1
|
Mullooly N, Smith DM, Gianni D. A multi-parametric high throughput assay for detecting beta-cell proliferation in dispersed primary islets. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:3-12. [PMID: 36646173 DOI: 10.1016/j.slasd.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/11/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Identification of novel compounds to selectively induce pancreatic beta-cell proliferation has the potential to restore functional beta-cell mass and insulin secretory demand in type 2 diabetes. The rarity of islet cell clusters (comprising of only 1% of the total pancreas mass) makes such a discovery a challenge. To address this obstacle a high throughput, 384 well, plate-based multi-parametric imaging assay was developed to capture ex vivo primary islet proliferation, allowing positive identification of compounds that can selectively enhance islet beta-cell proliferation. The use of microscopy-based, high-content imaging technology enables acquisition of additional multi-parametric information such as proliferating populations in the islet beta and non beta-cells, insulin intensity, and cell counts, improving understanding of on and off target effects in primary tissue. The protocol requires access to a high-throughput microscopy platform for automated image acquisition of treated islet cells in assay plates. High content image analysis software is required to extract multiparametric cellular features and aid identification of therapeutically relevant small molecules and perturbants. Several putative beta-cell proliferative compounds have validated in this high throughput assay format, including the pleiotropic hormone prolactin [1] and the small molecule DYRK1A inhibitor harmine [2]. It is recommended to include one, or both, as positive controls to provide a reference for image analysis, give confidence in assay performance and capture potential assay variability during experimental runs. The protocol outlined specifically focuses on the multiparametric assessment of betacell proliferation in mouse and rat ex vivo islets and provides the methodology required for the collection of high quality cellular material. The high throughput, plate based assay can additionally be adapted to evaluate and quantify other disease relevant endpoints by high content microscopy and be applied to other downstream measurements. One of the caveats of a high-throughput, 384 microplate beta-cell proliferative assay is its limitations to facilitate human beta-cell proliferation detection, especially for weak activators. Adult human beta-cell proliferation is an extremely rare biological event and assessment experimentally can be donor dependent. In addition lower human islet beta-cell subpopulations require large numbers of cells for accurate rare event measurement.
Collapse
Affiliation(s)
- Niamh Mullooly
- Functional Genomics, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - David M Smith
- Emerging Innovations Unit, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Davide Gianni
- Functional Genomics, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
2
|
Ching C, Iich E, Teo AKK. Harnessing Human Pluripotent Stem Cell-Derived Pancreatic In Vitro Models for High-Throughput Toxicity Testing and Diabetes Drug Discovery. Handb Exp Pharmacol 2023; 281:301-332. [PMID: 37306817 DOI: 10.1007/164_2023_655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The long-standing goals in diabetes research are to improve β-cell survival, functionality and increase β-cell mass. Current strategies to manage diabetes progression are still not ideal for sustained maintenance of normoglycemia, thereby increasing demand for the development of novel drugs. Available pancreatic cell lines, cadaveric islets, and their culture methods and formats, either 2D or 3D, allow for multiple avenues of experimental design to address diverse aims in the research setting. More specifically, these pancreatic cells have been employed in toxicity testing, diabetes drug screens, and with careful curation, can be optimized for use in efficient high-throughput screenings (HTS). This has since spearheaded the understanding of disease progression and related mechanisms, as well as the discovery of potential drug candidates which could be the cornerstone for diabetes treatment. This book chapter will touch on the pros and cons of the most widely used pancreatic cells, including the more recent human pluripotent stem cell-derived pancreatic cells, and HTS strategies (cell models, design, readouts) that can be used for the purpose of toxicity testing and diabetes drug discovery.
Collapse
Affiliation(s)
- Carmen Ching
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elhadi Iich
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022; 164:6836713. [PMID: 36412119 PMCID: PMC9923807 DOI: 10.1210/endocr/bqac193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.
Collapse
Affiliation(s)
- Roy A Goode
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Julia M Hum
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Michael A Kalwat
- Correspondence: Michael A. Kalwat, PhD, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, 1210 Waterway Blvd, Suite 2000, Indianapolis, IN 46202, USA. or
| |
Collapse
|
4
|
Jain C, Bilekova S, Lickert H. Targeting pancreatic β cells for diabetes treatment. Nat Metab 2022; 4:1097-1108. [PMID: 36131204 DOI: 10.1038/s42255-022-00618-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
Insulin is a life-saving drug for patients with type 1 diabetes; however, even today, no pharmacotherapy can prevent the loss or dysfunction of pancreatic insulin-producing β cells to stop or reverse disease progression. Thus, pancreatic β cells have been a main focus for cell-replacement and regenerative therapies as a curative treatment for diabetes. In this Review, we highlight recent advances toward the development of diabetes therapies that target β cells to enhance proliferation, redifferentiation and protection from cell death and/or enable selective killing of senescent β cells. We describe currently available therapies and their mode of action, as well as insufficiencies of glucagon-like peptide 1 (GLP-1) and insulin therapies. We discuss and summarize data collected over the last decades that support the notion that pharmacological targeting of β cell insulin signalling might protect and/or regenerate β cells as an improved treatment of patients with diabetes.
Collapse
Affiliation(s)
- Chirag Jain
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Sara Bilekova
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair of β-Cell Biology, Technische Universität München, School of Medicine, Klinikum Rechts der Isar, München, Germany.
| |
Collapse
|
5
|
Wagner BK. Small-molecule discovery in the pancreatic beta cell. Curr Opin Chem Biol 2022; 68:102150. [PMID: 35487100 DOI: 10.1016/j.cbpa.2022.102150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022]
Abstract
The pancreatic beta cell is the only cell type in the body responsible for insulin secretion, and thus plays a unique role in the control of glucose homeostasis. The loss of beta-cell mass and function plays an important role in both type 1 and type 2 diabetes. Thus, using chemical biology to identify small molecules targeting the beta cell could be an important component to developing future therapeutics for diabetes. This strategy provides an attractive path toward increasing beta-cell numbers in vivo. A regenerative strategy involves enhancing proliferation, differentiation, or neogenesis. On the other hand, protecting beta cells from cell death, or improving maturity and function, could preserve beta-cell mass. Here, we discuss the current state of chemical matter available to study beta-cell regeneration, and how they were discovered.
Collapse
Affiliation(s)
- Bridget K Wagner
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA.
| |
Collapse
|
6
|
Shcheglova E, Blaszczyk K, Borowiak M. Mitogen Synergy: An Emerging Route to Boosting Human Beta Cell Proliferation. Front Cell Dev Biol 2022; 9:734597. [PMID: 35155441 PMCID: PMC8829426 DOI: 10.3389/fcell.2021.734597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Decreased number and function of beta cells are a key aspect of diabetes mellitus (diabetes), a disease that remains an onerous global health problem. Means of restoring beta cell mass are urgently being sought as a potential cure for diabetes. Several strategies, such as de novo beta cell derivation via pluripotent stem cell differentiation or mature somatic cell transdifferentiation, have yielded promising results. Beta cell expansion is another promising strategy, rendered challenging by the very low proliferative capacity of beta cells. Many effective mitogens have been identified in rodents, but the vast majority do not have similar mitogenic effects in human beta cells. Extensive research has led to the identification of several human beta cell mitogens, but their efficacy and specificity remain insufficient. An approach based on the simultaneous application of several mitogens has recently emerged and can yield human beta cell proliferation rates of up to 8%. Here, we discuss recent advances in restoration of the beta cell population, focusing on mitogen synergy, and the contribution of RNA-sequencing (RNA-seq) to accelerating the elucidation of signaling pathways in proliferating beta cells and the discovery of novel mitogens. Together, these approaches have taken beta cell research up a level, bringing us closer to a cure for diabetes.
Collapse
Affiliation(s)
- Ekaterina Shcheglova
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Katarzyna Blaszczyk
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Malgorzata Borowiak, ;
| |
Collapse
|
7
|
Bartolomé A. Stem Cell-Derived β Cells: A Versatile Research Platform to Interrogate the Genetic Basis of β Cell Dysfunction. Int J Mol Sci 2022; 23:501. [PMID: 35008927 PMCID: PMC8745644 DOI: 10.3390/ijms23010501] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β cell dysfunction is a central component of diabetes progression. During the last decades, the genetic basis of several monogenic forms of diabetes has been recognized. Genome-wide association studies (GWAS) have also facilitated the identification of common genetic variants associated with an increased risk of diabetes. These studies highlight the importance of impaired β cell function in all forms of diabetes. However, how most of these risk variants confer disease risk, remains unanswered. Understanding the specific contribution of genetic variants and the precise role of their molecular effectors is the next step toward developing treatments that target β cell dysfunction in the era of personalized medicine. Protocols that allow derivation of β cells from pluripotent stem cells, represent a powerful research tool that allows modeling of human development and versatile experimental designs that can be used to shed some light on diabetes pathophysiology. This article reviews different models to study the genetic basis of β cell dysfunction, focusing on the recent advances made possible by stem cell applications in the field of diabetes research.
Collapse
Affiliation(s)
- Alberto Bartolomé
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, 28029 Madrid, Spain
| |
Collapse
|
8
|
Simonett SP, Shin S, Herring JA, Bacher R, Smith LA, Dong C, Rabaglia ME, Stapleton DS, Schueler KL, Choi J, Bernstein MN, Turkewitz DR, Perez-Cervantes C, Spaeth J, Stein R, Tessem JS, Kendziorski C, Keleş S, Moskowitz IP, Keller MP, Attie AD. Identification of direct transcriptional targets of NFATC2 that promote β cell proliferation. J Clin Invest 2021; 131:e144833. [PMID: 34491912 PMCID: PMC8553569 DOI: 10.1172/jci144833] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
The transcription factor NFATC2 induces β cell proliferation in mouse and human islets. However, the genomic targets that mediate these effects have not been identified. We expressed active forms of Nfatc2 and Nfatc1 in human islets. By integrating changes in gene expression with genomic binding sites for NFATC2, we identified approximately 2200 transcriptional targets of NFATC2. Genes induced by NFATC2 were enriched for transcripts that regulate the cell cycle and for DNA motifs associated with the transcription factor FOXP. Islets from an endocrine-specific Foxp1, Foxp2, and Foxp4 triple-knockout mouse were less responsive to NFATC2-induced β cell proliferation, suggesting the FOXP family works to regulate β cell proliferation in concert with NFATC2. NFATC2 induced β cell proliferation in both mouse and human islets, whereas NFATC1 did so only in human islets. Exploiting this species difference, we identified approximately 250 direct transcriptional targets of NFAT in human islets. This gene set enriches for cell cycle-associated transcripts and includes Nr4a1. Deletion of Nr4a1 reduced the capacity of NFATC2 to induce β cell proliferation, suggesting that much of the effect of NFATC2 occurs through its induction of Nr4a1. Integration of noncoding RNA expression, chromatin accessibility, and NFATC2 binding sites enabled us to identify NFATC2-dependent enhancer loci that mediate β cell proliferation.
Collapse
Affiliation(s)
- Shane P. Simonett
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Sunyoung Shin
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Jacob A. Herring
- Nutrition, Dietetics and Food Science Department, Brigham Young University, Provo, Utah, USA
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Linsin A. Smith
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Chenyang Dong
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Mary E. Rabaglia
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Donnie S. Stapleton
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Kathryn L. Schueler
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Jeea Choi
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Daniel R. Turkewitz
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Carlos Perez-Cervantes
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Jason Spaeth
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jeffery S. Tessem
- Nutrition, Dietetics and Food Science Department, Brigham Young University, Provo, Utah, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Sündüz Keleş
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Ivan P. Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Mark P. Keller
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Alan D. Attie
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
9
|
Pahlavanneshan S, Behmanesh M, Tahamtani Y, Hajizadeh-Saffar E, Basiri M, Baharvand H. Induction of ß Cell Replication by Small Molecule-Mediated Menin Inhibition and Combined PKC Activation and TGF‑ß Inhibition as Revealed by A Refined Primary Culture Screening. CELL JOURNAL 2021; 23:633-639. [PMID: 34939756 PMCID: PMC8665985 DOI: 10.22074/cellj.2021.7437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/04/2020] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Pancreatic β cells are recognized as central players in the pathogenesis of types 1 and 2 diabetes. Efficient and robust primary culture methods are required to interrogate β cell biology and screen potential anti-diabetic therapeutics. The aim of this study was to refine monolayer culture of beta cells and to investigate potential inducers of beta cell proliferation. MATERIALS AND METHODS In this experimental study, we compared different culture methods to optimize conditions required for a monolayer culture of rat pancreatic islet cells in order to facilitate image analysis-based assays. We also used the refined culture method to screen a group of rationally selected candidate small molecules and their combinations to determine their potential proliferative effects on the β cells. RESULTS Ham's F10 medium supplemented with 2% foetal bovine serum (FBS) in the absence of any surface coating provided a superior monolayer β cell culture, while other conditions induced fibroblast-like cell growth or multilayer cell aggregation over two weeks. Evaluation of candidate small molecules showed that a menin inhibitor MI-2 and a combination of transforming growth factor-β (TGF-β) inhibitor SB481542 and protein kinase C (PKC) activator indolactam V (IndV) significantly induced replication of pancreatic β cells. CONCLUSION Overall, our optimized culture condition provided a convenient approach to study the cultured pancreatic islet cells and enabled us to detect the proliferative effect of menin inhibition and combined TGF-β inhibition and PKC activation, which could be considered as potential strategies for inducing β cell proliferation and regeneration.
Collapse
Affiliation(s)
- Saghar Pahlavanneshan
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran,P.O.Box: 14115-154Department of Genetics, Faculty of Biological SciencesTarbiat Modares UniversityTehranIranP.O.Box: 16635-148Department of Stem Cells and Developmental BiologyCell Science Research CentreRoyan Institute for Stem Cell Biology
and TechnologyACECRTehranIran
Emails: ,
| | - Yaser Tahamtani
- . Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR,
Tehran, Iran
| | - Mohsen Basiri
- . Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- . Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Department of Developmental Biology, University of Science and Culture, Tehran, Iran,P.O.Box: 14115-154Department of Genetics, Faculty of Biological SciencesTarbiat Modares UniversityTehranIranP.O.Box: 16635-148Department of Stem Cells and Developmental BiologyCell Science Research CentreRoyan Institute for Stem Cell Biology
and TechnologyACECRTehranIran
Emails: ,
| |
Collapse
|
10
|
Iorio C, Rourke JL, Wells L, Sakamaki JI, Moon E, Hu Q, Kin T, Screaton RA. Silencing the G-protein coupled receptor 3-salt inducible kinase 2 pathway promotes human β cell proliferation. Commun Biol 2021; 4:907. [PMID: 34302056 PMCID: PMC8302759 DOI: 10.1038/s42003-021-02433-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Loss of pancreatic β cells is the hallmark of type 1 diabetes, for which provision of insulin is the standard of care. While regenerative and stem cell therapies hold the promise of generating single-source or host-matched tissue to obviate immune-mediated complications, these will still require surgical intervention and immunosuppression. Here we report the development of a high-throughput RNAi screening approach to identify upstream pathways that regulate adult human β cell quiescence and demonstrate in a screen of the GPCRome that silencing G-protein coupled receptor 3 (GPR3) leads to human pancreatic β cell proliferation. Loss of GPR3 leads to activation of Salt Inducible Kinase 2 (SIK2), which is necessary and sufficient to drive cell cycle entry, increase β cell mass, and enhance insulin secretion in mice. Taken together, our data show that targeting the GPR3-SIK2 pathway is a potential strategy to stimulate the regeneration of β cells.
Collapse
Affiliation(s)
| | - Jillian L Rourke
- Sunnybrook Research Institute, Toronto, Canada
- Mount Allison University, Sackville, NB, Canada
| | - Lisa Wells
- Sunnybrook Research Institute, Toronto, Canada
| | - Jun-Ichi Sakamaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Emily Moon
- Sunnybrook Research Institute, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Queenie Hu
- Sunnybrook Research Institute, Toronto, Canada
| | - Tatsuya Kin
- Clinical Islet Laboratory, University of Alberta Hospital, Edmonton, Canada
| | - Robert A Screaton
- Sunnybrook Research Institute, Toronto, Canada.
- Department of Biochemistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
11
|
Kumar K, Suebsuwong C, Wang P, Garcia-Ocana A, Stewart AF, DeVita RJ. DYRK1A Inhibitors as Potential Therapeutics for β-Cell Regeneration for Diabetes. J Med Chem 2021; 64:2901-2922. [PMID: 33682417 DOI: 10.1021/acs.jmedchem.0c02050] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
According to the World Health Organization (WHO), 422 million people are suffering from diabetes worldwide. Current diabetes therapies are focused on optimizing blood glucose control to prevent long-term diabetes complications. Unfortunately, current therapies have failed to achieve glycemic targets in the majority of people with diabetes. In this context, regeneration of functional insulin-producing human β-cells in people with diabetes through the use of DYRK1A inhibitor drugs has recently received special attention. Several small molecule DYRK1A inhibitors have been identified that induce human β-cell proliferation in vitro and in vivo. Furthermore, DYRK1A inhibitors have also been shown to synergize β-cell proliferation with other classes of drugs, such as TGFβ inhibitors and GLP-1 receptor agonists. In this perspective, we review the status of DYRK1A as a therapeutic target for β-cell proliferation and provide perspectives on technical and scientific challenges for future translational development.
Collapse
Affiliation(s)
- Kunal Kumar
- Drug Discovery Institute and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Chalada Suebsuwong
- Drug Discovery Institute and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Peng Wang
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Adolfo Garcia-Ocana
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Andrew F Stewart
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Robert J DeVita
- Drug Discovery Institute and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
12
|
Oakie A, Nostro MC. Harnessing Proliferation for the Expansion of Stem Cell-Derived Pancreatic Cells: Advantages and Limitations. Front Endocrinol (Lausanne) 2021; 12:636182. [PMID: 33716986 PMCID: PMC7947602 DOI: 10.3389/fendo.2021.636182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Restoring the number of glucose-responsive β-cells in patients living with diabetes is critical for achieving normoglycemia since functional β-cells are lost during the progression of both type 1 and 2 diabetes. Stem cell-derived β-cell replacement therapies offer an unprecedented opportunity to replace the lost β-cell mass, yet differentiation efficiencies and the final yield of insulin-expressing β-like cells are low when using established protocols. Driving cellular proliferation at targeted points during stem cell-derived pancreatic progenitor to β-like cell differentiation can serve as unique means to expand the final cell therapeutic product needed to restore insulin levels. Numerous studies have examined the effects of β-cell replication upon functionality, using primary islets in vitro and mouse models in vivo, yet studies that focus on proliferation in stem cell-derived pancreatic models are only just emerging in the field. This mini review will discuss the current literature on cell proliferation in pancreatic cells, with a focus on the proliferative state of stem cell-derived pancreatic progenitors and β-like cells during their differentiation and maturation. The benefits of inducing proliferation to increase the final number of β-like cells will be compared against limitations associated with driving replication, such as the blunted capacity of proliferating β-like cells to maintain optimal β-cell function. Potential strategies that may bypass the challenges induced by the up-regulation of cell cycle-associated factors during β-cell differentiation will be proposed.
Collapse
Affiliation(s)
- Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Lewis PL, Wells JM. Engineering-inspired approaches to study β-cell function and diabetes. Stem Cells 2021; 39:522-535. [PMID: 33497522 DOI: 10.1002/stem.3340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand β-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and β-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human β-cells, to cell lines, to pluripotent stem cell-derived β-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are β-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying β-cell biology, improving β-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic.
Collapse
Affiliation(s)
- Phillip L Lewis
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
14
|
Kh S, Haider KH. Stem Cells: A Renewable Source of Pancreatic β-Cells and Future for Diabetes Treatment. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
15
|
Spears E, Serafimidis I, Powers AC, Gavalas A. Debates in Pancreatic Beta Cell Biology: Proliferation Versus Progenitor Differentiation and Transdifferentiation in Restoring β Cell Mass. Front Endocrinol (Lausanne) 2021; 12:722250. [PMID: 34421829 PMCID: PMC8378310 DOI: 10.3389/fendo.2021.722250] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
In all forms of diabetes, β cell mass or function is reduced and therefore the capacity of the pancreatic cells for regeneration or replenishment is a critical need. Diverse lines of research have shown the capacity of endocrine as well as acinar, ductal and centroacinar cells to generate new β cells. Several experimental approaches using injury models, pharmacological or genetic interventions, isolation and in vitro expansion of putative progenitors followed by transplantations or a combination thereof have suggested several pathways for β cell neogenesis or regeneration. The experimental results have also generated controversy related to the limitations and interpretation of the experimental approaches and ultimately their physiological relevance, particularly when considering differences between mouse, the primary animal model, and human. As a result, consensus is lacking regarding the relative importance of islet cell proliferation or progenitor differentiation and transdifferentiation of other pancreatic cell types in generating new β cells. In this review we summarize and evaluate recent experimental approaches and findings related to islet regeneration and address their relevance and potential clinical application in the fight against diabetes.
Collapse
Affiliation(s)
- Erick Spears
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ioannis Serafimidis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
- VA Tennessee Valley Healthcare System, Nashville, TN, United States
- *Correspondence: Anthony Gavalas, ; Alvin C. Powers,
| | - Anthony Gavalas
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Neuherberg, Germany
- *Correspondence: Anthony Gavalas, ; Alvin C. Powers,
| |
Collapse
|
16
|
Fu A, Alvarez-Perez JC, Avizonis D, Kin T, Ficarro SB, Choi DW, Karakose E, Badur MG, Evans L, Rosselot C, Bridon G, Bird GH, Seo HS, Dhe-Paganon S, Kamphorst JJ, Stewart AF, James Shapiro AM, Marto JA, Walensky LD, Jones RG, Garcia-Ocana A, Danial NN. Glucose-dependent partitioning of arginine to the urea cycle protects β-cells from inflammation. Nat Metab 2020; 2:432-446. [PMID: 32694660 PMCID: PMC7568475 DOI: 10.1038/s42255-020-0199-4] [Citation(s) in RCA: 36] [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] [Received: 01/31/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023]
Abstract
Chronic inflammation is linked to diverse disease processes, but the intrinsic mechanisms that determine cellular sensitivity to inflammation are incompletely understood. Here, we show the contribution of glucose metabolism to inflammation-induced changes in the survival of pancreatic islet β-cells. Using metabolomic, biochemical and functional analyses, we investigate the protective versus non-protective effects of glucose in the presence of pro-inflammatory cytokines. When protective, glucose metabolism augments anaplerotic input into the TCA cycle via pyruvate carboxylase (PC) activity, leading to increased aspartate levels. This metabolic mechanism supports the argininosuccinate shunt, which fuels ureagenesis from arginine and conversely diminishes arginine utilization for production of nitric oxide (NO), a chief mediator of inflammatory cytotoxicity. Activation of the PC-urea cycle axis is sufficient to suppress NO synthesis and shield cells from death in the context of inflammation and other stress paradigms. Overall, these studies uncover a previously unappreciated link between glucose metabolism and arginine-utilizing pathways via PC-directed ureagenesis as a protective mechanism.
Collapse
Affiliation(s)
- Accalia Fu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Juan Carlos Alvarez-Perez
- Diabetes, Obesity and Metabolism Institute, Department of Medicine, Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daina Avizonis
- Rosalind and Morris Goodman Cancer Center Metabolomics Core, Montreal, Canada
| | - Tatsuya Kin
- Clinical Islet Transplant Program, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Scott B Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dong Wook Choi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Esra Karakose
- Diabetes, Obesity and Metabolism Institute, Department of Medicine, Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Lindsay Evans
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Carolina Rosselot
- Diabetes, Obesity and Metabolism Institute, Department of Medicine, Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gaelle Bridon
- Rosalind and Morris Goodman Cancer Center Metabolomics Core, Montreal, Canada
| | - Gregory H Bird
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | | | - Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Department of Medicine, Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A M James Shapiro
- Clinical Islet Transplant Program, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Jarrod A Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Loren D Walensky
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Russell G Jones
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Adolfo Garcia-Ocana
- Diabetes, Obesity and Metabolism Institute, Department of Medicine, Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nika N Danial
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Maugein A, Diedisheim M, Bailly K, Scharfmann R, Albagli O. The RB gene family controls the maturation state of the EndoC-βH2 human pancreatic β-cells. Differentiation 2020; 113:1-9. [PMID: 32120156 DOI: 10.1016/j.diff.2020.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 01/18/2023]
Abstract
The functional maturation of human pancreatic β-cells remains poorly understood. EndoC-βH2 is a human β-cell line with a reversible immortalized phenotype. Removal of the two oncogenes, SV40LT and hTERT introduced for its propagation, stops proliferation, triggers cell size increase and senescence, promotes mitochondrial activity and amplifies several β-cell traits and functions. Overall, these events recapitulate several aspects of functional β-cell maturation. We report here that selective depletion of SV40LT, but not of hTERT, is sufficient to revert EndoC-βH2 immortalization. SV40LT inhibits the activity of the RB family members and of P53. In EndoC-βH2 cells, the knock-down of RB itself, and, to a lesser extent, of its relative P130, precludes most events triggered by SV40LT depletion. In contrast, the knock-down of P53 does not prevent reversion of immortalization. Thus, an increase in RB and P130 activity, but not in P53 activity, is required for functional maturation of EndoC-βH2 cells upon SV40LT-depletion. In addition, RB and/or P130 depletion in SV40LT-expressing EndoC-βH2 cells decreases cell size, stimulates proliferation, and decreases the expression of key β-cell genes. Thus, despite SV40LT expression, EndoC-βH2 cells have a residual RB activity, which when suppressed reverts them to a more immature phenotype. These results show that the expression and activity levels of RB family members, especially RB itself, regulate the maturation state of EndoC-βH2 cells.
Collapse
Affiliation(s)
- Alicia Maugein
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France
| | - Marc Diedisheim
- Assistance Publique - Hôpitaux de Paris, Diabetology Department, Paris University, Cochin Hospital, and INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris University, 75006, Paris, France
| | - Karine Bailly
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France
| | - Raphaël Scharfmann
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France
| | - Olivier Albagli
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France.
| |
Collapse
|
18
|
Holowiecki A, Linstrum K, Ravisankar P, Chetal K, Salomonis N, Waxman JS. Pbx4 limits heart size and fosters arch artery formation by partitioning second heart field progenitors and restricting proliferation. Development 2020; 147:dev185652. [PMID: 32094112 PMCID: PMC7063670 DOI: 10.1242/dev.185652] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/06/2020] [Indexed: 12/11/2022]
Abstract
Vertebrate heart development requires the integration of temporally distinct differentiating progenitors. However, few signals are understood that restrict the size of the later-differentiating outflow tract (OFT). We show that improper specification and proliferation of second heart field (SHF) progenitors in zebrafish lazarus (lzr) mutants, which lack the transcription factor Pbx4, produces enlarged hearts owing to an increase in ventricular and smooth muscle cells. Specifically, Pbx4 initially promotes the partitioning of the SHF into anterior progenitors, which contribute to the OFT, and adjacent endothelial cell progenitors, which contribute to posterior pharyngeal arches. Subsequently, Pbx4 limits SHF progenitor (SHFP) proliferation. Single cell RNA sequencing of nkx2.5+ cells revealed previously unappreciated distinct differentiation states and progenitor subpopulations that normally reside within the SHF and arterial pole of the heart. Specifically, the transcriptional profiles of Pbx4-deficient nkx2.5+ SHFPs are less distinct and display characteristics of normally discrete proliferative progenitor and anterior, differentiated cardiomyocyte populations. Therefore, our data indicate that the generation of proper OFT size and arch arteries requires Pbx-dependent stratification of unique differentiation states to facilitate both homeotic-like transformations and limit progenitor production within the SHF.
Collapse
Affiliation(s)
- Andrew Holowiecki
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Kelsey Linstrum
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Padmapriyadarshini Ravisankar
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Kashish Chetal
- Bioinformatics Division, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Nathan Salomonis
- Bioinformatics Division, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
19
|
Scharfmann R, Staels W, Albagli O. The supply chain of human pancreatic β cell lines. J Clin Invest 2019; 129:3511-3520. [PMID: 31478912 PMCID: PMC6715382 DOI: 10.1172/jci129484] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patients with type 1 or type 2 diabetes have an insufficiency in their functional β cell mass. To advance diabetes treatment and to work toward a cure, a better understanding of how to protect the pancreatic β cells against autoimmune or metabolic assaults (e.g., obesity, gestation) will be required. Over the past decades, β cell protection has been extensively investigated in rodents both in vivo and in vitro using isolated islets or rodent β cell lines. Transferring these rodent data to humans has long been challenging, at least partly for technical reasons: primary human islet preparations were scarce and functional human β cell lines were lacking. In 2011, we described a robust protocol of targeted oncogenesis in human fetal pancreas and produced the first functional human β cell line, and in subsequent years additional lines with specific traits. These cell lines are currently used by more than 150 academic and industrial laboratories worldwide. In this Review, we first explain how we developed the human β cell lines and why we think we succeeded where others, despite major efforts, did not. Next, we discuss the use of such functional human β cell lines and share some perspectives on their use to advance diabetes research.
Collapse
Affiliation(s)
- Raphael Scharfmann
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Willem Staels
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels, Belgium
| | - Olivier Albagli
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| |
Collapse
|
20
|
Zhong F, Jiang Y. Endogenous Pancreatic β Cell Regeneration: A Potential Strategy for the Recovery of β Cell Deficiency in Diabetes. Front Endocrinol (Lausanne) 2019; 10:101. [PMID: 30842756 PMCID: PMC6391341 DOI: 10.3389/fendo.2019.00101] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/04/2019] [Indexed: 12/11/2022] Open
Abstract
Endogenous pancreatic β cell regeneration is a potential strategy for β cell expansion or neogenesis to treat diabetes. Regeneration can occur through stimulation of existing β cell replication or conversion of other pancreatic cells into β cells. Recently, various strategies and approaches for stimulation of endogenous β cell regeneration have been evaluated, but they were not suitable for clinical application. In this paper, we comprehensively review these strategies, and further discuss various factors involved in regulation of β cell regeneration under physiological or pathological conditions, such as mediators, transcription factors, signaling pathways, and potential pharmaceutical drugs. Furthermore, we discuss possible reasons for the failure of regenerative medicines in clinical trials, and possible strategies for improving β cell regeneration. As β cell heterogeneity and plasticity determines their function and environmental adaptability, we focus on β cell subtype markers and discuss the importance of research evaluating the characteristics of new β cells. In addition, based on the autoimmunologic features of type 1 diabetes, NOD/Lt-SCID-IL2rg null (NSG) mice grafted with human immune cells and β cells are recommended for use in evaluation of antidiabetic regenerative medicines. This review will further understand current advances in endogenous β cell regeneration, and provide potential new strategies for the treatment of diabetes focused on cell therapy.
Collapse
Affiliation(s)
- Fan Zhong
- Department of Gastroenterology, Songjiang Hospital Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Jiang
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Akbib S, Stichelmans J, Stangé G, Ling Z, Assefa Z, Hellemans KH. Glucocorticoids and checkpoint tyrosine kinase inhibitors stimulate rat pancreatic beta cell proliferation differentially. PLoS One 2019; 14:e0212210. [PMID: 30779812 PMCID: PMC6380609 DOI: 10.1371/journal.pone.0212210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cell therapy for diabetes could benefit from the identification of small-molecule compounds that increase the number of functional pancreatic beta cells. Using a newly developed screening assay, we previously identified glucocorticoids as potent stimulators of human and rat beta cell proliferation. We now compare the stimulatory action of these steroid hormones to a selection of checkpoint tyrosine kinase inhibitors that were also found to activate the cell cycle-in beta cells and analyzed their respective effects on DNA-synthesis, beta cell numbers and expression of cell cycle regulators. Our data using glucocorticoids in combination with a receptor antagonist, mifepristone, show that 48h exposure is sufficient to allow beta cells to pass the cell cycle restriction point and to become committed to cell division regardless of sustained glucocorticoid-signaling. To reach the end-point of mitosis another 40h is required. Within 14 days glucocorticoids stimulate up to 75% of the cells to undergo mitosis, which indicates that these steroid hormones act as proliferation competence-inducing factors. In contrast, by correlating thymidine-analogue incorporation to changes in absolute cell numbers, we show that the checkpoint kinase inhibitors, as compared to glucocorticoids, stimulate DNA-synthesis only during a short time-window in a minority of cells, insufficient to give a measurable increase of beta cell numbers. Glucocorticoids, but not the kinase inhibitors, were also found to induce changes in the expression of checkpoint regulators. Our data, using checkpoint kinase-specific inhibitors further point to a role for Chk1 and Cdk1 in G1/S transition and progression of beta cells through the cell cycle upon stimulation with glucocorticoids.
Collapse
Affiliation(s)
- Sarah Akbib
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jordy Stichelmans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Stangé
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Zhidong Ling
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Beta Cell Bank, University Hospital Brussels, Brussels, Belgium
| | - Zerihun Assefa
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine H. Hellemans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Center for Beta Cell Therapy in Diabetes, Brussels, Belgium
| |
Collapse
|
22
|
Moyce BL, Dolinsky VW. Maternal β-Cell Adaptations in Pregnancy and Placental Signalling: Implications for Gestational Diabetes. Int J Mol Sci 2018; 19:ijms19113467. [PMID: 30400566 PMCID: PMC6274918 DOI: 10.3390/ijms19113467] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/24/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Rates of gestational diabetes mellitus (GDM) are on the rise worldwide, and the number of pregnancies impacted by GDM and resulting complications are also increasing. Pregnancy is a period of unique metabolic plasticity, during which mild insulin resistance is a physiological adaptation to prioritize fetal growth. To compensate for this, the pancreatic β-cell utilizes a variety of adaptive mechanisms, including increasing mass, number and insulin-secretory capacity to maintain glucose homeostasis. When insufficient insulin production does not overcome insulin resistance, hyperglycemia can occur. Changes in the maternal system that occur in GDM such as lipotoxicity, inflammation and oxidative stress, as well as impairments in adipokine and placental signalling, are associated with impaired β-cell adaptation. Understanding these pathways, as well as mechanisms of β-cell dysfunction in pregnancy, can identify novel therapeutic targets beyond diet and lifestyle interventions, insulin and antihyperglycemic agents currently used for treating GDM.
Collapse
Affiliation(s)
- Brittany L Moyce
- Department of Pharmacology & Therapeutics and the Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba and the Manitoba Developmental Origins of Chronic Diseases in Children Network (DEVOTION), University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| | - Vernon W Dolinsky
- Department of Pharmacology & Therapeutics and the Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba and the Manitoba Developmental Origins of Chronic Diseases in Children Network (DEVOTION), University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| |
Collapse
|
23
|
Yi L, Swensen AC, Qian WJ. Serum biomarkers for diagnosis and prediction of type 1 diabetes. Transl Res 2018; 201:13-25. [PMID: 30144424 PMCID: PMC6177288 DOI: 10.1016/j.trsl.2018.07.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/02/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022]
Abstract
Type 1 diabetes (T1D) culminates in the autoimmune destruction of the pancreatic βcells, leading to insufficient production of insulin and development of hyperglycemia. Serum biomarkers including a combination of glucose, glycated molecules, C-peptide, and autoantibodies have been well established for the diagnosis of T1D. However, these molecules often mark a late stage of the disease when ∼90% of the pancreatic insulin-producing β-cells have already been lost. With the prevalence of T1D increasing worldwide and because of the physical and psychological burden induced by this disease, there is a great need for prognostic biomarkers to predict T1D development or progression. This would allow us to identify individuals at high risk for early prevention and intervention. Therefore, considerable efforts have been dedicated to the understanding of disease etiology and the discovery of novel biomarkers in the last few decades. The advent of high-throughput and sensitive "-omics" technologies for the study of proteins, nucleic acids, and metabolites have allowed large scale profiling of protein expression and gene changes in T1D patients relative to disease-free controls. In this review, we briefly discuss the classical diagnostic biomarkers of T1D but mainly focus on the novel biomarkers that are identified as markers of β-cell destruction and screened with the use of state-of-the-art "-omics" technologies.
Collapse
Affiliation(s)
- Lian Yi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Adam C Swensen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington.
| |
Collapse
|
24
|
Dominguez Gutierrez G, Xin Y, Okamoto H, Kim J, Lee AH, Ni M, Adler C, Yancopoulos GD, Murphy AJ, Gromada J. Gene Signature of Proliferating Human Pancreatic α Cells. Endocrinology 2018; 159:3177-3186. [PMID: 30010845 DOI: 10.1210/en.2018-00469] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022]
Abstract
Pancreatic α cells proliferate at a low rate, and little is known about the control of this process. Here we report the characterization of human α cells by large-scale, single-cell RNA sequencing coupled with pseudotime ordering. We identified two large subpopulations and a smaller cluster of proliferating α cells with increased expression of genes involved in cell-cycle regulation. The proliferating α cells were differentiated, had normal levels of GCG expression, and showed no signs of cellular stress. Proliferating α cells were detected in both the G1S and G2M phases of the cell cycle. Human α cells proliferate at a fivefold higher rate than human β cells and express lower levels of the cell-cycle inhibitors CDKN1A and CDKN1C. Collectively, this study provides the gene signatures of human α cells and the genes involved in their cell division. The lower expression of two cell-cycle inhibitors in human α cells could account for their higher rate of proliferation compared with their insulin-producing counterparts.
Collapse
Affiliation(s)
| | - Yurong Xin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Jinrang Kim
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Ann-Hwee Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | | | | |
Collapse
|
25
|
Karakose E, Ackeifi C, Wang P, Stewart AF. Advances in drug discovery for human beta cell regeneration. Diabetologia 2018; 61:1693-1699. [PMID: 29770834 PMCID: PMC6239977 DOI: 10.1007/s00125-018-4639-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/05/2018] [Indexed: 12/15/2022]
Abstract
The numbers of insulin-secreting pancreatic beta cells are reduced in people with type 1 and type 2 diabetes. Driving beta cell regeneration in the pancreases of people with diabetes would be an attractive approach to reversing diabetes. While adult human beta cells have long been believed to be terminally differentiated and, therefore, irreversibly quiescent, it has become clear over recent years that this is not true. More specifically, both candidate and unbiased high-throughput screen approaches have revealed several classes of molecules that are clearly able to induce human beta cell proliferation. Here, we review recent approaches and accomplishments in human beta cell regenerative drug discovery. We also list the challenges that this rapidly moving field must confront to translate beta cell regenerative therapy from the laboratory to the clinic.
Collapse
Affiliation(s)
- Esra Karakose
- The Diabetes, Obesity and Metabolism Institute, The Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Courtney Ackeifi
- The Diabetes, Obesity and Metabolism Institute, The Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Peng Wang
- The Diabetes, Obesity and Metabolism Institute, The Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Andrew F Stewart
- The Diabetes, Obesity and Metabolism Institute, The Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY, 10029, USA.
| |
Collapse
|
26
|
Aguayo-Mazzucato C, Bonner-Weir S. Pancreatic β Cell Regeneration as a Possible Therapy for Diabetes. Cell Metab 2018; 27:57-67. [PMID: 28889951 PMCID: PMC5762410 DOI: 10.1016/j.cmet.2017.08.007] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/05/2017] [Accepted: 08/08/2017] [Indexed: 02/08/2023]
Abstract
Diabetes is the result of having inadequate supply of functional insulin-producing β cells. Two possible approaches for replenishing the β cells are: (1) replacement by transplanting cadaveric islets or β cells derived from human embryonic stem cells/induced pluripotent stem cells and (2) induction of endogenous regeneration. This review focuses on endogenous regeneration, which can follow two pathways: enhanced replication of existing β cells and formation of new β cells from cells not expressing insulin, either by conversion from a differentiated cell type (transdifferentiation) or differentiation from progenitors (neogenesis). Exciting progress on both pathways suggest that regeneration may have therapeutic promise.
Collapse
Affiliation(s)
| | - Susan Bonner-Weir
- Joslin Diabetes Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA.
| |
Collapse
|
27
|
Sisino G, Zhou AX, Dahr N, Sabirsh A, Soundarapandian MM, Perera R, Larsson-Lekholm E, Magnone MC, Althage M, Tyrberg B. Long noncoding RNAs are dynamically regulated during β-cell mass expansion in mouse pregnancy and control β-cell proliferation in vitro. PLoS One 2017; 12:e0182371. [PMID: 28796801 PMCID: PMC5552087 DOI: 10.1371/journal.pone.0182371] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/17/2017] [Indexed: 11/25/2022] Open
Abstract
Pregnancy is associated with increased β-cell proliferation driven by prolactin. Long noncoding RNAs (lncRNA) are the most abundant RNA species in the mammalian genome, yet, their functional importance is mainly elusive. Aims/hypothesis: This study tests the hypothesis that lncRNAs regulate β-cell proliferation in response to prolactin in the context of β-cell mass compensation in pregnancy. Methods: The expression profile of lncRNAs in mouse islets at day 14.5 of pregnancy was explored by a bioinformatics approach, further confirmed by quantitative PCR at different days of pregnancy, and islet specificity was evaluated by comparing expression in islets versus other tissues. In order to establish the role of the candidate lncRNAs we studied cell proliferation in mouse islets and the MIN6 β-cell line by EdU incorporation and cell count. Results: We found that a group of lncRNAs is differentially regulated in mouse islets at 14.5 days of pregnancy. At different stages of pregnancy, these lncRNAs are dynamically expressed, and expression is prolactin dependent in mouse islets and MIN6 cells. One of those lncRNAs, Gm16308 (Lnc03), is dynamically regulated during pregnancy, prolactin-dependent and islet-enriched. Silencing Lnc03 in primary β-cells and MIN6 cells inhibits, whereas over-expression stimulates, proliferation even in the absence of prolactin, demonstrating that Lnc03 regulates β-cell growth. Conclusions/interpretation: During pregnancy mouse islet proliferation is correlated with dynamic changes of lncRNA expression. In particular, Lnc03 regulates mouse β-cell proliferation and may be a crucial component of β-cell proliferation in β-cell mass adaptation in both health and disease.
Collapse
Affiliation(s)
- Giorgia Sisino
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Alex-Xianghua Zhou
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Niklas Dahr
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Alan Sabirsh
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | | | - Ranjan Perera
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, United States of America
| | | | - Maria Chiara Magnone
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Magnus Althage
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Björn Tyrberg
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
- * E-mail:
| |
Collapse
|
28
|
Iwata Y, Klaren WD, Lebakken CS, Grimm FA, Rusyn I. High-Content Assay Multiplexing for Vascular Toxicity Screening in Induced Pluripotent Stem Cell-Derived Endothelial Cells and Human Umbilical Vein Endothelial Cells. Assay Drug Dev Technol 2017; 15:267-279. [PMID: 28771372 DOI: 10.1089/adt.2017.786] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Endothelial cells (ECs) play a major role in blood vessel formation and function. While there is longstanding evidence for the potential of chemical exposures to adversely affect EC function and vascular development, the hazard potential of chemicals with respect to vascular effects is not routinely evaluated in safety assessments. Induced pluripotent stem cell (iPSC)-derived ECs promise to provide a physiologically relevant, organotypic culture model that is amenable for high-throughput (HT) EC toxicant screening and may represent a viable alternative to traditional in vitro models, including human umbilical vein endothelial cells (HUVECs). To evaluate the utility of iPSC-ECs for multidimensional HT toxicity profiling of chemicals, both iPSC-ECs and HUVECs were exposed to selected positive (angiogenesis inhibitors, cytotoxic agents) and negative compounds in concentration response for either 16 or 24 h in a 384-well plate format. Furthermore, chemical effects on vascularization were quantified using EC angiogenesis on biological (Geltrex™) and synthetic (SP-105 angiogenesis hydrogel) extracellular matrices. Cellular toxicity was assessed using high-content live cell imaging and the CellTiter-Glo® assay. Assay performance indicated good to excellent assay sensitivity and reproducibility for both cell types investigated. Both iPSC-derived ECs and HUVECs formed tube-like structures on Geltrex™ and hydrogel, an effect that was inhibited by angiogenesis inhibitors and cytotoxic agents in a concentration-dependent manner. The quality of HT assays in HUVECs was generally higher than that in iPSC-ECs. Altogether, this study demonstrates the capability of ECs for comprehensive assessment of the biological effects of chemicals on vasculature in a HT compatible format.
Collapse
Affiliation(s)
- Yasuhiro Iwata
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - William D Klaren
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | | | - Fabian A Grimm
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Ivan Rusyn
- 1 Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| |
Collapse
|
29
|
Xu EE, Sasaki S, Speckmann T, Nian C, Lynn FC. SOX4 Allows Facultative β-Cell Proliferation Through Repression of Cdkn1a. Diabetes 2017; 66:2213-2219. [PMID: 28495880 DOI: 10.2337/db16-1074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 05/03/2017] [Indexed: 11/13/2022]
Abstract
The high-mobility group box transcription factor SOX4 is the most highly expressed SOX family protein in pancreatic islets, and mutations in Sox4 are associated with an increased risk of developing type 2 diabetes. We used an inducible β-cell knockout mouse model to test the hypothesis that Sox4 is essential for the maintenance of β-cell number during the development of type 2 diabetes. Knockout of Sox4 at 6 weeks of age resulted in time-dependent worsening of glucose tolerance, impairment of insulin secretion, and diabetes by 30 weeks of age. Immunostaining revealed a decrease in β-cell mass in knockout mice that was caused by a 39% reduction in β-cell proliferation. Gene expression studies revealed that induction of the cell cycle inhibitor Cdkn1a was responsible for the decreased proliferation in the knockout animals. Altogether, this study demonstrates that SOX4 is necessary for adult β-cell replication through direct regulation of the β-cell cycle.
Collapse
Affiliation(s)
- Eric E Xu
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Cell and Developmental Biology Graduate Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shugo Sasaki
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Thilo Speckmann
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Cell and Developmental Biology Graduate Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cuilan Nian
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Francis C Lynn
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Cell and Developmental Biology Graduate Program, University of British Columbia, Vancouver, British Columbia, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Aamodt KI, Aramandla R, Brown JJ, Fiaschi-Taesch N, Wang P, Stewart AF, Brissova M, Powers AC. Development of a reliable automated screening system to identify small molecules and biologics that promote human β-cell regeneration. Am J Physiol Endocrinol Metab 2016; 311:E859-E868. [PMID: 27624103 PMCID: PMC5130356 DOI: 10.1152/ajpendo.00515.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 09/09/2016] [Indexed: 11/22/2022]
Abstract
Numerous compounds stimulate rodent β-cell proliferation; however, translating these findings to human β-cells remains a challenge. To examine human β-cell proliferation in response to such compounds, we developed a medium-throughput in vitro method of quantifying adult human β-cell proliferation markers. This method is based on high-content imaging of dispersed islet cells seeded in 384-well plates and automated cell counting that identifies fluorescently labeled β-cells with high specificity using both nuclear and cytoplasmic markers. β-Cells from each donor were assessed for their function and ability to enter the cell cycle by cotransduction with adenoviruses encoding cell cycle regulators cdk6 and cyclin D3. Using this approach, we tested 12 previously identified mitogens, including neurotransmitters, hormones, growth factors, and molecules, involved in adenosine and Tgf-1β signaling. Each compound was tested in a wide concentration range either in the presence of basal (5 mM) or high (11 mM) glucose. Treatment with the control compound harmine, a Dyrk1a inhibitor, led to a significant increase in Ki-67+ β-cells, whereas treatment with other compounds had limited to no effect on human β-cell proliferation. This new scalable approach reduces the time and effort required for sensitive and specific evaluation of human β-cell proliferation, thus allowing for increased testing of candidate human β-cell mitogens.
Collapse
Affiliation(s)
- Kristie I Aamodt
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Radhika Aramandla
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Judy J Brown
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nathalie Fiaschi-Taesch
- Division of Endocrinology, Diabetes, and Bone Disease, Department of Medicine, Mount Sinai Medical Center, New York, New York; and
| | - Peng Wang
- Division of Endocrinology, Diabetes, and Bone Disease, Department of Medicine, Mount Sinai Medical Center, New York, New York; and
| | - Andrew F Stewart
- Division of Endocrinology, Diabetes, and Bone Disease, Department of Medicine, Mount Sinai Medical Center, New York, New York; and
| | - Marcela Brissova
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
31
|
Benthuysen JR, Carrano AC, Sander M. Advances in β cell replacement and regeneration strategies for treating diabetes. J Clin Invest 2016; 126:3651-3660. [PMID: 27694741 DOI: 10.1172/jci87439] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In the past decade, new approaches have been explored that are aimed at restoring functional β cell mass as a treatment strategy for diabetes. The two most intensely pursued strategies are β cell replacement through conversion of other cell types and β cell regeneration by enhancement of β cell replication. The approach closest to clinical implementation is the replacement of β cells with human pluripotent stem cell-derived (hPSC-derived) cells, which are currently under investigation in a clinical trial to assess their safety in humans. In addition, there has been success in reprogramming developmentally related cell types into β cells. Reprogramming approaches could find therapeutic applications by inducing β cell conversion in vivo or by reprogramming cells ex vivo followed by implantation. Finally, recent studies have revealed novel pharmacologic targets for stimulating β cell replication. Manipulating these targets or the pathways they regulate could be a strategy for promoting the expansion of residual β cells in diabetic patients. Here, we provide an overview of progress made toward β cell replacement and regeneration and discuss promises and challenges for clinical implementation of these strategies.
Collapse
|
32
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
33
|
Saunders D, Powers AC. Replicative capacity of β-cells and type 1 diabetes. J Autoimmun 2016; 71:59-68. [PMID: 27133598 DOI: 10.1016/j.jaut.2016.03.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 03/28/2016] [Indexed: 12/16/2022]
Abstract
Efforts to restore β-cell number or mass in type 1 diabetes (T1D) must combine an intervention to stimulate proliferation of remaining β-cells and an intervention to mitigate or control the β-cell-directed autoimmunity. This review highlights features of the β-cell, including it being part of a pancreatic islet, a mini-organ that is highly vascularized and highly innervated, and efforts to promote β-cell proliferation. In addition, the β-cell in T1D exists in a microenvironment with interactions and input from other islet cell types, extracellular matrix, vascular endothelial cells, neuronal projections, and immune cells, all of which likely influence the β-cell's capacity for replication. Physiologic β-cell proliferation occurs in human and rodents in the neonatal period and early in life, after which there is an age-dependent decline in β-cell proliferation, and also as part of the β-cell's compensatory response to the metabolic challenges of pregnancy and insulin resistance. This review reviews the molecular pathways involved in this β-cell proliferation and highlights recent work in two areas: 1) Investigators, using high-throughput screening to discover small molecules that promote human β-cell proliferation, are now focusing on the dual-specificity tyrosine-regulated kinase-1a and cell cycle-dependent kinase inhibitors CDKN2C/p18 or CDKN1A/p21as targets of compounds to stimulate adult human β-cell proliferation. 2) Local inflammation, macrophages, and the local β-cell microenvironment promote β-cell proliferation. Future efforts to harness the responsible mechanisms may lead to new approaches to promote β-cell proliferation in T1D.
Collapse
Affiliation(s)
- Diane Saunders
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, United States; Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; VA Tennessee Valley Healthcare System, Nashville, TN, United States.
| |
Collapse
|