cell cycle progression and protection from DNA damage. Taken together, these results provide a high-resolution map depicting a sophisticated genetic circuit for replication of the β-cells. Single cell RNA sequencing dissects a sequence of replication process of beta cells ER stress responders are transiently activated in initiation of the proliferation Physiological replication accompanied with induced expression of tumor suppressors Fine balance of proliferation genes and tumor suppressors is a key of the replication
Collapse
Affiliation(s)
- Hisato Tatsuoka
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoko Sakamoto
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Diabetes and Endocrinology, Gifu University Graduate School of Medicine, Gifu, Japan.,Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Hyogo, Japan.,Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Ryotaro Kabai
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Unyanee Kato
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tatsuya Okumura
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ainur Botagarova
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinsuke Tokumoto
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryota Usui
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahito Ogura
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuaki Nagashima
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Eri Mukai
- Laboratory of Medical Physiology and Metabolism, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Shiga, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular & Cellular Regulation, Gunma University, Gunma, Japan
| | - Akira Watanabe
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
8
|
Wu L, Xiong L, Li J, Peng Z, Zhang L, Shi P, Gong Y, Xiao H. Circ-Tulp4 promotes β-cell adaptation to lipotoxicity by regulating soat1 expression. J Mol Endocrinol 2020; 65:149-161. [PMID: 33064661 PMCID: PMC7576671 DOI: 10.1530/jme-20-0079] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 09/11/2020] [Indexed: 12/18/2022]
Abstract
This study aimed to identify circular RNAs differentially expressed in the islets of type 2 diabetes (T2DM) models and clarify their roles in the control of β-cell functions. Circular RNAs dysregulated in the islets of diabetic db/db mice were identified by high-throughput RNA sequencing. Then, the expression level of the selected circular RNA circ-Tulp4 was confirmed by real-time PCR in the islets of diabetic models and Min6 cells. MTS, EdU, western blot, flow cytometric analysis, and luciferase assay were performed to investigate the impact of circ-Tulp4 on β-cell functions. This study identified thousands of circular RNAs in mouse pancreatic islets. The circ-Tulp4 level significantly decreased in the diabetic models and altered in the Min6 cells under lipotoxic condition. The modulation of circ-Tulp4 level in Min6 cells regulated cell proliferation. Furthermore, an interaction was demonstrated between circ-Tulp4 and miR-7222-3p, which suppressed the expression of cholesterol esterification-related gene, sterol O-acyltransferase 1 (SOAT1). The accumulation of soat1 activated cyclin D1 expression, thus promoting cell cycle progression. These findings showed that circ-Tulp4 regulated β-cell proliferation via miR-7222-3p/soat1/cyclin D1 signaling. Our research suggested that circ-Tulp4 might be a potential therapeutic intervention for T2DM. Besides, soat1 might be important for β-cell adaptation to lipotoxicity.
Collapse
Affiliation(s)
- Liting Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Xiong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jin Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zishan Peng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Luyao Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Peijie Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingying Gong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Correspondence should be addressed to Y Gong or H Xiao: or
| | - Haipeng Xiao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Correspondence should be addressed to Y Gong or H Xiao: or
| |
Collapse
|
9
|
Lin YCD, Huang HY, Shrestha S, Chou CH, Chen YH, Chen CR, Hong HC, Li J, Chang YA, Chiew MY, Huang YR, Tu SJ, Sun TH, Weng SL, Tseng CP, Huang HD. Multi-omics profiling reveals microRNA-mediated insulin signaling networks. BMC Bioinformatics 2020; 21:389. [PMID: 32938376 PMCID: PMC7496206 DOI: 10.1186/s12859-020-03678-0] [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] [Indexed: 01/17/2023] Open
Abstract
Background MicroRNAs (miRNAs) play a key role in mediating the action of insulin on cell growth and the development of diabetes. However, few studies have been conducted to provide a comprehensive overview of the miRNA-mediated signaling network in response to glucose in pancreatic beta cells. In our study, we established a computational framework integrating multi-omics profiles analyses, including RNA sequencing (RNA-seq) and small RNA sequencing (sRNA-seq) data analysis, inverse expression pattern analysis, public data integration, and miRNA targets prediction to illustrate the miRNA-mediated regulatory network at different glucose concentrations in INS-1 pancreatic beta cells (INS-1), which display important characteristics of the pancreatic beta cells. Results We applied our computational framework to the expression profiles of miRNA/mRNA of INS-1, at different glucose concentrations. A total of 1437 differentially expressed genes (DEGs) and 153 differentially expressed miRNAs (DEmiRs) were identified from multi-omics profiles. In particular, 121 DEmiRs putatively regulated a total of 237 DEGs involved in glucose metabolism, fatty acid oxidation, ion channels, exocytosis, homeostasis, and insulin gene regulation. Moreover, Argonaute 2 immunoprecipitation sequencing, qRT-PCR, and luciferase assay identified Crem, Fn1, and Stc1 are direct targets of miR-146b and elucidated that miR-146b acted as a potential regulator and promising target to understand the insulin signaling network. Conclusions In this study, the integration of experimentally verified data with system biology framework extracts the miRNA network for exploring potential insulin-associated miRNA and their target genes. The findings offer a potentially significant effect on the understanding of miRNA-mediated insulin signaling network in the development and progression of pancreatic diabetes.
Collapse
Affiliation(s)
- Yang-Chi-Dung Lin
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Hsi-Yuan Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Sirjana Shrestha
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Chih-Hung Chou
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Yen-Hua Chen
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, 10021, USA
| | - Chi-Ru Chen
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Hsiao-Chin Hong
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Jing Li
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China.,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China
| | - Yi-An Chang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Men-Yee Chiew
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Ya-Rong Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Siang-Jyun Tu
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Ting-Hsuan Sun
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Shun-Long Weng
- Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital, Hsinchu, 300, Taiwan
| | - Ching-Ping Tseng
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan.
| | - Hsien-Da Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China. .,Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Longgang District, Shenzhen, 518172, Guangdong Province, China. .,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 300, Taiwan.
| |
Collapse
|
10
|
Zhang JR, Sun HJ. Roles of circular RNAs in diabetic complications: From molecular mechanisms to therapeutic potential. Gene 2020; 763:145066. [PMID: 32827686 DOI: 10.1016/j.gene.2020.145066] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Diabetes is characterized by changed homeostasis of blood glucose levels, which is associated with various complications, including cardiomyopathy, atherosclerosis, endothelial dysfunction, nephropathy, retinopathy and neuropathy. In recent years, accumulative evidence has demonstrated that circular RNAs are identified as a novel type of noncoding RNAs (ncRNAs) involving in the regulation of various physiological processes and pathologic conditions. Specifically, the emergence of complications response to diabetes is finely controlled by a complex gene regulatory network in which circular RNAs play a critical role. Recently, circular RNAs are emerging as messengers that could influence cellular functions under diabetic conditions. Dysregulation of circular RNAs has been closely linked to the pathophysiology of diabetes-related complications. In this review, we aimed to summarize the current progression and underlying mechanisms of circular RNA in the development of diabetes-related complications. We will also provide an overview of circular RNA-regulated cell communications in different types of cells that have been linked to diabetic complications. We anticipated that the completion of this review will provide potential clues for developing novel circular RNAs-based biomarkers or therapeutic targets for diabetes and its associated complications.
Collapse
Affiliation(s)
- Ji-Ru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Wuxi 214062, PR China
| | - Hai-Jian Sun
- Department of Basic Medicine, Wuxi Medical School, Jiangnan University, Wuxi 214122, PR China; Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
11
|
Xu W, Cui J, Zhou F, Bai M, Deng R, Wang W. Leonurine protects against dexamethasone-induced cytotoxicity in pancreatic β-cells via PI3K/Akt signaling pathway. Biochem Biophys Res Commun 2020; 529:652-658. [DOI: 10.1016/j.bbrc.2020.05.184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/27/2023]
|
12
|
Li W, Li P, Li R, Yu Z, Sun X, Ji G, Yang X, Zhu L, Zhu S. GLP1R Single-Nucleotide Polymorphisms rs3765467 and rs10305492 Affect β Cell Insulin Secretory Capacity and Apoptosis Through GLP-1. DNA Cell Biol 2020; 39:1700-1710. [PMID: 32721233 DOI: 10.1089/dna.2020.5424] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The increased secretion of glucagon-like peptide-1 (GLP-1) after Roux-en-Y gastric bypass (RYGB) is regarded as the main reason for the improvement of blood glucose. However, the single-nucleotide polymorphisms (SNPs) of GLP-1 Receptor (GLP1R) impair receptor function, subsequently affecting β cell insulin secretion function, ultimately affecting the efficacy of RYGB. In this study, we revealed that two SNPs in GLP1R gene, rs3765467 and rs10305492, could significantly reduce the insulin secreted by β cells and the cyclic AMP concentration, whereas promote β cell apoptosis. Under high glucose exposure, rs3765467 and rs10305492 impaired β cell secretion of insulin and β cell viability in the same way; in other words, GLP1R rs3765467 and rs10305492 exert an effect on pancreatic β cell glucose-stimulated insulin secretion. Moreover, GLP-1 antagonist Exendin (9-39) further enhanced, whereas GLP-1 agonist Exendin-4 partially attenuated the effects of SNPs on the functions and apoptosis of β cells. In conclusion, the rs3765467 and rs10305492 SNPs in GLP1R show to exert a critical effect on regulating insulin secretory capacity of β cells and β cell mass. Through leading to the dysfunction and apoptosis of β cells, GLP1R rs3765467 and rs10305492 might also impair GLP-1 interaction with GLP1R, therefore attenuating the therapeutic effect of RYGB.
Collapse
Affiliation(s)
- Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Rao Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhaomei Yu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guangnian Ji
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiangwu Yang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Abbaszadeh-Goudarzi K, Radbakhsh S, Pourhanifeh MH, Khanbabaei H, Davoodvandi A, Fathizadeh H, Sahebkar A, Shahrzad MK, Mirzaei H. Circular RNA and Diabetes: Epigenetic Regulator with Diagnostic Role. Curr Mol Med 2020; 20:516-526. [DOI: 10.2174/1566524020666200129142106] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/12/2019] [Accepted: 01/01/2020] [Indexed: 11/22/2022]
Abstract
Circular RNAs, a group of endogenous non-coding RNAs, are characterized
by covalently closed cyclic structures with no poly-adenylated tails. It has been recently
recommended that cirRNAs have an essential role in regulating genes expression by
functioning as a translational regulator, RNA binding protein sponge and microRNA
sponge. Due to their close relation to the progression of various diseases such as
diabetes, circRNAs have become a research hotspot. A number of circRNAs (i.e.,
circRNA_0054633, circHIPK3, circANKRD36, and circRNA11783-2) have been shown
to be associated with initiation and progression of diabetes. Based on reports, in a
tissue, some circRNAs are expressed in a developmental stage-specific manner. In this
study, we reviewed research on circular RNAs involved in the pathogenesis and
diagnosis of diabetes and their prognostic roles.
Collapse
Affiliation(s)
- Kazem Abbaszadeh-Goudarzi
- Cellular and Molecular Research Center, Department of Biochemistry and Nutrition, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Shabnam Radbakhsh
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hashem Khanbabaei
- Radiobiology Laboratory, Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Hadis Fathizadeh
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Karim Shahrzad
- Department of Internal Medicine and endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
14
|
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
|
15
|
Abstract
PURPOSE OF REVIEW Pancreatic β-cells play a critical role in whole-body glucose homeostasis by regulating the release of insulin in response to minute by minute alterations in metabolic demand. As such, β-cells are staunchly resilient but there are circumstances where they can become functionally compromised or physically lost due to pathophysiological changes which culminate in overt hyperglycemia and diabetes. RECENT FINDINGS In humans, β-cell mass appears to be largely defined in the postnatal period and this early replicative and generative phase is followed by a refractory state which persists throughout life. Despite this, efforts to identify physiological and pharmacological factors which might re-initiate β-cell replication (or cause the replenishment of β-cells by neogenesis or transdifferentiation) are beginning to bear fruit. Controlled manipulation of β-cell mass in humans still represents a holy grail for therapeutic intervention in diabetes, but progress is being made which may lead to ultimate success.
Collapse
Affiliation(s)
- Giorgio Basile
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Rohit N. Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Noel G. Morgan
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter EX2 5DW, UK
| |
Collapse
|
16
|
Chen X, Zhang J, Zhou Z. Targeting Islets: Metabolic Surgery Is More than a Bariatric Surgery. Obes Surg 2019; 29:3001-3009. [DOI: 10.1007/s11695-019-03979-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
17
|
Singh A, Gibert Y, Dwyer KM. The adenosine, adrenergic and opioid pathways in the regulation of insulin secretion, beta cell proliferation and regeneration. Pancreatology 2018; 18:615-623. [PMID: 29937364 DOI: 10.1016/j.pan.2018.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/25/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Insulin, a key hormone produced by pancreatic beta cells precisely regulates glucose metabolism in vertebrates. In type 1 diabetes, the beta cell mass is destroyed, a process triggered by a combination of environmental and genetic factors. This ultimately results in absolute insulin deficiency and dysregulated glucose metabolism resulting in a number of detrimental pathophysiological effects. The traditional focus of treating type 1 diabetes has been to control blood sugar levels through the administration of exogenous insulin. Newer approaches aim to replace the beta cell mass through pancreatic or islet transplantation. Type 2 diabetes results from a relative insulin deficiency for the prevailing insulin resistance. Treatments are generally aimed at reducing insulin resistance and/or augmenting insulin secretion and the use of insulin itself is often required. It is increasingly being recognized that the beta cell mass is dynamic and increases insulin secretion in response to beta cell mitogens and stress signals to maintain glycemia within a very narrow physiological range. This review critically discusses the role of adrenergic, adenosine and opioid pathways and their interrelationship in insulin secretion, beta cell proliferation and regeneration.
Collapse
Affiliation(s)
- Amitoj Singh
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Yann Gibert
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Karen M Dwyer
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia.
| |
Collapse
|
18
|
Stoll L, Sobel J, Rodriguez-Trejo A, Guay C, Lee K, Venø MT, Kjems J, Laybutt DR, Regazzi R. Circular RNAs as novel regulators of β-cell functions in normal and disease conditions. Mol Metab 2018; 9:69-83. [PMID: 29396373 PMCID: PMC5870096 DOI: 10.1016/j.molmet.2018.01.010] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE There is strong evidence for an involvement of different classes of non-coding RNAs, including microRNAs and long non-coding RNAs, in the regulation of β-cell activities and in diabetes development. Circular RNAs were recently discovered to constitute a substantial fraction of the mammalian transcriptome but the contribution of these non-coding RNAs in physiological and disease processes remains largely unknown. The goal of this study was to identify the circular RNAs expressed in pancreatic islets and to elucidate their possible role in the control of β-cells functions. METHODS We used a microarray approach to identify circular RNAs expressed in human islets and searched their orthologues in RNA sequencing data from mouse islets. We then measured the level of four selected circular RNAs in the islets of different Type 1 and Type 2 diabetes models and analyzed the role of these circular transcripts in the regulation of insulin secretion, β-cell proliferation, and apoptosis. RESULTS We identified thousands of circular RNAs expressed in human pancreatic islets, 497 of which were conserved in mouse islets. The level of two of these circular transcripts, circHIPK3 and ciRS-7/CDR1as, was found to be reduced in the islets of diabetic db/db mice. Mimicking this decrease in the islets of wild type animals resulted in impaired insulin secretion, reduced β-cell proliferation, and survival. ciRS-7/CDR1as has been previously proposed to function by blocking miR-7. Transcriptomic analysis revealed that circHIPK3 acts by sequestering a group of microRNAs, including miR-124-3p and miR-338-3p, and by regulating the expression of key β-cell genes, such as Slc2a2, Akt1, and Mtpn. CONCLUSIONS Our findings point to circular RNAs as novel regulators of β-cell activities and suggest an involvement of this novel class of non-coding RNAs in β-cell dysfunction under diabetic conditions.
Collapse
Affiliation(s)
- Lisa Stoll
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland
| | - Jonathan Sobel
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland
| | | | - Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland
| | - Kailun Lee
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Morten Trillingsgaard Venø
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - D Ross Laybutt
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland.
| |
Collapse
|
19
|
Bhalla N, Sathish S, Sinha A, Shen AQ. Large-Scale Nanophotonic Structures for Long-Term Monitoring of Cell Proliferation. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201700258] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Nikhil Bhalla
- Micro/Bio/Nanofluidics Unit; Okinawa Institute of Science and Technology Graduate University; 1919-1 Tancha, Onna Kunigami District Okinawa Prefecture 904-0495 Japan
| | - Shivani Sathish
- Micro/Bio/Nanofluidics Unit; Okinawa Institute of Science and Technology Graduate University; 1919-1 Tancha, Onna Kunigami District Okinawa Prefecture 904-0495 Japan
| | - Abhishek Sinha
- Micro/Bio/Nanofluidics Unit; Okinawa Institute of Science and Technology Graduate University; 1919-1 Tancha, Onna Kunigami District Okinawa Prefecture 904-0495 Japan
| | - Amy Q. Shen
- Micro/Bio/Nanofluidics Unit; Okinawa Institute of Science and Technology Graduate University; 1919-1 Tancha, Onna Kunigami District Okinawa Prefecture 904-0495 Japan
| |
Collapse
|
20
|
Ackeifi CA, Swartz EA, Wang P. Cell-Based Methods to Identify Inducers of Human Pancreatic Beta-Cell Proliferation. Methods Mol Biol 2018; 1787:87-100. [PMID: 29736712 DOI: 10.1007/978-1-4939-7847-2_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Diabetes is the result of the insufficiency or dysfunction of pancreatic beta cells alone or in combination with insulin resistance. The replacement or regeneration of beta cells can effectively reverse diabetes in humans and rodents. Therefore, the identification of novel small molecules that promote pancreatic beta-cell proliferation is an attractive approach for diabetic therapy. While numerous hormones, small molecules, and growth factors are able to drive rodent beta cells to replicate, only a few small molecules have demonstrated the ability to stimulate human beta-cell proliferation. Hence, there is an urgent need for therapeutic agents that induce regeneration and expansion of adult human beta cells. Here, we describe a detailed protocol for coating chamber slides, culturing primary islets, performing islet cell disassociation, seeding cells on chamber slides, treating islet cells with compounds or infecting them with adenovirus, immunostaining of proliferation markers and imaging, and data analysis.
Collapse
Affiliation(s)
- Courtney A Ackeifi
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ethan A Swartz
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peng Wang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
21
|
Wang T, Pan W, Hu J, Zhang Z, Li G, Liang Y. Circular RNAs in Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1087:275-285. [PMID: 30259374 DOI: 10.1007/978-981-13-1426-1_22] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metabolic diseases include diabetes mellitus (DM), obesity, metabolic syndrome, and non-alcoholic fatty liver disease (NAFLD). Circular RNA is a new type of RNA that is different from traditional linear RNA and has a closed loop structure. However, the function of circular RNA is not yet well elucidated in metabolic diseases. Only a few studies have reported about the relationship between circular RNA and metabolic diseases such as DM and NAFLD. This chapter presents a brief review of epidemiology, pathophysiology, or treatment of DM and NAFLD and then discusses the relationship between circular RNA and DM or NAFLD. Besides, this chapter further provides an updated discussion of the most relevant discoveries regarding circular RNA and their potential applications in molecular diagnostics, nucleic acid therapy, and biomarkers.
Collapse
Affiliation(s)
- Tianhui Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, China
| | - Wen Pan
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Jun Hu
- Department of Pediatric Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongrong Zhang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yajun Liang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
22
|
Riddy DM, Delerive P, Summers RJ, Sexton PM, Langmead CJ. G Protein-Coupled Receptors Targeting Insulin Resistance, Obesity, and Type 2 Diabetes Mellitus. Pharmacol Rev 2018; 70:39-67. [PMID: 29233848 DOI: 10.1124/pr.117.014373] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/13/2017] [Indexed: 03/21/2025] Open
Abstract
G protein-coupled receptors (GPCRs) continue to be important discovery targets for the treatment of type 2 diabetes mellitus (T2DM). Many GPCRs are directly involved in the development of insulin resistance and β-cell dysfunction, and in the etiology of inflammation that can lead to obesity-induced T2DM. This review summarizes the current literature describing a number of well-validated GPCR targets, but also outlines several new and promising targets for drug discovery. We highlight the importance of understanding the role of these receptors in the disease pathology, and their basic pharmacology, which will pave the way to the development of novel pharmacological probes that will enable these targets to fulfill their promise for the treatment of these metabolic disorders.
Collapse
Affiliation(s)
- Darren M Riddy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Philippe Delerive
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| |
Collapse
|
23
|
Fan J, Wang Y, Liu L, Zhang H, Zhang F, Shi L, Yu M, Gao F, Xu Z. cTAGE5 deletion in pancreatic β cells impairs proinsulin trafficking and insulin biogenesis in mice. J Cell Biol 2017; 216:4153-4164. [PMID: 29133483 PMCID: PMC5716288 DOI: 10.1083/jcb.201705027] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/31/2017] [Accepted: 09/08/2017] [Indexed: 01/02/2023] Open
Abstract
In this study, Fan et al. show that cTAGE5 interacts with the v-SNARE Sec22b to regulate proinsulin processing and COPII-dependent trafficking from the ER to the Golgi, thereby influencing glucose tolerance. Proinsulin is synthesized in the endoplasmic reticulum (ER) in pancreatic β cells and transported to the Golgi apparatus for proper processing and secretion into plasma. Defects in insulin biogenesis may cause diabetes. However, the underlying mechanisms for proinsulin transport are still not fully understood. We show that β cell–specific deletion of cTAGE5, also known as Mea6, leads to increased ER stress, reduced insulin biogenesis in the pancreas, and severe glucose intolerance in mice. We reveal that cTAGE5/MEA6 interacts with vesicle membrane soluble N-ethyl-maleimide sensitive factor attachment protein receptor Sec22b. Sec22b and its interaction with cTAGE5/MEA6 are essential for proinsulin processing. cTAGE5/MEA6 may coordinate with Sec22b to control the release of COPII vesicles from the ER, and thereby the ER-to-Golgi trafficking of proinsulin. Importantly, transgenic expression of human cTAGE5/MEA6 in β cells can rescue not only the defect in islet structure, but also dysfunctional insulin biogenesis and glucose intolerance on cTAGE5/Mea6 conditional knockout background. Together our data provide more insight into the underlying mechanism of the proinsulin trafficking pathway.
Collapse
Affiliation(s)
- Junwan Fan
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yaqing Wang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Liang Liu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongsheng Zhang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Feng Zhang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lei Shi
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Mei Yu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China .,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
24
|
Mondal P, Prasad A, Girdhar K. Interventions to improve β-cell mass and function. ANNALES D'ENDOCRINOLOGIE 2017; 78:469-477. [PMID: 28870707 DOI: 10.1016/j.ando.2016.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/18/2016] [Accepted: 11/09/2016] [Indexed: 01/09/2023]
Abstract
Diabetes mellitus (T2DM) has become an epidemiologically important disease worldwide and is also becoming a great matter of concern due to the effects associated with it like: high morbidity, elevated health care cost and shortened life span. T2DM is a chronic metabolic disease characterized by insulin resistance as well as β-cell dysfunction. It is widely accepted that in the face of insulin resistance, euglycemia can be maintained by increase in pancreatic β-cell mass and insulin secretion. This compensation is largely due to enhanced secretion of insulin by the β-cell mass, which is present initially, and thereby subsequent increases in β-cell mass provide additional insulin secretion. However, the mechanism by which β-cell anatomical plasticity and functional plasticity for insulin secretion is coordinated and executed in different physiological and pathophysiological states is complex and has been poorly understood. As the incidence of T2DM continues to increase at an alarming rate, it is becoming imperative to shift the research focus towards the β-cell physiology where identification of novel pathways that influence the β-cell proliferation and/or contribute to increase insulin secretion has the potential to lead to new therapies for preventing or delaying onset of disease.
Collapse
Affiliation(s)
- Prosenjit Mondal
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India.
| | - Amit Prasad
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India
| | - Khyati Girdhar
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India
| |
Collapse
|
25
|
Chau GC, Im DU, Kang TM, Bae JM, Kim W, Pyo S, Moon EY, Um SH. mTOR controls ChREBP transcriptional activity and pancreatic β cell survival under diabetic stress. J Cell Biol 2017; 216:2091-2105. [PMID: 28606928 PMCID: PMC5496625 DOI: 10.1083/jcb.201701085] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/05/2017] [Accepted: 05/01/2017] [Indexed: 02/06/2023] Open
Abstract
Through in vivo analyses of mTOR deficiency and in vitro studies of human and mouse pancreatic islets, Chau et al. show that mTOR plays a critical role in β cell survival in diabetes. mTOR associates with and inhibits the transcriptional ChREBP–Mlx complex, suppressing TXNIP expression and β cell death. Impaired nutrient sensing and dysregulated glucose homeostasis are common in diabetes. However, how nutrient-sensitive signaling components control glucose homeostasis and β cell survival under diabetic stress is not well understood. Here, we show that mice lacking the core nutrient-sensitive signaling component mammalian target of rapamycin (mTOR) in β cells exhibit reduced β cell mass and smaller islets. mTOR deficiency leads to a severe reduction in β cell survival and increased mitochondrial oxidative stress in chemical-induced diabetes. Mechanistically, we find that mTOR associates with the carbohydrate-response element–binding protein (ChREBP)–Max-like protein complex and inhibits its transcriptional activity, leading to decreased expression of thioredoxin-interacting protein (TXNIP), a potent inducer of β cell death and oxidative stress. Consistent with this, the levels of TXNIP and ChREBP were highly elevated in human diabetic islets and mTOR-deficient mouse islets. Thus, our results suggest that a nutrient-sensitive mTOR-regulated transcriptional network could be a novel target to improve β cell survival and glucose homeostasis in diabetes.
Collapse
Affiliation(s)
- Gia Cac Chau
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, Korea
| | - Dong Uk Im
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| | - Tong Mook Kang
- Department of Physiology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, Korea .,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
26
|
Kim SY, Lee JH, Merrins MJ, Gavrilova O, Bisteau X, Kaldis P, Satin LS, Rane SG. Loss of Cyclin-dependent Kinase 2 in the Pancreas Links Primary β-Cell Dysfunction to Progressive Depletion of β-Cell Mass and Diabetes. J Biol Chem 2017; 292:3841-3853. [PMID: 28100774 DOI: 10.1074/jbc.m116.754077] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/13/2017] [Indexed: 11/06/2022] Open
Abstract
The failure of pancreatic islet β-cells is a major contributor to the etiology of type 2 diabetes. β-Cell dysfunction and declining β-cell mass are two mechanisms that contribute to this failure, although it is unclear whether they are molecularly linked. Here, we show that the cell cycle regulator, cyclin-dependent kinase 2 (CDK2), couples primary β-cell dysfunction to the progressive deterioration of β-cell mass in diabetes. Mice with pancreas-specific deletion of Cdk2 are glucose-intolerant, primarily due to defects in glucose-stimulated insulin secretion. Accompanying this loss of secretion are defects in β-cell metabolism and perturbed mitochondrial structure. Persistent insulin secretion defects culminate in progressive deficits in β-cell proliferation, reduced β-cell mass, and diabetes. These outcomes may be mediated directly by the loss of CDK2, which binds to and phosphorylates the transcription factor FOXO1 in a glucose-dependent manner. Further, we identified a requirement for CDK2 in the compensatory increases in β-cell mass that occur in response to age- and diet-induced stress. Thus, CDK2 serves as an important nexus linking primary β-cell dysfunction to progressive β-cell mass deterioration in diabetes.
Collapse
Affiliation(s)
- So Yoon Kim
- From the Cell Growth and Metabolism Section, Diabetes, Endocrinology, and Obesity Branch and
| | - Ji-Hyeon Lee
- From the Cell Growth and Metabolism Section, Diabetes, Endocrinology, and Obesity Branch and
| | - Matthew J Merrins
- the Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin, Madison, Wisconsin 53705
| | - Oksana Gavrilova
- the Mouse Metabolism Core Laboratory, NIDDK, National Institutes of Health, Clinical Research Center, Bethesda, Maryland 20892
| | - Xavier Bisteau
- the Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos#3-09, Singapore 138673, Singapore
| | - Philipp Kaldis
- the Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos#3-09, Singapore 138673, Singapore.,the Department of Biochemistry, National University of Singapore, Singapore 117597, Singapore, and
| | - Leslie S Satin
- the Department of Pharmacology and Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Sushil G Rane
- From the Cell Growth and Metabolism Section, Diabetes, Endocrinology, and Obesity Branch and
| |
Collapse
|
27
|
Tsukita S, Yamada T, Takahashi K, Munakata Y, Hosaka S, Takahashi H, Gao J, Shirai Y, Kodama S, Asai Y, Sugisawa T, Chiba Y, Kaneko K, Uno K, Sawada S, Imai J, Katagiri H. MicroRNAs 106b and 222 Improve Hyperglycemia in a Mouse Model of Insulin-Deficient Diabetes via Pancreatic β-Cell Proliferation. EBioMedicine 2016; 15:163-172. [PMID: 27974246 PMCID: PMC5233820 DOI: 10.1016/j.ebiom.2016.12.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/16/2016] [Accepted: 12/04/2016] [Indexed: 01/01/2023] Open
Abstract
Major symptoms of diabetes mellitus manifest, once pancreatic β-cell numbers have become inadequate. Although natural regeneration of β-cells after injury is very limited, bone marrow (BM) transplantation (BMT) promotes their regeneration through undetermined mechanism(s) involving inter-cellular (BM cell-to-β-cell) crosstalk. We found that two microRNAs (miRNAs) contribute to BMT-induced β-cell regeneration. Screening murine miRNAs in serum exosomes after BMT revealed 42 miRNAs to be increased. Two of these miRNAs (miR-106b-5p and miR-222-3p) were shown to be secreted by BM cells and increased in pancreatic islet cells after BMT. Treatment with the corresponding anti-miRNAs inhibited BMT-induced β-cell regeneration. Furthermore, intravenous administration of the corresponding miRNA mimics promoted post-injury β-cell proliferation through Cip/Kip family down-regulation, thereby ameliorating hyperglycemia in mice with insulin-deficient diabetes. Thus, these identified miRNAs may lead to the development of therapeutic strategies for diabetes. BMT regenerates β-cells in mice with STZ-induced diabetes and increases miR-106b and miR-222 in serum exosomes and islets. Inhibition with anti-miRs against these miRs suppresses BMT-induced β-cell regeneration. Injection of miR-106b and miR-222 mimics promotes β-cell proliferation and improves hyperglycemia in STZ-treated mice.
Regeneration of pancreatic β-cells is a promising therapeutic strategy not only for type 1 diabetes but also for certain forms of type 2 diabetes. However, natural regeneration of β-cells hardly ever occurs. Interestingly, bone marrow transplantation (BMT) has been shown to promote β-cell regeneration through an undetermined mechanism(s). In this study, we found that two microRNAs (miR-106b/-222) contribute to BMT-induced β-cell proliferation. Inhibition of miR-106b/-222 using specific anti-miRNAs significantly suppressed BMT-induced β-cell proliferation. Furthermore, intravenously administered miR-106b/222 promoted β-cell proliferation, thereby ameliorating hyperglycemia in mice with insulin-deficient diabetes. Thus, these identified miRNAs may lead to novel therapeutic strategies for diabetes.
Collapse
Affiliation(s)
- Sohei Tsukita
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan; Centre for Metabolic Diseases, Tohoku University Graduate School of Medicine, Japan.
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Yuichiro Munakata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Shinichiro Hosaka
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Hironobu Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Junhong Gao
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan; Tohoku University Frontier Research Institute for Interdisciplinary Science, Miyagi, Japan
| | - Yuta Shirai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Yoichiro Asai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Takashi Sugisawa
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Yumiko Chiba
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Kenji Uno
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Shojiro Sawada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan; Centre for Metabolic Diseases, Tohoku University Graduate School of Medicine, Japan; Japan Agency for Medical Research and Development, CREST, Japan
| |
Collapse
|
28
|
Assefa Z, Akbib S, Lavens A, Stangé G, Ling Z, Hellemans KH, Pipeleers D. Direct effect of glucocorticoids on glucose-activated adult rat β-cells increases their cell number and their functional mass for transplantation. Am J Physiol Endocrinol Metab 2016; 311:E698-E705. [PMID: 27555297 DOI: 10.1152/ajpendo.00070.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/17/2016] [Indexed: 01/02/2023]
Abstract
Compounds that increase β-cell number can serve as β-cell replacement therapies in diabetes. In vitro studies have identified several agents that can activate DNA synthesis in primary β-cells but only in small percentages of cells and without demonstration of increases in cell number. We used whole well multiparameter imaging to first screen a library of 1,280 compounds for their ability to recruit adult rat β-cells into DNA synthesis and then assessed influences of stimulatory agents on the number of living cells. The four compounds with highest β-cell recruitment were glucocorticoid (GC) receptor ligands. The GC effect occurred in glucose-activated β-cells and was associated with increased glucose utilization and oxidation. Hydrocortisone and methylprednisolone almost doubled the number of β-cells in 2 wk. The expanded cell population provided an increased functional β-cell mass for transplantation in diabetic animals. These effects are age dependent; they did not occur in neonatal rat β-cells, where GC exposure suppressed basal replication and was cytotoxic. We concluded that GCs can induce the replication of adult rat β-cells through a direct action, with intercellular differences in responsiveness that have been related to differences in glucose activation and in age. These influences can explain variability in GC-induced activation of DNA synthesis in rat and human β-cells. Our study also demonstrated that β-cells can be expanded in vitro to increase the size of metabolically adequate grafts.
Collapse
Affiliation(s)
- Zerihun Assefa
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Sarah Akbib
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Astrid Lavens
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Geert Stangé
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Zhidong Ling
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Karine H Hellemans
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Daniel Pipeleers
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| |
Collapse
|
29
|
Yang Z, Zhang Q, Lu Q, Jia Z, Chen P, Ma K, Wang W, Zhou C. ISL-1 promotes pancreatic islet cell proliferation by forming an ISL-1/Set7/9/PDX-1 complex. Cell Cycle 2016; 14:3820-9. [PMID: 26176407 PMCID: PMC4825718 DOI: 10.1080/15384101.2015.1069926] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Islet-1 (ISL-1), a LIM-homeodomain transcription factor, has been recently found to be essential for promoting postnatal pancreatic islet proliferation. However, the detailed mechanism has not yet been elucidated. In the present study, we investigated the mechanism by which ISL-1 promotes β-cell proliferation through regulation of CyclinD1 in HIT-T15 and NIT-1 cells, as well in rat islet mass. Our results provide the evidence that ISL-1 promotes adult pancreatic islet β-cell proliferation by activating CyclinD1 transcription through cooperation with Set7/9 and PDX-1 to form an ISL-1/Set7/9/PDX-1 complex. This complex functions in an ISL-1-dependent manner, with Set7/9 functioning not only as a histone methyltransferase, which increases the histone H3K4 tri-methylation of the CyclinD1 promoter region, but also an adaptor to bridge ISL-1 and PDX-1, while PDX-1 functions as a RNA pol II binding modulator. Furthermore, the formation of the ISL-1/Set7/9/PDX-1 complex is positively associated with insulin-like growth factor-1 treatment in NIT and HIT-T15 cells in vitro, while may be negatively correlated with age in vivo.
Collapse
Affiliation(s)
- Zhe Yang
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Qiao Zhang
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China.,b Current address: Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Kunming Medical University ; Kunming , China
| | - Qin Lu
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Zhuqing Jia
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Ping Chen
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Kangtao Ma
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Weiping Wang
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Chunyan Zhou
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| |
Collapse
|
30
|
Guan B, Li W, Li F, Xie Y, Ni Q, Gu Y, Li X, Wang Q, Zhang H, Ning G. Sfrp5 mediates glucose-induced proliferation in rat pancreatic β-cells. J Endocrinol 2016; 229:73-83. [PMID: 26869333 DOI: 10.1530/joe-15-0535] [Citation(s) in RCA: 13] [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: 02/07/2016] [Accepted: 02/11/2016] [Indexed: 01/07/2023]
Abstract
The cellular and molecular mechanisms of glucose-stimulated β-cell proliferation are poorly understood. Recently, secreted frizzled-related protein 5 (encoded by Sfrp5; a Wnt signaling inhibitor) has been demonstrated to be involved in β-cell proliferation in obesity. A previous study demonstrated that glucose enhanced Wnt signaling to promote cell proliferation. We hypothesized that inhibition of SFRP5 contributes to glucose-stimulated β-cell proliferation. In this study, we found that the Sfrp5 level was significantly reduced in high glucose-treated INS-1 cells, primary rat β-cells, and islets isolated from glucose-infused rats. Overexpression of SFRP5 diminished glucose-stimulated proliferation in both INS-1 cells and primary β-cells, with a concomitant inhibition of the Wnt signaling pathway and decreased cyclin D2 expression. In addition, we showed that glucose-induced Sfrp5 suppression was modulated by the PI3K/AKT pathway. Therefore, we conclude that glucose inhibits Sfrp5 expression via the PI3K/AKT pathway and hence promotes rat pancreatic β-cell proliferation.
Collapse
Affiliation(s)
- Binbin Guan
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China Department of EndocrinologyFuJian Union hospital, Fuzhou, P R China
| | - Wenyi Li
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Fengying Li
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Yun Xie
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Qicheng Ni
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Yanyun Gu
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Xiaoying Li
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Qidi Wang
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Hongli Zhang
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Guang Ning
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| |
Collapse
|
31
|
Abstract
The mechanisms or causes of pancreatic β-cell death as well as impaired insulin secretion, which are the principal events of diabetic etiopathology, are largely unknown. Diabetic complications are known to be associated with abnormal plasma lipid profile, mainly elevated level of cholesterol and free fatty acids. However, in recent years, elevated plasma cholesterol has been implicated as a primary modulator of pancreatic β-cell functions as well as death. High-cholesterol diet in animal models or excess cholesterol in pancreatic β-cell causes transporter desensitization and results in morphometric changes in insulin granules. Moreover, cholesterol is also held responsible to cause oxidative stress, mitochondrial dysfunction, and activation of proapoptotic markers leading to β-cell death. The present review focuses on the pathways and molecularevents that occur in the β-cell under the influence of excess cholesterol that hampers the basal physiology of the cell leading to the progression of diabetes.
Collapse
|
32
|
El-Badawy A, El-Badri N. The cell cycle as a brake for β-cell regeneration from embryonic stem cells. Stem Cell Res Ther 2016; 7:9. [PMID: 26759123 PMCID: PMC4711007 DOI: 10.1186/s13287-015-0274-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The generation of insulin-producing β cells from stem cells in vitro provides a promising source of cells for cell transplantation therapy in diabetes. However, insulin-producing cells generated from human stem cells show deficiency in many functional characteristics compared with pancreatic β cells. Recent reports have shown molecular ties between the cell cycle and the differentiation mechanism of embryonic stem (ES) cells, assuming that cell fate decisions are controlled by the cell cycle machinery. Both β cells and ES cells possess unique cell cycle machinery yet with significant contrasts. In this review, we compare the cell cycle control mechanisms in both ES cells and β cells, and highlight the fundamental differences between pluripotent cells of embryonic origin and differentiated β cells. Through critical analysis of the differences of the cell cycle between these two cell types, we propose that the cell cycle of ES cells may act as a brake for β-cell regeneration. Based on these differences, we discuss the potential of modulating the cell cycle of ES cells for the large-scale generation of functionally mature β cells in vitro. Further understanding of the factors that modulate the ES cell cycle will lead to new approaches to enhance the production of functional mature insulin-producing cells, and yield a reliable system to generate bona fide β cells in vitro.
Collapse
Affiliation(s)
- Ahmed El-Badawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt.
| |
Collapse
|
33
|
Zhou Y, Hu Q, Chen F, Zhang J, Guo J, Wang H, Gu J, Ma L, Ho G. Human umbilical cord matrix-derived stem cells exert trophic effects on β-cell survival in diabetic rats and isolated islets. Dis Model Mech 2015; 8:1625-33. [PMID: 26398949 PMCID: PMC4728317 DOI: 10.1242/dmm.021857] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/11/2015] [Indexed: 02/05/2023] Open
Abstract
Human umbilical cord matrix-derived stem cells (uMSCs), owing to their cellular and procurement advantages compared with mesenchymal stem cells derived from other tissue sources, are in clinical trials to treat type 1 (T1D) and type 2 diabetes (T2D). However, the therapeutic basis remains to be fully understood. The immunomodulatory property of uMSCs could explain the use in treating T1D; however, the mere immune modulation might not be sufficient to support the use in T2D. We thus tested whether uMSCs could exert direct trophic effects on β-cells. Infusion of uMSCs into chemically induced diabetic rats prevented hyperglycemic progression with a parallel preservation of islet size and cellularity, demonstrating the protective effect of uMSCs on β-cells. Mechanistic analyses revealed that uMSCs engrafted long-term in the injured pancreas and the engraftment markedly activated the pancreatic PI3K pathway and its downstream anti-apoptotic machinery. The pro-survival pathway activation was associated with the expression and secretion of β-cell growth factors by uMSCs, among which insulin-like growth factor 1 (IGF1) was highly abundant. To establish the causal relationship between the uMSC-secreted factors and β-cell survival, isolated rat islets were co-cultured with uMSCs in the transwell system. Co-culturing improved the islet viability and insulin secretion. Furthermore, reduction of uMSC-secreted IGF1 via siRNA knockdown diminished the protective effects on islets in the co-culture. Thus, our data support a model whereby uMSCs exert trophic effects on islets by secreting β-cell growth factors such as IGF1. The study reveals a novel therapeutic role of uMSCs and suggests that multiple mechanisms are employed by uMSCs to treat diabetes.
Collapse
Affiliation(s)
- Yunting Zhou
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Qi Hu
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Fuyi Chen
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Juan Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jincheng Guo
- Department of Molecular Pathology, Shantou University Medical College, Shantou 515041, China
| | - Hongwu Wang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jiang Gu
- Department of Molecular Pathology, Shantou University Medical College, Shantou 515041, China
| | - Lian Ma
- Department of Pediatrics, The Women and Children's Hospital of Shenzhen University, Shenzhen 518122, China
| | - Guyu Ho
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
34
|
Mohan R, Mao Y, Zhang S, Zhang YW, Xu CR, Gradwohl G, Tang X. Differentially Expressed MicroRNA-483 Confers Distinct Functions in Pancreatic β- and α-Cells. J Biol Chem 2015; 290:19955-66. [PMID: 26109062 DOI: 10.1074/jbc.m115.650705] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Indexed: 01/08/2023] Open
Abstract
Insulin secreted from pancreatic β-cells and glucagon secreted from pancreatic α-cells are the two major hormones working in the pancreas in an opposing manner to regulate and maintain a normal glucose homeostasis. How microRNAs (miRNAs), a population of non-coding RNAs so far demonstrated to be differentially expressed in various types of cells, regulate gene expression in pancreatic β-cells and its closely associated α-cells is not completely clear. In this study, miRNA profiling was performed and compared between pancreatic β-cells and their partner α-cells. One novel miRNA, miR-483, was identified for its highly differential expression in pancreatic β-cells when compared to its expression in α-cells. Overexpression of miR-483 in β-cells increased insulin transcription and secretion by targeting SOCS3, a member of suppressor of cytokine signaling family. In contrast, overexpression of miR-483 decreased glucagon transcription and secretion in α-cells. Moreover, overexpressed miR-483 protected against proinflammatory cytokine-induced apoptosis in β-cells. This correlates with a higher expression level of miR-483 and the expanded β-cell mass observed in the islets of prediabetic db/db mice. Together, our data suggest that miR-483 has opposite effects in α- and β-cells by targeting SOCS3, and the imbalance of miR-483 and its targets may play a crucial role in diabetes pathogenesis.
Collapse
Affiliation(s)
- Ramkumar Mohan
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931
| | - Yiping Mao
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931
| | - Shungang Zhang
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931
| | - Yu-Wei Zhang
- the College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China, and
| | - Cheng-Ran Xu
- the College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China, and
| | - Gérard Gradwohl
- the Institute of Genetics and Molecular and Cellular Biology, Department of Development and Stem cells, University of Strasbourg, 67404 Illkirch, France
| | - Xiaoqing Tang
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931,
| |
Collapse
|
35
|
Li Q, Lai ZC. Recent progress in studies of factors that elicit pancreatic β-cell expansion. Protein Cell 2015; 6:81-7. [PMID: 25492376 PMCID: PMC4312764 DOI: 10.1007/s13238-014-0123-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 11/13/2014] [Indexed: 12/26/2022] Open
Abstract
The loss of or decreased functional pancreatic β-cell is a major cause of type 1 and type 2 diabetes. Previous studies have shown that adult β-cells can maintain their ability for a low level of turnover through replication and neogenesis. Thus, a strategy to prevent and treat diabetes would be to enhance the ability of β-cells to increase the mass of functional β-cells. Consequently, much effort has been devoted to identify factors that can effectively induce β-cell expansion. This review focuses on recent reports on small molecules and protein factors that have been shown to promote β-cell expansion.
Collapse
Affiliation(s)
- Qiu Li
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021 China
| | - Zhi-Chun Lai
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021 China
- Department of Biology, The Pennsylvania State University, University Park, PA 16802 USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802 USA
| |
Collapse
|
36
|
Yang Y, Chan L. Gene Therapy for Diabetes. TRANSLATING GENE THERAPY TO THE CLINIC 2015:115-128. [DOI: 10.1016/b978-0-12-800563-7.00008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
37
|
da Silva SRM, Perrone GC, Dinis JM, de Almeida RMC. Reproducibility enhancement and differential expression of non predefined functional gene sets in human genome. BMC Genomics 2014; 15:1181. [PMID: 25539829 PMCID: PMC4326474 DOI: 10.1186/1471-2164-15-1181] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 12/11/2014] [Indexed: 12/27/2022] Open
Abstract
Background Transcriptogram profiling is a method to present and analyze transcription data in a genome-wide scale that reduces noise and facilitates biological interpretation. An ordered gene list is produced, such that the probability that the genes are functionally associated exponentially decays with their distance on the list. This list presents a biological logic, evinced by the selective enrichment of successive intervals with Gene Ontology terms or KEGG pathways. Transcriptograms are expression profiles obtained by taking the average of gene expression over neighboring genes on this list. Transcriptograms enhance reproducibility and precision for expression measurements of functionally correlated gene sets. Results Here we present an ordering list for Homo sapiens and apply the transcriptogram profiling method to different datasets. We show that this method enhances experiment reproducibility and enhances signal. We applied the method to a diabetes study by Hwang and collaborators, which focused on expression differences between cybrids produced by the hybridization of mitochondria of diabetes mellitus donors with osteosarcoma cell lines, depleted of mitochondria. We found that the transcriptogram method revealed significant differential expression in gene sets linked to blood coagulation and wound healing pathways, and also to gene sets that do not represent any metabolic pathway or Gene Ontology term. These gene sets are connected to ECM-receptor interaction and secreted proteins. Conclusion The transcriptogram profiling method provided an automatic way to define sets of genes with correlated expression, reduce noise in genome-wide transcription profiles, and enhance measure reproducibility and sensitivity. These advantages enabled biologic interpretation and pointed to differentially expressed gene sets in diabetes mellitus which were not previously defined. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-1181) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Rita M C de Almeida
- Instituto de Física, Universidade Federal do Rio Grande do Sul, Av, Bento Gonçalves, 9500, 91501-970 Porto Alegre, RS, Brazil.
| |
Collapse
|
38
|
Masjkur J, Arps-Forker C, Poser SW, Nikolakopoulou P, Toutouna L, Chenna R, Chavakis T, Chatzigeorgiou A, Chen LS, Dubrovska A, Choudhary P, Uphues I, Mark M, Bornstein SR, Androutsellis-Theotokis A. Hes3 is expressed in the adult pancreatic islet and regulates gene expression, cell growth, and insulin release. J Biol Chem 2014; 289:35503-35516. [PMID: 25371201 PMCID: PMC4271235 DOI: 10.1074/jbc.m114.590687] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 10/31/2014] [Indexed: 12/16/2022] Open
Abstract
The transcription factor Hes3 is a component of a signaling pathway that supports the growth of neural stem cells with profound consequences in neurodegenerative disease models. Here we explored whether Hes3 also regulates pancreatic islet cells. We showed that Hes3 is expressed in human and rodent pancreatic islets. In mouse islets it co-localizes with alpha and beta cell markers. We employed the mouse insulinoma cell line MIN6 to perform in vitro characterization and functional studies in conditions known to modulate Hes3 based upon our previous work using neural stem cell cultures. In these conditions, cells showed elevated Hes3 expression and nuclear localization, grew efficiently, and showed higher evoked insulin release responses, compared with serum-containing conditions. They also exhibited higher expression of the transcription factor Pdx1 and insulin. Furthermore, they were responsive to pharmacological treatments with the GLP-1 analog Exendin-4, which increased nuclear Hes3 localization. We employed a transfection approach to address specific functions of Hes3. Hes3 RNA interference opposed cell growth and affected gene expression as revealed by DNA microarrays. Western blotting and PCR approaches specifically showed that Hes3 RNA interference opposes the expression of Pdx1 and insulin. Hes3 overexpression (using a Hes3-GFP fusion construct) confirmed a role of Hes3 in regulating Pdx1 expression. Hes3 RNA interference reduced evoked insulin release. Mice lacking Hes3 exhibited increased islet damage by streptozotocin. These data suggest roles of Hes3 in pancreatic islet function.
Collapse
Affiliation(s)
| | | | | | | | | | - Ramu Chenna
- the Applied Bioinformatics Group, BioInnovations Zentrum, University of Dresden, 01307 Dresden, Germany
| | - Triantafyllos Chavakis
- the Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and
| | - Antonios Chatzigeorgiou
- the Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and
| | - Lan-Sun Chen
- the Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and
| | - Anna Dubrovska
- Department of Medicine, OncoRay National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Pratik Choudhary
- the Diabetes Research Group, King's College London, London SE5 9RS, United Kingdom
| | - Ingo Uphues
- the Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach, Germany, and
| | - Michael Mark
- the Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach, Germany, and
| | | | | |
Collapse
|
39
|
Ma X, Guan Y, Hua X. Glucagon-like peptide 1-potentiated insulin secretion and proliferation of pancreatic β-cells. J Diabetes 2014; 6:394-402. [PMID: 24725840 DOI: 10.1111/1753-0407.12161] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/28/2014] [Accepted: 04/07/2014] [Indexed: 02/06/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is the primary incretin hormone secreted from the intestine upon uptake of food to stimulate insulin secretion from pancreatic β-cells. GLP-1 exerts its effects by binding to its G-protein coupled receptors and subsequently activating adenylate cyclase, leading to generation of cyclic adenosine monophosphate (cAMP). cAMP stimulates insulin secretion via activation of its effectors PKA and Epac2 in pancreatic β-cells. In addition to its insulinotropic effects, GLP-1 also preserves pancreatic β-cell mass by stimulating β-cell proliferation. Unlike the action of sulphonylureas in lowering blood glucose levels, action of GLP-1 is affected by and interplays with glucose levels. Due to such advantages, GLP-1-based therapeutics have been rapidly developed and used clinically for treatment of type 2 diabetes. However, molecular mechanisms underlying how GLP-1 potentiates diminished glucose-stimulated insulin secretion and β-cell proliferation under diabetic conditions are not well understood. Here, we review the actions of GLP-1 in regulation of insulin secretion and pancreatic β-cell proliferation.
Collapse
Affiliation(s)
- Xiaosong Ma
- Shenzhen University Diabetes Center, Shenzhen, China
| | | | | |
Collapse
|
40
|
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
|
41
|
Schvartz D, Couté Y, Sanchez JC. Quantitative proteomics reveals the link between minichromosome maintenance complex and glucose-induced proliferation of rat pancreatic INS-1E β-cells. J Proteomics 2014; 108:163-70. [DOI: 10.1016/j.jprot.2014.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/16/2014] [Accepted: 05/19/2014] [Indexed: 12/21/2022]
|
42
|
Sun Y, Zhang Y, Li N, Zhang H, Zhou L, Shao L. Exposure to high levels of glucose increases the expression levels of genes involved in cholesterol biosynthesis in rat islets. Exp Ther Med 2014; 8:991-997. [PMID: 25120636 PMCID: PMC4113575 DOI: 10.3892/etm.2014.1812] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 06/05/2014] [Indexed: 02/01/2023] Open
Abstract
Cells continually adjust their gene expression profiles in order to adapt to the availability of nutrients. Glucose is a major regulator of pxancreatic β-cell function and cell growth. However, the mechanism of β-cell adaptation to high levels of glucose remains uncertain. To identify the specific targets responsible for adaptation to high levels of glucose, the differentially expressed genes from primary rat islets treated with 3.3 and 16.7 mmol/l glucose for 24 h were detected by DNA microarray. The results revealed that the expression levels of genes that encode enzymes required for de novo cholesterol biosynthesis [3-hydroxy-3-methylglutaryl-CoA synthase 1 (Hmgcs1), 3-hydroxy-3-methylglutaryl-CoA reductase (Hmgcr), mevalonate (diphospho) decarboxylase (Mvd), isopentenyl-diphosphate δ-isomerase 1 (Idi1), squalene epoxidase (Sqle) and 7-dehydrocholesterol reductase (Dhcr7)] were significantly increased in islets treated with high levels of glucose compared with those in the islets treated with lower glucose levels. Quantitative polymerase chain reaction further confirmed that glucose stimulated the expression levels of these genes in a dose- and time-dependent manner. A similar result was obtained in islets isolated from rats subjected to 12, 24, 48 and 72 h of continuous glucose infusion. It has previously been recognized that cholesterol homeostasis is important for β-cell function. The present study provides, to the best of our knowledge, the first evidence for the involvement of the de novo cholesterol biosynthesis pathway in the adaptation of rat islets to high levels of glucose in vitro and in vivo.
Collapse
Affiliation(s)
- Yixuan Sun
- Department of Geratology, East Hospital, Shanghai Tongji University, Shanghai 200120, P.R. China
| | - Yuqing Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Na Li
- Department of Gastroenterology, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| | - Hua Zhang
- Department of Geratology, East Hospital, Shanghai Tongji University, Shanghai 200120, P.R. China
| | - Libin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Li Shao
- Department of Geratology, East Hospital, Shanghai Tongji University, Shanghai 200120, P.R. China
| |
Collapse
|
43
|
Wang R, Munoz EE, Zhu S, McGrath BC, Cavener DR. Perk gene dosage regulates glucose homeostasis by modulating pancreatic β-cell functions. PLoS One 2014; 9:e99684. [PMID: 24915520 PMCID: PMC4051701 DOI: 10.1371/journal.pone.0099684] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 05/16/2014] [Indexed: 11/18/2022] Open
Abstract
Background Insulin synthesis and cell proliferation are under tight regulation in pancreatic β-cells to maintain glucose homeostasis. Dysfunction in either aspect leads to development of diabetes. PERK (EIF2AK3) loss of function mutations in humans and mice exhibit permanent neonatal diabetes that is characterized by insufficient β-cell mass and reduced proinsulin trafficking and insulin secretion. Unexpectedly, we found that Perk heterozygous mice displayed lower blood glucose levels. Methodology Longitudinal studies were conducted to assess serum glucose and insulin, intracellular insulin synthesis and storage, insulin secretion, and β-cell proliferation in Perk heterozygous mice. In addition, modulation of Perk dosage specifically in β-cells showed that the glucose homeostasis phenotype of Perk heterozygous mice is determined by reduced expression of PERK in the β-cells. Principal Findings We found that Perk heterozygous mice first exhibited enhanced insulin synthesis and secretion during neonatal and juvenile development followed by enhanced β-cell proliferation and a substantial increase in β-cell mass at the adult stage. These differences are not likely to entail the well-known function of PERK to regulate the ER stress response in cultured cells as several markers for ER stress were not differentially expressed in Perk heterozygous mice. Conclusions In addition to the essential functions of PERK in β-cells as revealed by severely diabetic phenotype in humans and mice completely deficient for PERK, reducing Perk gene expression by half showed that intermediate levels of PERK have a profound impact on β-cell functions and glucose homeostasis. These results suggest that an optimal level of PERK expression is necessary to balance several parameters of β-cell function and growth in order to achieve normoglycemia.
Collapse
Affiliation(s)
- Rong Wang
- The Pennsylvania State University, Department of Biology, Center of Cellular Dynamics, University Park, Pennsylvania, United States of America
| | - Elyse E. Munoz
- The Pennsylvania State University, Department of Biology, Center of Cellular Dynamics, University Park, Pennsylvania, United States of America
| | - Siying Zhu
- The Pennsylvania State University, Department of Biology, Center of Cellular Dynamics, University Park, Pennsylvania, United States of America
| | - Barbara C. McGrath
- The Pennsylvania State University, Department of Biology, Center of Cellular Dynamics, University Park, Pennsylvania, United States of America
| | - Douglas R. Cavener
- The Pennsylvania State University, Department of Biology, Center of Cellular Dynamics, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
44
|
Jiao Y, Le Lay J, Yu M, Naji A, Kaestner KH. Elevated mouse hepatic betatrophin expression does not increase human β-cell replication in the transplant setting. Diabetes 2014; 63:1283-8. [PMID: 24353178 PMCID: PMC3964501 DOI: 10.2337/db13-1435] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The recent discovery of betatrophin, a protein secreted by the liver and white adipose tissue in conditions of insulin resistance and shown to dramatically stimulate replication of mouse insulin-producing β-cells, has raised high hopes for the rapid development of a novel therapeutic approach for the treatment of diabetes. At present, however, the effects of betatrophin on human β-cells are not known. Here we use administration of the insulin receptor antagonist S961, shown to increase betatrophin gene expression and stimulate β-cell replication in mice, to test its effect on human β-cells. Although mouse β-cells, in their normal location in the pancreas or when transplanted under the kidney capsule, respond with a dramatic increase in β-cell DNA replication, human β-cells are completely unresponsive. These results put into question whether betatrophin can be developed as a therapeutic approach for treating human diabetes.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - John Le Lay
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ming Yu
- Department of Surgery and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ali Naji
- Department of Surgery and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Klaus H. Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Corresponding author: Klaus H. Kaestner,
| |
Collapse
|
45
|
Scharfmann R, Pechberty S, Hazhouz Y, von Bülow M, Bricout-Neveu E, Grenier-Godard M, Guez F, Rachdi L, Lohmann M, Czernichow P, Ravassard P. Development of a conditionally immortalized human pancreatic β cell line. J Clin Invest 2014; 124:2087-98. [PMID: 24667639 DOI: 10.1172/jci72674] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/22/2014] [Indexed: 12/25/2022] Open
Abstract
Diabetic patients exhibit a reduction in β cells, which secrete insulin to help regulate glucose homeostasis; however, little is known about the factors that regulate proliferation of these cells in human pancreas. Access to primary human β cells is limited and a challenge for both functional studies and drug discovery progress. We previously reported the generation of a human β cell line (EndoC-βH1) that was generated from human fetal pancreas by targeted oncogenesis followed by in vivo cell differentiation in mice. EndoC-βH1 cells display many functional properties of adult β cells, including expression of β cell markers and insulin secretion following glucose stimulation; however, unlike primary β cells, EndoC-βH1 cells continuously proliferate. Here, we devised a strategy to generate conditionally immortalized human β cell lines based on Cre-mediated excision of the immortalizing transgenes. The resulting cell line (EndoC-βH2) could be massively amplified in vitro. After expansion, transgenes were efficiently excised upon Cre expression, leading to an arrest of cell proliferation and pronounced enhancement of β cell-specific features such as insulin expression, content, and secretion. Our data indicate that excised EndoC-βH2 cells are highly representative of human β cells and should be a valuable tool for further analysis of human β cells.
Collapse
|
46
|
Alismail H, Jin S. Microenvironmental stimuli for proliferation of functional islet β-cells. Cell Biosci 2014; 4:12. [PMID: 24594290 PMCID: PMC3974598 DOI: 10.1186/2045-3701-4-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/29/2014] [Indexed: 12/31/2022] Open
Abstract
Diabetes is characterized by high blood glucose level due to either autoimmune destruction of islet β-cells or insufficient insulin secretion or glucose non-responsive production of insulin by β-cells. It is highly desired to replace biological functional β-cells for the treatment of diabetes. Unfortunately, β-cells proliferate with an extremely low rate. This cellular property hinders cell-based therapy for clinical application. Many attempts have been made to develop techniques that allow production of large quantities of clinically relevant islet β-cells in vitro. A line of studies evidently demonstrate that β-cells can proliferate under certain circumstances, giving the hopes for generating and expanding these cells in vitro and transplanting them to the recipient. In this review, we discuss the requirements of microenvironmental stimuli that stimulate β-cell proliferation in cell cultures. We highlight advanced approaches for augmentation of β-cell expansion that have recently emerged in this field. Furthermore, knowing the signaling pathways and molecular mechanisms would enable manipulating cell proliferation and optimizing its insulin secretory function. Thus, signaling pathways involved in the enhancement of cell proliferation are discussed as well.
Collapse
Affiliation(s)
| | - Sha Jin
- Department of Bioengineering, Thomas J, Watson School of Engineering and Applied Sciences, State University of New York in Binghamton, Binghamton, NY 13902, USA.
| |
Collapse
|
47
|
Vetere A, Choudhary A, Burns SM, Wagner BK. Targeting the pancreatic β-cell to treat diabetes. Nat Rev Drug Discov 2014; 13:278-89. [PMID: 24525781 DOI: 10.1038/nrd4231] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes is a leading cause of morbidity and mortality worldwide, and predicted to affect over 500 million people by 2030. However, this growing burden of disease has not been met with a comparable expansion in therapeutic options. The appreciation of the pancreatic β-cell as a central player in the pathogenesis of both type 1 and type 2 diabetes has renewed focus on ways to improve glucose homeostasis by preserving, expanding and improving the function of this key cell type. Here, we provide an overview of the latest developments in this field, with an emphasis on the most promising strategies identified to date for treating diabetes by targeting the β-cell.
Collapse
Affiliation(s)
- Amedeo Vetere
- Chemical Biology Program, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Amit Choudhary
- 1] Chemical Biology Program, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA. [2] Society of Fellows, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Sean M Burns
- Medical & Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Bridget K Wagner
- Chemical Biology Program, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
48
|
Assefa Z, Lavens A, Steyaert C, Stangé G, Martens GA, Ling Z, Hellemans K, Pipeleers D. Glucose regulates rat beta cell number through age-dependent effects on beta cell survival and proliferation. PLoS One 2014; 9:e85174. [PMID: 24416358 PMCID: PMC3887027 DOI: 10.1371/journal.pone.0085174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 11/24/2013] [Indexed: 11/22/2022] Open
Abstract
Background Glucose effects on beta cell survival and DNA-synthesis suggest a role as regulator of beta cell mass but data on beta cell numbers are lacking. We examined outcome of these influences on the number of beta cells isolated at different growth stages in their population. Methods Beta cells from neonatal, young-adult and old rats were cultured serum-free for 15 days. Their number was counted by automated whole-well imaging distinguishing influences on cell survival and on proliferative activity. Results Elevated glucose (10–20 versus 5 mmol/l) increased the number of living beta cells from 8-week rats to 30%, following a time- and concentration-dependent recruitment of quiescent cells into DNA-synthesis; a glucokinase-activator lowered the threshold but did not raise total numbers of glucose-recruitable cells. No glucose-induced increase occurred in beta cells from 40-week rats. Neonatal beta cells doubled in number at 5 mmol/l involving a larger activated fraction that did not increase at higher concentrations; however, their higher susceptibility to glucose toxicity at 20 mmol/l resulted in 20% lower living cell numbers than at start. None of the age groups exhibited a repetitively proliferating subpopulation. Conclusions Chronically elevated glucose levels increased the number of beta cells from young-adult but not from old rats; they interfered with expansion of neonatal beta cells and reduced their number. These effects are attributed to age-dependent differences in basal and glucose-induced proliferative activity and in cellular susceptibility to glucose toxicity. They also reflect age-dependent variations in the functional heterogeneity of the rat beta cell population.
Collapse
Affiliation(s)
- Zerihun Assefa
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| | - Astrid Lavens
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| | - Christophe Steyaert
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| | - Geert Stangé
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| | - Geert A Martens
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| | - Zhidong Ling
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| | - Karine Hellemans
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| | - Daniel Pipeleers
- Diabetes Pathology and Therapy Unit, Diabetes Research Center and Center for Beta Cell Therapy, Brussels Free University-VUB, Brussels, Belgium
| |
Collapse
|
49
|
Abstract
Evidence supports a relationship between the neuroendocrine and the immune systems. Data from mice that overexpress or are deficient in growth hormone (GH) indicate that GH stimulates T and B-cell proliferation and Ig synthesis, and enhances maturation of myeloid progenitor cells. The effect of GH on autoimmune pathologies has nonetheless been little studied. Using a murine model of type 1 diabetes, a T-cell-mediated autoimmune disease characterized by immune cell infiltration of pancreatic islets and destruction of insulin-producing β-cells, we observed that sustained GH expression reduced prodromal disease symptoms and eliminated progression to overt diabetes. The effect involves several GH-mediated mechanisms; GH altered the cytokine environment, triggered anti-inflammatory macrophage (M2) polarization, maintained activity of the suppressor T-cell population, and limited Th17 cell plasticity. In addition, GH reduced apoptosis and/or increased the proliferative rate of β-cells. These results support a role for GH in immune response regulation and identify a unique target for therapeutic intervention in type 1 diabetes.
Collapse
|
50
|
Villani V, Milanesi A, Sedrakyan S, Da Sacco S, Angelow S, Conconi MT, Di Liddo R, De Filippo R, Perin L. Amniotic fluid stem cells prevent β-cell injury. Cytotherapy 2013; 16:41-55. [PMID: 24210784 DOI: 10.1016/j.jcyt.2013.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 08/19/2013] [Accepted: 08/25/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND AIMS The contribution of amniotic fluid stem cells (AFSC) to tissue protection and regeneration in models of acute and chronic kidney injuries and lung failure has been shown in recent years. In the present study, we used a chemically induced mouse model of type 1 diabetes to determine whether AFSC could play a role in modulating β-cell injury and restoring β-cell function. METHODS Streptozotocin-induced diabetic mice were given intracardial injection of AFSC; morphological and physiological parameters and gene expression profile for the insulin pathway were evaluated after cell transplantation. RESULTS AFSC injection resulted in protection from β-cell damage and increased β-cell regeneration in a subset of mice as indicated by glucose and insulin levels, increased islet mass and preservation of islet structure. Moreover, β-cell preservation/regeneration correlated with activation of the insulin receptor/Pi3K/Akt signaling pathway and vascular endothelial growth factor-A expression involved in maintaining β-cell mass and function. CONCLUSIONS Our results suggest a therapeutic role for AFSC in preserving and promoting endogenous β-cell functionality and proliferation. The protective role of AFSC is evident when stem cell transplantation is performed before severe hyperglycemia occurs, which suggests the importance of early intervention. The present study demonstrates the possible benefits of the application of a non-genetically engineered stem cell population derived from amniotic fluid for the treatment of type 1 diabetes mellitus and gives new insight on the mechanism by which the beneficial effect is achieved.
Collapse
Affiliation(s)
- Valentina Villani
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Anna Milanesi
- Division of Endocrinology, VA Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California
| | - Sargis Sedrakyan
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Stefano Da Sacco
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Susanne Angelow
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | | | - Rosa Di Liddo
- Department of Pharmaceutical Sciences, University of Padua, Padua, Italy
| | - Roger De Filippo
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Laura Perin
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California.
| |
Collapse
|