1
|
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
|
2
|
Grygoryev D, Ekstrom T, Manalo E, Link JM, Alshaikh A, Keith D, Allen-Petersen BL, Sheppard B, Morgan T, Soufi A, Sears RC, Kim J. Sendai virus is robust and consistent in delivering genes into human pancreatic cancer cells. Heliyon 2024; 10:e27221. [PMID: 38463758 PMCID: PMC10923719 DOI: 10.1016/j.heliyon.2024.e27221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly intratumorally heterogeneous disease that includes several subtypes and is highly plastic. Effective gene delivery to all PDAC cells is essential for modulating gene expression and identifying potential gene-based therapeutic targets in PDAC. Most current gene delivery systems for pancreatic cells are optimized for islet or acinar cells. Lentiviral vectors are the current main gene delivery vectors for PDAC, but their transduction efficiencies vary depending on pancreatic cell type, and are especially poor for the classical subtype of PDAC cells from both primary tumors and cell lines. Methods We systemically compare transduction efficiencies of glycoprotein G of vesicular stomatitis virus (VSV-G)-pseudotyped lentiviral and Sendai viral vectors in human normal pancreatic ductal and PDAC cells. Results We find that the Sendai viral vector gives the most robust gene delivery efficiency regardless of PDAC cell type. Therefore, we propose using Sendai viral vectors to transduce ectopic genes into PDAC cells.
Collapse
Affiliation(s)
- Dmytro Grygoryev
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Taelor Ekstrom
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Elise Manalo
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Jason M. Link
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Amani Alshaikh
- The University of Edinburgh, Centre for Regenerative Medicine, Institute of Regeneration and Repair, Institute of Stem Cell Research, Edinburgh, UK
- King Abdulaziz City for Science and Technology, Health Sector (KACST), Riyadh, Saudi Arabia
| | - Dove Keith
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Brittany L. Allen-Petersen
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Brett Sheppard
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
- Department of Surgery, Oregon Health & Science University School of Medicine, USA
| | - Terry Morgan
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
- Department of Pathology, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| | - Abdenour Soufi
- The University of Edinburgh, Centre for Regenerative Medicine, Institute of Regeneration and Repair, Institute of Stem Cell Research, Edinburgh, UK
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| | - Jungsun Kim
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| |
Collapse
|
3
|
Francis M, Bhaskar S, Komanduri S, Sheshadri P, Prasanna J, Kumar A. Deubiquitinase USP1 influences the dedifferentiation of mouse pancreatic β-cells. iScience 2023; 26:106771. [PMID: 37250303 PMCID: PMC10214732 DOI: 10.1016/j.isci.2023.106771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Loss of insulin-secreting β-cells in diabetes may be either due to apoptosis or dedifferentiation of β-cell mass. The ubiquitin-proteasome system comprising E3 ligase and deubiquitinases (DUBs) controls several aspects of β-cell functions. In this study, screening for key DUBs identified USP1 to be specifically involved in dedifferentiation process. Inhibition of USP1 either by genetic intervention or small-molecule inhibitor ML323 restored epithelial phenotype of β-cells, but not with inhibition of other DUBs. In absence of dedifferentiation cues, overexpression of USP1 was sufficient to induce dedifferentiation in β-cells; mechanistic insight showed USP1 to mediate its effect via modulating the expression of inhibitor of differentiation (ID) 2. In an in vivo streptozotocin (STZ)-induced dedifferentiation mouse model system, administering ML323 alleviated hyperglycemic state. Overall, this study identifies USP1 to be involved in dedifferentiation of β-cells and its inhibition may have a therapeutic application of reducing β-cell loss during diabetes.
Collapse
Affiliation(s)
- Meenal Francis
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Smitha Bhaskar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Saarwani Komanduri
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Preethi Sheshadri
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
4
|
Liu Y, Liu Z, Hu L, He L, Yang L, Qin Z, Xie Y, Peng X, Dai L. Function of stem cells in radiation-induced damage. Int J Radiat Biol 2023; 99:1483-1494. [PMID: 36912588 DOI: 10.1080/09553002.2023.2188935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/27/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE The aim of this review is to discuss previous studies on the function of stem cells in radiation-induced damage, and the factors affecting these processes, in the hope of improving our understanding of the different stem cells and the communication networks surrounding them. This is essential for the development of effective stem cell-based therapies to regenerate or replace normal tissues damaged by radiation. CONCLUSION In salivary glands, senescence-associated cytokines and inflammation-associated cells have a greater effect on stem cells. In the intestinal glands, Paneth cells strongly affect stem cell-mediated tissue regeneration after radiation treatment. In the pancreas, β-cells as well as protein C receptor positive (Procr) cells are expected to be key cells in the treatment of diabetes. In the bone marrow, a variety of cytokines such as CXC-chemokine ligand 12 (CXCL12) and stem cell factor (SCF), contribute to the functional recovery of hematopoietic stem cells after irradiation.
Collapse
Affiliation(s)
- Yingtong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Hu
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ling He
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lianlian Yang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuping Xie
- Department of Oncology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Cell Replacement Therapy for Type 1 Diabetes Patients: Potential Mechanisms Leading to Stem-Cell-Derived Pancreatic β-Cell Loss upon Transplant. Cells 2023; 12:cells12050698. [PMID: 36899834 PMCID: PMC10000642 DOI: 10.3390/cells12050698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cell replacement therapy using stem-cell-derived insulin-producing β-like cells (sBCs) has been proposed as a practical cure for patients with type one diabetes (T1D). sBCs can correct diabetes in preclinical animal models, demonstrating the promise of this stem cell-based approach. However, in vivo studies have demonstrated that most sBCs, similarly to cadaveric human islets, are lost upon transplantation due to ischemia and other unknown mechanisms. Hence, there is a critical knowledge gap in the current field concerning the fate of sBCs upon engraftment. Here we review, discuss effects, and propose additional potential mechanisms that could contribute toward β-cell loss in vivo. We summarize and highlight some of the literature on phenotypic loss in β-cells under both steady, stressed, and diseased diabetic conditions. Specifically, we focus on β-cell death, dedifferentiation into progenitors, trans-differentiation into other hormone-expressing cells, and/or interconversion into less functional β-cell subtypes as potential mechanisms. While current cell replacement therapy efforts employing sBCs carry great promise as an abundant cell source, addressing the somewhat neglected aspect of β-cell loss in vivo will further accelerate sBC transplantation as a promising therapeutic modality that could significantly enhance the life quality of T1D patients.
Collapse
|
6
|
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
|
7
|
Iuliano C, Absmaier-Kijak M, Sinnberg T, Hoffard N, Hils M, Köberle M, Wölbing F, Shumilina E, Heise N, Fehrenbacher B, Schaller M, Lang F, Kaesler S, Biedermann T. Fetal Tissue-Derived Mast Cells (MC) as Experimental Surrogate for In Vivo Connective Tissue MC. Cells 2022; 11:cells11060928. [PMID: 35326379 PMCID: PMC8946182 DOI: 10.3390/cells11060928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
Bone-marrow-derived mast cells are matured from bone marrow cells in medium containing 20% fetal calf serum (FCS), interleukin (IL)-3 and stem-cell factor (SCF) and are used as in vitro models to study mast cells (MC) and their role in health and disease. In vivo, however, BM-derived hematopoietic stem cells account for only a fraction of MC; the majority of MC in vivo are and remain tissue resident. In this study we established a side-by-side culture with BMMC, fetal skin MC (FSMC) or fetal liver MC (FLMC) for comparative studies to identify the best surrogates for mature connective tissue MC (CTMC). All three MC types showed comparable morphology by histology and MC phenotype by flow cytometry. Heterogeneity was detected in the transcriptome with the most differentially expressed genes in FSMC compared to BMMC being Hdc and Tpsb2. Expression of ST2 was highly expressed in BMMC and FSMC and reduced in FLMC, diminishing their secretion of type 2 cytokines. Higher granule content, stronger response to FcεRI activation and significantly higher release of histamine from FSMC compared to FLMC and BMMC indicated differences in MC development in vitro dependent on the tissue of origin. Thus, tissues of origin imprint MC precursor cells to acquire distinct phenotypes and signatures despite identical culture conditions. Fetal-derived MC resemble mature CTMC, with FSMC being the most developed.
Collapse
Affiliation(s)
- Caterina Iuliano
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
| | - Magdalena Absmaier-Kijak
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
| | - Tobias Sinnberg
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.F.); (M.S.)
| | - Nils Hoffard
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
| | - Miriam Hils
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
| | - Martin Köberle
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
| | - Florian Wölbing
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
| | - Ekaterina Shumilina
- Department of Physiology, University of Tübingen, 72076 Tübingen, Germany; (E.S.); (N.H.); (F.L.)
| | - Nicole Heise
- Department of Physiology, University of Tübingen, 72076 Tübingen, Germany; (E.S.); (N.H.); (F.L.)
| | - Birgit Fehrenbacher
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.F.); (M.S.)
| | - Martin Schaller
- Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany; (T.S.); (B.F.); (M.S.)
| | - Florian Lang
- Department of Physiology, University of Tübingen, 72076 Tübingen, Germany; (E.S.); (N.H.); (F.L.)
| | - Susanne Kaesler
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
- Correspondence: (S.K.); (T.B.); Tel.: +49-89-4141-3170 (S.K. & T.B.); Fax: 49-89-4141-3171 (S.K. & T.B.)
| | - Tilo Biedermann
- Department of Dermatology and Allergology, School of Medicine, Technical University Munich, Biedersteiner Str. 29, 80802 Munich, Germany; (C.I.); (M.A.-K.); (N.H.); (M.H.); (M.K.); (F.W.)
- Correspondence: (S.K.); (T.B.); Tel.: +49-89-4141-3170 (S.K. & T.B.); Fax: 49-89-4141-3171 (S.K. & T.B.)
| |
Collapse
|
8
|
Wang HL, Wang L, Zhao CY, Lan HY. Role of TGF-Beta Signaling in Beta Cell Proliferation and Function in Diabetes. Biomolecules 2022; 12:373. [PMID: 35327565 PMCID: PMC8945211 DOI: 10.3390/biom12030373] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 12/27/2022] Open
Abstract
Beta (β) cell dysfunction or loss is the common pathological feature in all types of diabetes mellitus (diabetes). Resolving the underlying mechanism may facilitate the treatment of diabetes by preserving the β cell population and function. It is known that TGF-β signaling plays diverse roles in β cell development, function, proliferation, apoptosis, and dedifferentiation. Inhibition of TGF-β signaling expands β cell lineage in the development. However, deletion of Tgfbr1 has no influence on insulin demand-induced but abolishes inflammation-induced β cell proliferation. Among canonical TGF-β signaling, Smad3 but not Smad2 is the predominant repressor of β cell proliferation in response to systemic insulin demand. Deletion of Smad3 simultaneously improves β cell function, apoptosis, and systemic insulin resistance with the consequence of eliminated overt diabetes in diabetic mouse models, revealing Smad3 as a key mediator and ideal therapeutic target for type-2 diabetes. However, Smad7 shows controversial effects on β cell proliferation and glucose homeostasis in animal studies. On the other hand, overexpression of Tgfb1 prevents β cells from autoimmune destruction without influence on β cell function. All these findings reveal the diverse regulatory roles of TGF-β signaling in β cell biology.
Collapse
Affiliation(s)
- Hong-Lian Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (H.-L.W.); (L.W.)
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (H.-L.W.); (L.W.)
| | - Chang-Ying Zhao
- Department of Endocrinology, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China;
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Guangdong Academy of Sciences, Guangdong Provincial People’s Hospital Joint Research Laboratory on Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
9
|
Title AC, Karsai M, Mir-Coll J, Grining ÖY, Rufer C, Sonntag S, Forschler F, Jawurek S, Klein T, Yesildag B. Evaluation of the Effects of Harmine on β-cell Function and Proliferation in Standardized Human Islets Using 3D High-Content Confocal Imaging and Automated Analysis. Front Endocrinol (Lausanne) 2022; 13:854094. [PMID: 35860702 PMCID: PMC9289187 DOI: 10.3389/fendo.2022.854094] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/10/2022] [Indexed: 01/09/2023] Open
Abstract
Restoration of β-cell mass through the induction of proliferation represents an attractive therapeutic approach for the treatment of diabetes. However, intact and dispersed primary islets suffer from rapidly deteriorating viability and function ex vivo, posing a significant challenge for their experimental use in proliferation studies. Here, we describe a novel method for the assessment of compound effects on β-cell proliferation and count using reaggregated primary human islets, or islet microtissues (MTs), which display homogeneous size and tissue architecture as well as robust and stable functionality and viability for 4 weeks in culture. We utilized this platform to evaluate the dose-dependent short- and long-term effects of harmine on β-cell proliferation and function. Following compound treatment and EdU incorporation, islet MTs were stained and confocal-imaged for DAPI (nuclear marker), NKX6.1 (β-cell marker), and EdU (proliferation marker), allowing automated 3D-analysis of number of total cells, β-cells, and proliferating β- and non-β-cells per islet MT. In parallel, insulin secretion, intracellular insulin and ATP contents, and Caspase 3/7 activity were analyzed to obtain a comprehensive overview of islet MT function and viability. We observed that 4-day harmine treatment increased β- and non-β-cell proliferation, NKX6.1 expression, and basal and stimulated insulin secretion in a dose-dependent manner, while fold-stimulation of secretion peaked at intermediate harmine doses. Interestingly, 15-day harmine treatment led to a general reduction in harmine's proliferative effects as well as altered dose-dependent trends. The described methodology provides a unique tool for in vitro high-throughput evaluation of short- and long-term changes in human β-cell proliferation, count and fraction along with a variety of functional parameters, in a representative 3D human islet model.
Collapse
Affiliation(s)
| | - Maria Karsai
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | - Joan Mir-Coll
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | | | - Chantal Rufer
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | | | | | - Sayro Jawurek
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | - Thomas Klein
- Department of Cardio-Metabolic Diseases, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Burcak Yesildag
- Diabetes Research, InSphero AG, Schlieren, Switzerland
- *Correspondence: Burcak Yesildag,
| |
Collapse
|
10
|
Cai Z, Liu F, Yang Y, Li D, Hu S, Song L, Yu S, Li T, Liu B, Luo H, Zhang W, Zhou Z, Zhang J. GRB10 regulates β cell mass by inhibiting β cell proliferation and stimulating β cell dedifferentiation. J Genet Genomics 2021; 49:208-216. [PMID: 34861413 DOI: 10.1016/j.jgg.2021.11.006] [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: 05/31/2021] [Revised: 11/07/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Abstract
Decreased functional β-cell mass is the hallmark of diabetes, but the cause of this metabolic defect remains elusive. Here, we show that the expression levels of the growth factor receptor-bound protein 10 (GRB10), a negative regulator of insulin and mTORC1 signaling, are markedly induced in islets of diabetic mice and high glucose-treated insulinoma cell line INS-1cells. β-cell-specific knockout of Grb10 in mice increased β-cell mass and improved β-cell function. Grb10-deficient β-cells exhibit enhanced mTORC1 signaling and reduced β-cell dedifferentiation, which could be blocked by rapamycin. On the contrary, Grb10 overexpression induced β-cell dedifferentiation in MIN6 cells. Our study identifies GRB10 as a critical regulator of β-cell dedifferentiation and β-cell mass, which exerts its effect by inhibiting mTORC1 signaling.
Collapse
Affiliation(s)
- Zixin Cai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Fen Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yan Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Dandan Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Shanbiao Hu
- Department of Urological Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lei Song
- Department of Urological Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shaojie Yu
- Department of Urological Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ting Li
- Department of Liver Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Bilian Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Weiping Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
11
|
Title AC, Silva PN, Godbersen S, Hasenöhrl L, Stoffel M. The miR-200-Zeb1 axis regulates key aspects of β-cell function and survival in vivo. Mol Metab 2021; 53:101267. [PMID: 34116231 PMCID: PMC8258987 DOI: 10.1016/j.molmet.2021.101267] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The miR-200-Zeb1 axis regulates the epithelial-to-mesenchymal transition (EMT), differentiation, and resistance to apoptosis. A better understanding of these processes in diabetes is highly relevant, as β-cell dedifferentiation and apoptosis contribute to the loss of functional β-cell mass and diabetes progression. Furthermore, EMT promotes the loss of β-cell identity in the in vitro expansion of human islets. Though the miR-200 family has previously been identified as a regulator of β-cell apoptosis in vivo, studies focusing on Zeb1 are lacking. The aim of this study was thus to investigate the role of Zeb1 in β-cell function and survival in vivo. METHODS miR-200 and Zeb1 are involved in a double-negative feedback loop. We characterized a mouse model in which miR-200 binding sites in the Zeb1 3'UTR are mutated (Zeb1200), leading to a physiologically relevant upregulation of Zeb1 mRNA expression. The role of Zeb1 was investigated in this model via metabolic tests and analysis of isolated islets. Further insights into the distinct contributions of the miR-200 and Zeb1 branches of the feedback loop were obtained by crossing the Zeb1200 allele into a background of miR-141-200c overexpression. RESULTS Mild Zeb1 derepression in vivo led to broad transcriptional changes in islets affecting β-cell identity, EMT, insulin secretion, cell-cell junctions, the unfolded protein response (UPR), and the response to ER stress. The aggregation and insulin secretion of dissociated islets of mice homozygous for the Zeb1200 mutation (Zeb1200M) were impaired, and Zeb1200M islets were resistant to thapsigargin-induced ER stress ex vivo. Zeb1200M mice had increased circulating proinsulin levels but no overt metabolic phenotype, reflecting the strong compensatory ability of islets to maintain glucose homeostasis. CONCLUSIONS This study signifies the importance of the miR-200-Zeb1 axis in regulating key aspects of β-cell function and survival. A better understanding of this axis is highly relevant in developing therapeutic strategies for inducing β-cell redifferentiation and maintaining β-cell identity in in vitro islet expansion.
Collapse
Affiliation(s)
- Alexandra C Title
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland; Competence Center Personalized Medicine, ETH Zürich, Voltastrasse 24, 8044, Zürich, Switzerland
| | - Pamuditha N Silva
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Svenja Godbersen
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Lynn Hasenöhrl
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Markus Stoffel
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland; Competence Center Personalized Medicine, ETH Zürich, Voltastrasse 24, 8044, Zürich, Switzerland; Medical Faculty, University of Zürich, 8091, Zürich, Switzerland.
| |
Collapse
|
12
|
Czernichow P, Reynaud K, Ravassard P. Production and Characterization of a Conditionally Immortalized Dog Beta-Cell Line from Fetal Canine Pancreas. Cell Transplant 2021; 29:963689720971204. [PMID: 33150791 PMCID: PMC7784601 DOI: 10.1177/0963689720971204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Since the 1970s, rodent and human insulin-secreting pancreatic beta-cell lines have been developed and found useful for studying beta-cell biology. Surprisingly, although the dog has been widely used as a translational model for diabetes, no canine insulin-secreting beta cells have ever been produced. Here, a targeted oncogenesis protocol previously described by some of us for generating human beta cells was adapted to produce canine beta cells. Canine fetal pancreata were obtained by cesarean section between 42 and 55 days of gestation, and fragments of fetal glands were transduced with a lentiviral vector expressing SV40LT under the control of the insulin promoter. Two Lox P sites flanking the sequence allowed subsequent transgene excision by Cre recombinase expression. When grafted into SCID mice, these transduced pancreata formed insulinomas. ACT-164 is the cell line described in this report. Insulin mRNA expression and protein content were lower than reported with adult cells, but the ACT-164 cells were functional, and their insulin production in vitro increased under glucose stimulation. Transgene excision upon Cre expression arrested proliferation and enhanced insulin expression and production. When grafted in SCID mice, intact and excised cells reversed chemically induced diabetes. We have thus produced an excisable canine beta-cell line. These cells may play an important role in the study of several aspects of the cell transplantation procedure including the encapsulation process, which is difficult to investigate in rodents. Although much more work is needed to improve the excision procedure and achieve 100% removal of large T antigen expression, we have shown that functional cells can be obtained and might in the future be used for replacement therapy in diabetic dogs.
Collapse
Affiliation(s)
- P Czernichow
- Animal Cell Therapy, Sorbonne Universités, Campus des Cordeliers, Paris, France
| | - K Reynaud
- Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France.,PRC, UMR INRA0085, CNRS 7247, Centre INRA Val de Loire, Nouzilly, France
| | - P Ravassard
- Paris Brain Institute (ICM) Sorbonne Universités, Inserm, CNRS - Hôpital Pitié-Salpêtrière, Boulevard de l'Hôpital, Paris, France
| |
Collapse
|
13
|
Amanzougaghene N, Tajeri S, Yalaoui S, Lorthiois A, Soulard V, Gego A, Rametti A, Risco-Castillo V, Moreno A, Tefit M, van Gemert GJ, Sauerwein RW, Vaillant JC, Ravassard P, Pérignon JL, Froissard P, Mazier D, Franetich JF. The Host Protein Aquaporin-9 is Required for Efficient Plasmodium falciparum Sporozoite Entry into Human Hepatocytes. Front Cell Infect Microbiol 2021; 11:704662. [PMID: 34268141 PMCID: PMC8276244 DOI: 10.3389/fcimb.2021.704662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocyte invasion by Plasmodium sporozoites represents a promising target for innovative antimalarial therapy, but the molecular events mediating this process are still largely uncharacterized. We previously showed that Plasmodium falciparum sporozoite entry into hepatocytes strictly requires CD81. However, CD81-overexpressing human hepatoma cells remain refractory to P. falciparum infection, suggesting the existence of additional host factors necessary for sporozoite entry. Here, through differential transcriptomic analysis of human hepatocytes and hepatoma HepG2-CD81 cells, the transmembrane protein Aquaporin-9 (AQP9) was found to be among the most downregulated genes in hepatoma cells. RNA silencing showed that sporozoite invasion of hepatocytes requires AQP9 expression. AQP9 overexpression in hepatocytes increased their permissiveness to P. falciparum. Moreover, chemical disruption with the AQP9 inhibitor phloretin markedly inhibited hepatocyte infection. Our findings identify AQP9 as a novel host factor required for P. falciparum sporozoite hepatocyte-entry and indicate that AQP9 could be a potential therapeutic target.
Collapse
Affiliation(s)
- Nadia Amanzougaghene
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Shahin Tajeri
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Samir Yalaoui
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Audrey Lorthiois
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Valérie Soulard
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Audrey Gego
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Armelle Rametti
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | | | - Alicia Moreno
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Maurel Tefit
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Jean-Christophe Vaillant
- AP-HP, Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Philippe Ravassard
- CR-ICM - LGN CNRS UMR-7991, IFR des Neurosciences, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jean-Louis Pérignon
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Patrick Froissard
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Dominique Mazier
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| |
Collapse
|
14
|
Maachi H, Ghislain J, Tremblay C, Poitout V. Pronounced proliferation of non-beta cells in response to beta-cell mitogens in isolated human islets of Langerhans. Sci Rep 2021; 11:11283. [PMID: 34050242 PMCID: PMC8163757 DOI: 10.1038/s41598-021-90643-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 05/06/2021] [Indexed: 11/09/2022] Open
Abstract
The potential to treat diabetes by increasing beta-cell mass is driving a major effort to identify beta-cell mitogens. Demonstration of mitogen activity in human beta cells is frequently performed in ex vivo assays. However, reported disparities in the efficacy of beta-cell mitogens led us to investigate the sources of this variability. We studied 35 male (23) and female (12) human islet batches covering a range of donor ages and BMI. Islets were kept intact or dispersed into single cells and cultured in the presence of harmine, glucose, or heparin-binding epidermal growth factor-like growth factor (HB-EGF), and subsequently analyzed by immunohistochemistry or flow cytometry. Proliferating cells were identified by double labeling with EdU and Ki67 and glucagon, c-peptide or Nkx6.1, and cytokeratin-19 to respectively label alpha, beta, and ductal cells. Harmine and HB-EGF stimulated human beta-cell proliferation, but the effect of glucose was dependent on the assay and the donor. Harmine potently stimulated alpha-cell proliferation and both harmine and HB-EGF increased proliferation of insulin- and glucagon-negative cells, including cytokeratin 19-positive cells. Given the abundance of non-beta cells in human islet preparations, our results suggest that assessment of beta-cell mitogens requires complementary approaches and rigorous identification of cell identity using multiple markers.
Collapse
Affiliation(s)
- Hasna Maachi
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada.,Department of Pharmacology and Physiology, University of Montreal, Montreal, QC, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada. .,Department of Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
15
|
Ma J, Xing B, Cao Y, He X, Bennett KE, Tong C, An C, Hojnacki T, Feng Z, Deng S, Ling S, Xie G, Wu Y, Ren Y, Yu M, Katona BW, Li H, Naji A, Hua X. Menin-regulated Pbk controls high fat diet-induced compensatory beta cell proliferation. EMBO Mol Med 2021; 13:e13524. [PMID: 33821572 PMCID: PMC8103087 DOI: 10.15252/emmm.202013524] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/07/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic beta cells undergo compensatory proliferation in the early phase of type 2 diabetes. While pathways such as FoxM1 are involved in regulating compensatory beta cell proliferation, given the lack of therapeutics effectively targeting beta cell proliferation, other targetable pathways need to be identified. Herein, we show that Pbk, a serine/threonine protein kinase, is essential for high fat diet (HFD)‐induced beta cell proliferation in vivo using a Pbk kinase deficiency knock‐in mouse model. Mechanistically, JunD recruits menin and HDAC3 complex to the Pbk promoter to reduce histone H3 acetylation, leading to epigenetic repression of Pbk expression. Moreover, menin inhibitor (MI) disrupts the menin–JunD interaction and augments Pbk transcription. Importantly, MI administration increases beta cell proliferation, ameliorating hyperglycemia, and impaired glucose tolerance (IGT) in HFD‐induced diabetic mice. Notably, Pbk is required for the MI‐induced beta cell proliferation and improvement of IGT. Together, these results demonstrate the repressive role of the menin/JunD/Pbk axis in regulating HFD‐induced compensatory beta cell proliferation and pharmacologically regulating this axis may serve as a novel strategy for type 2 diabetes therapy.
Collapse
Affiliation(s)
- Jian Ma
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bowen Xing
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yan Cao
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Xin He
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kate E Bennett
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Chao Tong
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiying An
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Taylor Hojnacki
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zijie Feng
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sunbin Deng
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunbin Ling
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gengchen Xie
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuan Wu
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ming Yu
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Hongzhe Li
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ali Naji
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Xianxin Hua
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
16
|
Efrat S. Epigenetic Memory: Lessons From iPS Cells Derived From Human β Cells. Front Endocrinol (Lausanne) 2021; 11:614234. [PMID: 33584546 PMCID: PMC7874102 DOI: 10.3389/fendo.2020.614234] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/02/2020] [Indexed: 11/18/2022] Open
Abstract
Incomplete reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) may be responsible for the heterogeneity in differentiation capacity observed among iPSC lines. It remains unclear whether it results from stochastic reprogramming events, or reflects consistent genetic or cell-of-origin differences. Some evidence suggests that epigenetic memory predisposes iPSCs to enhanced differentiation into the parental cell type. We investigated iPSCs reprogrammed from human pancreatic islet β cells (BiPSCs), as a step in development of a robust differentiation protocol for generation of β-like cells. BiPSCs derived from multiple human donors manifested enhanced and reproducible spontaneous and induced differentiation towards insulin-producing cells, compared with iPSCs derived from isogenic non-β-cell types and fibroblast-derived iPSCs (FiPSCs). Genome-wide analyses of open chromatin in BiPSCs and FiPSCs identified thousands of differential open chromatin sites (DOCs) between the two iPSC types. DOCs more open in BiPSCs (Bi-DOCs) were significantly enriched for known regulators of endodermal development, including bivalent and weak enhancers, and FOXA2 binding sites. Bi-DOCs were associated with genes related to pancreas development and β-cell function. These studies provide evidence for reproducible epigenetic memory in BiPSCs. Bi-DOCs may provide clues to genes and pathways involved in the differentiation process, which could be manipulated to increase the efficiency and reproducibility of differentiation of pluripotent stem cells from non-β-cell sources.
Collapse
Affiliation(s)
- Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Wang D, Wang J, Bai L, Pan H, Feng H, Clevers H, Zeng YA. Long-Term Expansion of Pancreatic Islet Organoids from Resident Procr + Progenitors. Cell 2020; 180:1198-1211.e19. [PMID: 32200801 DOI: 10.1016/j.cell.2020.02.048] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/03/2019] [Accepted: 02/21/2020] [Indexed: 12/31/2022]
Abstract
It has generally proven challenging to produce functional β cells in vitro. Here, we describe a previously unidentified protein C receptor positive (Procr+) cell population in adult mouse pancreas through single-cell RNA sequencing (scRNA-seq). The cells reside in islets, do not express differentiation markers, and feature epithelial-to-mesenchymal transition characteristics. By genetic lineage tracing, Procr+ islet cells undergo clonal expansion and generate all four endocrine cell types during adult homeostasis. Sorted Procr+ cells, representing ∼1% of islet cells, can robustly form islet-like organoids when cultured at clonal density. Exponential expansion can be maintained over long periods by serial passaging, while differentiation can be induced at any time point in culture. β cells dominate in differentiated islet organoids, while α, δ, and PP cells occur at lower frequencies. The organoids are glucose-responsive and insulin-secreting. Upon transplantation in diabetic mice, these organoids reverse disease. These findings demonstrate that the adult mouse pancreatic islet contains a population of Procr+ endocrine progenitors.
Collapse
Affiliation(s)
- Daisong Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingqiang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lanyue Bai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hong Pan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua Feng
- Omics Core, Bio-Med Big Data Center, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hans Clevers
- Hubrecht Institute and Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht, Utrecht, the Netherlands; Utrecht University and Princess Maxima Center, Utrecht, the Netherlands
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
18
|
Zhang J, Liu F. The De-, Re-, and trans-differentiation of β-cells: Regulation and function. Semin Cell Dev Biol 2020; 103:68-75. [DOI: 10.1016/j.semcdb.2020.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/09/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
|
19
|
Qadir MMF, Álvarez-Cubela S, Weitz J, Panzer JK, Klein D, Moreno-Hernández Y, Cechin S, Tamayo A, Almaça J, Hiller H, Beery M, Kusmartseva I, Atkinson M, Speier S, Ricordi C, Pugliese A, Caicedo A, Fraker CA, Pastori RL, Domínguez-Bendala J. Long-term culture of human pancreatic slices as a model to study real-time islet regeneration. Nat Commun 2020; 11:3265. [PMID: 32601271 PMCID: PMC7324563 DOI: 10.1038/s41467-020-17040-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 06/04/2020] [Indexed: 01/02/2023] Open
Abstract
The culture of live pancreatic tissue slices is a powerful tool for the interrogation of physiology and pathology in an in vitro setting that retains near-intact cytoarchitecture. However, current culture conditions for human pancreatic slices (HPSs) have only been tested for short-term applications, which are not permissive for the long-term, longitudinal study of pancreatic endocrine regeneration. Using a culture system designed to mimic the physiological oxygenation of the pancreas, we demonstrate high viability and preserved endocrine and exocrine function in HPS for at least 10 days after sectioning. This extended lifespan allowed us to dynamically lineage trace and quantify the formation of insulin-producing cells in HPS from both non-diabetic and type 2 diabetic donors. This technology is expected to be of great impact for the conduct of real-time regeneration/developmental studies in the human pancreas.
Collapse
Affiliation(s)
- Mirza Muhammad Fahd Qadir
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Jonathan Weitz
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Julia K Panzer
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Yaisa Moreno-Hernández
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Universidad Francisco de Vitoria, Madrid, Spain
| | - Sirlene Cechin
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Tamayo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Joana Almaça
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Helmut Hiller
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Beery
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Irina Kusmartseva
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Mark Atkinson
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Institute of Physiology, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alberto Pugliese
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Caicedo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Christopher A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
20
|
Téllez N, Montanya E. Determining Beta Cell Mass, Apoptosis, Proliferation, and Individual Beta Cell Size in Pancreatic Sections. Methods Mol Biol 2020; 2128:313-337. [PMID: 32180202 DOI: 10.1007/978-1-0716-0385-7_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pancreatic beta cells have a significant remodeling capacity which plays an essential role in the maintenance of glucose homeostasis. Beta cell apoptosis, replication, size, dedifferentiation, and (neo)generation contribute to the beta cell mass regulation. However, the extent of their respective contribution varies significantly depending on the specific condition, and it is the balance among them that determines the eventual change in beta cell mass. Thus, the study of the pancreatic beta cell mass regulation requires the determination of all these factors. In this chapter, we describe the quantification of beta cell replication based on the incorporation of thymidine analogs into replicated DNA strands and on the expression of Ki67 antigen and phosphorylation of histone H3. Beta cell apoptosis is analyzed by the TUNEL technique, and beta cell mass and cross-sectional area of individual beta cells are determined by computerized image processing methods.
Collapse
Affiliation(s)
- Noèlia Téllez
- CIBER of Diabetes and Metabolic Diseases, CIBERDEM, Barcelona, Spain. .,Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain. .,University of Barcelona, Barcelona, Spain.
| | - Eduard Montanya
- CIBER of Diabetes and Metabolic Diseases, CIBERDEM, Barcelona, Spain.,Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.,University of Barcelona, Barcelona, Spain.,Endocrine Unit, Hospital Universitari de Bellvitge, Barcelona, Spain
| |
Collapse
|
21
|
Dayem AA, Lee SB, Kim K, Lim KM, Jeon TI, Cho SG. Recent advances in organoid culture for insulin production and diabetes therapy: methods and challenges. BMB Rep 2019. [PMID: 30940326 PMCID: PMC6549913 DOI: 10.5483/bmbrep.2019.52.5.089] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Breakthroughs in stem cell technology have contributed to disease modeling and drug screening via organoid technology. Organoid are defined as three-dimensional cellular aggregations derived from adult tissues or stem cells. They recapitulate the intricate pattern and functionality of the original tissue. Insulin is secreted mainly by the pancreatic β cells. Large-scale production of insulin-secreting β cells is crucial for diabetes therapy. Here, we provide a brief overview of organoids and focus on recent advances in protocols for the generation of pancreatic islet organoids from pancreatic tissue or pluripotent stem cells for insulin secretion. The feasibility and limitations of organoid cultures derived from stem cells for insulin production will be described. As the pancreas and gut share the same embryological origin and produce insulin, we will also discuss the possible application of gut organoids for diabetes therapy. Better understanding of the challenges associated with the current protocols for organoid culture facilitates development of scalable organoid cultures for applications in biomedicine.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Soo Bin Lee
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Kyung Min Lim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Tak-Il Jeon
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
22
|
Highly efficient ex vivo lentiviral transduction of primary human pancreatic exocrine cells. Sci Rep 2019; 9:15870. [PMID: 31676849 PMCID: PMC6825235 DOI: 10.1038/s41598-019-51763-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/30/2019] [Indexed: 01/09/2023] Open
Abstract
The lack of efficient gene transfer methods into primary human pancreatic exocrine cells hampers studies on the plasticity of these cells and their possible role in beta cell regeneration. Therefore, improved gene transfer protocols are needed. Lentiviral vectors are widely used to drive ectopic gene expression in mammalian cells, including primary human islet cells. Here we aimed to optimize gene transfer into primary human exocrine cells using modified lentiviral vectors or transduction conditions. We evaluated different promoters, viral envelopes, medium composition and transduction adjuvants. Transduction efficiency of a reporter vector was evaluated by fluorescence microscopy and flow cytometry. We show that protamine sulfate-assisted transduction of a VSV-G-pseudotyped vector expressing eGFP under the control of a CMV promoter in a serum-free environment resulted in the best transduction efficiency of exocrine cells, reaching up to 90% of GFP-positive cells 5 days after transduction. Our findings will enable further studies on pancreas (patho)physiology that require gene transfer such as gene overexpression, gene knockdown or lineage tracing studies.
Collapse
|
23
|
Lenz A, Lenz G, Ku HT, Ferreri K, Kandeel F. Islets from human donors with higher but not lower hemoglobin A1c levels respond to gastrin treatment in vitro. PLoS One 2019; 14:e0221456. [PMID: 31430329 PMCID: PMC6701795 DOI: 10.1371/journal.pone.0221456] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Gastrin is a peptide hormone, which in combination with other factors such as TGFα, EGF or GLP-1, is capable of increasing beta cell mass and lowering blood glucose levels in adult diabetic mice. In humans, administration of a bolus of gastrin alone induces insulin secretion suggesting that gastrin may target islet cells. However, whether gastrin alone is sufficient to exert an effect on isolated human islets has been controversial and the mechanism remained poorly understood. Therefore, in this study we started to examine the effects of gastrin alone on cultured adult human islets. Treatment of isolated human islets with gastrin I for 48 h resulted in increased expression of insulin, glucagon and somatostatin transcripts. These increases were significantly correlated with the levels of donor hemoglobin A1c (HbA1c) but not BMI or age. In addition, gastrin treatment resulted in increased expression of PDX1, NKX6.1, NKX2.2, MNX1 and HHEX in islets from donors with HbA1c greater than 42 mmol/mol. The addition of YM022, an antagonist of the gastrin receptor cholecystokinin B receptor (CCKBR), together with gastrin eliminated these effects, verifying that the effects of gastrin are mediated through CCKBR.CCKBR is expressed in somatostatin-expressing delta cells in islets from all donors. However, in the islets from donors with higher HbA1c (greater than 42 mmol/mol [6.0%]), cells triple-positive for CCKBR, somatostatin and insulin were detected, suggesting a de-differentiation or trans-differentiation of endocrine cells. Our results demonstrate a direct effect of gastrin on human islets from prediabetic or diabetic individuals that is mediated through CCKBR+ cells. Further, our data imply that gastrin may be a potential treatment for diabetic patients.
Collapse
Affiliation(s)
- Ayelet Lenz
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States of America
- * E-mail:
| | - Gal Lenz
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Kevin Ferreri
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| |
Collapse
|
24
|
Efrat S. Beta-Cell Dedifferentiation in Type 2 Diabetes: Concise Review. Stem Cells 2019; 37:1267-1272. [PMID: 31298804 DOI: 10.1002/stem.3059] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is caused by an inherited predisposition to pancreatic islet β-cell failure, which is manifested under cellular stress induced by metabolic overload. The decrease in the functional β-cell mass associated with T2D has been attributed primarily to β-cell death; however, studies in recent years suggested that β-cell dedifferentiation may contribute to this decline. The mechanisms linking genetic factors and cellular stress to β-cell dedifferentiation remain largely unknown. This study evaluated the evidence for β-cell dedifferentiation in T2D, and T2D and examined experimental systems in which its mechanisms may be studied. Understanding these mechanisms may allow prevention of β-cell dedifferentiation or induction of cell redifferentiation for restoration of the functional β-cell mass. Stem Cells 2019;37:1267-1272.
Collapse
Affiliation(s)
- Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
25
|
Dayem AA, Lee SB, Kim K, Lim KM, Jeon TI, Cho SG. Recent advances in organoid culture for insulin production and diabetes therapy: methods and challenges. BMB Rep 2019; 52:295-303. [PMID: 30940326 PMCID: PMC6549913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 10/13/2023] Open
Abstract
Breakthroughs in stem cell technology have contributed to disease modeling and drug screening via organoid technology. Organoid are defined as three-dimensional cellular aggregations derived from adult tissues or stem cells. They recapitulate the intricate pattern and functionality of the original tissue. Insulin is secreted mainly by the pancreatic β cells. Large-scale production of insulin-secreting β cells is crucial for diabetes therapy. Here, we provide a brief overview of organoids and focus on recent advances in protocols for the generation of pancreatic islet organoids from pancreatic tissue or pluripotent stem cells for insulin secretion. The feasibility and limitations of organoid cultures derived from stem cells for insulin production will be described. As the pancreas and gut share the same embryological origin and produce insulin, we will also discuss the possible application of gut organoids for diabetes therapy. Better understanding of the challenges associated with the current protocols for organoid culture facilitates development of scalable organoid cultures for applications in biomedicine. [BMB Reports 2019; 52(5): 295-303].
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Soo Bin Lee
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Kyung Min Lim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Tak-il Jeon
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| |
Collapse
|
26
|
Loomans CJM, Williams Giuliani N, Balak J, Ringnalda F, van Gurp L, Huch M, Boj SF, Sato T, Kester L, de Sousa Lopes SMC, Roost MS, Bonner-Weir S, Engelse MA, Rabelink TJ, Heimberg H, Vries RGJ, van Oudenaarden A, Carlotti F, Clevers H, de Koning EJP. Expansion of Adult Human Pancreatic Tissue Yields Organoids Harboring Progenitor Cells with Endocrine Differentiation Potential. Stem Cell Reports 2019. [PMID: 29539434 PMCID: PMC5918840 DOI: 10.1016/j.stemcr.2018.02.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Generating an unlimited source of human insulin-producing cells is a prerequisite to advance β cell replacement therapy for diabetes. Here, we describe a 3D culture system that supports the expansion of adult human pancreatic tissue and the generation of a cell subpopulation with progenitor characteristics. These cells display high aldehyde dehydrogenase activity (ALDHhi), express pancreatic progenitors markers (PDX1, PTF1A, CPA1, and MYC), and can form new organoids in contrast to ALDHlo cells. Interestingly, gene expression profiling revealed that ALDHhi cells are closer to human fetal pancreatic tissue compared with adult pancreatic tissue. Endocrine lineage markers were detected upon in vitro differentiation. Engrafted organoids differentiated toward insulin-positive (INS+) cells, and circulating human C-peptide was detected upon glucose challenge 1 month after transplantation. Engrafted ALDHhi cells formed INS+ cells. We conclude that adult human pancreatic tissue has potential for expansion into 3D structures harboring progenitor cells with endocrine differentiation potential.
Collapse
Affiliation(s)
- Cindy J M Loomans
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands; Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Nerys Williams Giuliani
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands; Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jeetindra Balak
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Femke Ringnalda
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - Léon van Gurp
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - Meritxell Huch
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands; Wellcome Trust/Cancer Research UK, Gurdon Institute, Cambridge CB2 1QN, UK
| | - Sylvia F Boj
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - Toshiro Sato
- Department of Gastroenterology, Keio University, Tokyo 108-8345, Japan
| | - Lennart Kester
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | | | - Matthias S Roost
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Susan Bonner-Weir
- Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Marten A Engelse
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Robert G J Vries
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | | | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Hans Clevers
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - Eelco J P de Koning
- Hubrecht Institute/KNAW and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands; Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
27
|
Navarro-Tableros V, Gomez Y, Brizzi MF, Camussi G. Generation of Human Stem Cell-Derived Pancreatic Organoids (POs) for Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:179-220. [PMID: 31025308 DOI: 10.1007/5584_2019_340] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-dependent diabetes mellitus or type 1 diabetes mellitus (T1DM) is an auto-immune condition characterized by the loss of pancreatic β-cells. The curative approach for highly selected patients is the pancreas or the pancreatic islet transplantation. Nevertheless, these options are limited by a growing shortage of donor organs and by the requirement of immunosuppression.Xenotransplantation of porcine islets has been extensively investigated. Nevertheless, the strong xenoimmunity and the risk of transmission of porcine endogenous retroviruses, have limited their application in clinic. Generation of β-like cells from stem cells is one of the most promising strategies in regenerative medicine. Embryonic, and more recently, adult stem cells are currently the most promising cell sources exploited to generate functional β-cells in vitro. A number of studies demonstrated that stem cells could generate functional pancreatic organoids (POs), able to restore normoglycemia when implanted in different preclinical diabetic models. Nevertheless, a gradual loss of function and cell dead are commonly detected when POs are transplanted in immunocompetent animals. So far, the main issue to be solved is the post-transplanted islet loss, due to the host immune attack. To avoid this hurdle, nanotechnology has provided a number of polymers currently under investigation for islet micro and macro-encapsulation. These new approaches, besides conferring PO immune protection, are able to supply oxygen and nutrients and to preserve PO morphology and long-term viability.Herein, we summarize the current knowledge on bioengineered POs and the stem cell differentiation platforms. We also discuss the in vitro strategies used to generate functional POs, and the protocols currently used to confer immune-protection against the host immune attack (micro- and macro-encapsulation). In addition, the most relevant ongoing clinical trials, and the most relevant hurdles met to move towards clinical application are revised.
Collapse
Affiliation(s)
- Victor Navarro-Tableros
- 2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Turin, Turin, Italy
| | - Yonathan Gomez
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy.
- Fondazione per la Ricerca Biomedica-ONLUS, Turin, Italy.
| |
Collapse
|
28
|
Baeyens L, Lemper M, Staels W, De Groef S, De Leu N, Heremans Y, German MS, Heimberg H. (Re)generating Human Beta Cells: Status, Pitfalls, and Perspectives. Physiol Rev 2018; 98:1143-1167. [PMID: 29717931 DOI: 10.1152/physrev.00034.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.
Collapse
Affiliation(s)
- Luc Baeyens
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Marie Lemper
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Willem Staels
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Sofie De Groef
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Nico De Leu
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Yves Heremans
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Michael S German
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| |
Collapse
|
29
|
Cohen H, Barash H, Meivar-Levy I, Molakandov K, Ben-Shimon M, Gurevich M, Zoabi F, Har-Zahav A, Gebhardt R, Gaunitz F, Gurevich M, Mor E, Ravassard P, Greenberger S, Ferber S. The Wnt/β-catenin pathway determines the predisposition and efficiency of liver-to-pancreas reprogramming. Hepatology 2018; 68:1589-1603. [PMID: 29394503 DOI: 10.1002/hep.29827] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 12/30/2017] [Accepted: 01/31/2018] [Indexed: 12/13/2022]
Abstract
UNLABELLED Transdifferentiation (TD) is the direct reprogramming of adult cells into cells of alternate fate and function. We have previously shown that liver cells can be transdifferentiated into beta-like, insulin-producing cells through ectopic expression of pancreatic transcription factors (pTFs). However, the efficiency of the process was consistently limited to <15% of the human liver cells treated in culture. The data in the current study suggest that liver-to-pancreas TD is restricted to a specific population of liver cells that is predisposed to undergo reprogramming. We isolated TD-predisposed subpopulation of liver cells from >15 human donors using a lineage tracing system based on the Wnt response element, part of the pericentral-specific promoter of glutamine synthetase. The cells, that were propagated separately, consistently exhibited efficient fate switch and insulin production and secretion in >60% of the cells upon pTF expression. The rest of the cells, which originated from 85% of the culture, resisted TD. Both populations expressed the ectopic pTFs with similar efficiencies, followed by similar repression of hepatic genes. Our data suggest that the TD-predisposed cells originate from a distinct population of liver cells that are enriched for Wnt signaling, which is obligatory for efficient TD. In TD-resistant populations, Wnt induction is insufficient to induce TD. An additional step of chromatin opening enables TD of these cells. CONCLUSION Liver-to-pancreas TD occurs in defined predisposed cells. These cells' predisposition is maintained by Wnt signaling that endows the cells with the plasticity needed to alter their transcriptional program and developmental fate when triggered by ectopic pTFs. These results may have clinical implications by drastically increasing the efficacy of TD in future clinical uses. (Hepatology 2018).
Collapse
Affiliation(s)
- Helit Cohen
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Hila Barash
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Irit Meivar-Levy
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Kfir Molakandov
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Marina Ben-Shimon
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michael Gurevich
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Fatima Zoabi
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Adi Har-Zahav
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Frank Gaunitz
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Germany
| | - Michael Gurevich
- The Organ Transplantation Division, Schneider Children Medical Center, Petach Tikvah, Israel
| | - Eytan Mor
- The Organ Transplantation Division, Schneider Children Medical Center, Petach Tikvah, Israel
| | - Philippe Ravassard
- Biotechnology and Biotherapy Group, Centre de Recherche, Institut du Cerveau et de la Moelle CNRS UMR7225, INSERM UMRS795, Université Pierre et Marie Curie, Paris, France
| | - Shoshana Greenberger
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| | - Sarah Ferber
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
30
|
The Polycomb-Dependent Epigenome Controls β Cell Dysfunction, Dedifferentiation, and Diabetes. Cell Metab 2018; 27:1294-1308.e7. [PMID: 29754954 PMCID: PMC5989056 DOI: 10.1016/j.cmet.2018.04.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/10/2018] [Accepted: 04/12/2018] [Indexed: 12/28/2022]
Abstract
To date, it remains largely unclear to what extent chromatin machinery contributes to the susceptibility and progression of complex diseases. Here, we combine deep epigenome mapping with single-cell transcriptomics to mine for evidence of chromatin dysregulation in type 2 diabetes. We find two chromatin-state signatures that track β cell dysfunction in mice and humans: ectopic activation of bivalent Polycomb-silenced domains and loss of expression at an epigenomically unique class of lineage-defining genes. β cell-specific Polycomb (Eed/PRC2) loss of function in mice triggers diabetes-mimicking transcriptional signatures and highly penetrant, hyperglycemia-independent dedifferentiation, indicating that PRC2 dysregulation contributes to disease. The work provides novel resources for exploring β cell transcriptional regulation and identifies PRC2 as necessary for long-term maintenance of β cell identity. Importantly, the data suggest a two-hit (chromatin and hyperglycemia) model for loss of β cell identity in diabetes.
Collapse
|
31
|
MicroRNA Expression Analysis of In Vitro Dedifferentiated Human Pancreatic Islet Cells Reveals the Activation of the Pluripotency-Related MicroRNA Cluster miR-302s. Int J Mol Sci 2018; 19:ijms19041170. [PMID: 29649109 PMCID: PMC5979342 DOI: 10.3390/ijms19041170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/06/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022] Open
Abstract
β-cell dedifferentiation has been recently suggested as an additional mechanism contributing to type-1 and to type-2 diabetes pathogenesis. Moreover, several studies demonstrated that in vitro culture of native human pancreatic islets derived from non-diabetic donors resulted in the generation of an undifferentiated cell population. Additional evidence from in vitro human β-cell lineage tracing experiments, demonstrated that dedifferentiated cells derive from β-cells, thus representing a potential in vitro model of β-cell dedifferentiation. Here, we report the microRNA expression profiles analysis of in vitro dedifferentiated islet cells in comparison to mature human native pancreatic islets. We identified 13 microRNAs upregulated and 110 downregulated in islet cells upon in vitro dedifferentiation. Interestingly, among upregulated microRNAs, we observed the activation of microRNA miR-302s cluster, previously defined as pluripotency-associated. Bioinformatic analysis indicated that miR-302s are predicted to target several genes involved in the control of β-cell/epithelial phenotype maintenance; accordingly, such genes were downregulated upon human islet in vitro dedifferentiation. Moreover, we uncovered that cell–cell contacts are needed to maintain low/null expression levels of miR-302. In conclusion, we showed that miR-302 microRNA cluster genes are involved in in vitro dedifferentiation of human pancreatic islet cells and inhibits the expression of multiple genes involved in the maintenance of β-cell mature phenotype.
Collapse
|
32
|
Aloy-Reverté C, Moreno-Amador JL, Nacher M, Montanya E, Semino CE. Use of RGD-Functionalized Sandwich Cultures to Promote Redifferentiation of Human Pancreatic Beta Cells AfterIn VitroExpansion. Tissue Eng Part A 2018; 24:394-406. [DOI: 10.1089/ten.tea.2016.0493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Caterina Aloy-Reverté
- Department of Bioengineering, Tissue Engineering Laboratory, IQS School of Engineering, Barcelona, Spain
| | - José L. Moreno-Amador
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Montserrat Nacher
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Eduard Montanya
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Carlos E. Semino
- Department of Bioengineering, Tissue Engineering Laboratory, IQS School of Engineering, Barcelona, Spain
| |
Collapse
|
33
|
Notch signaling regulates metabolic heterogeneity in glioblastoma stem cells. Oncotarget 2017; 8:64932-64953. [PMID: 29029402 PMCID: PMC5630302 DOI: 10.18632/oncotarget.18117] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) stem cells (GSCs) reside in both hypoxic and vascular microenvironments within tumors. The molecular mechanisms that allow GSCs to occupy such contrasting niches are not understood. We used patient-derived GBM cultures to identify GSC subtypes with differential activation of Notch signaling, which co-exist in tumors but occupy distinct niches and match their metabolism accordingly. Multipotent GSCs with Notch pathway activation reside in perivascular niches, and are unable to entrain anaerobic glycolysis during hypoxia. In contrast, most CD133-expressing GSCs do not depend on canonical Notch signaling, populate tumors regardless of local vascularity and selectively utilize anaerobic glycolysis to expand in hypoxia. Ectopic activation of Notch signaling in CD133-expressing GSCs is sufficient to suppress anaerobic glycolysis and resistance to hypoxia. These findings demonstrate a novel role for Notch signaling in regulating GSC metabolism and suggest intratumoral GSC heterogeneity ensures metabolic adaptations to support tumor growth in diverse tumor microenvironments.
Collapse
|
34
|
Wang W, Liu C, Jimenez-Gonzalez M, Song WJ, Hussain MA. The undoing and redoing of the diabetic β-cell. J Diabetes Complications 2017; 31:912-917. [PMID: 28242267 PMCID: PMC5450161 DOI: 10.1016/j.jdiacomp.2017.01.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/23/2016] [Accepted: 01/31/2017] [Indexed: 02/08/2023]
Abstract
A hallmark of type 2 diabetes (T2DM) is the reduction in functional β-cell mass, which is considered at least in part to result from an imbalance of β-cell renewal and apoptosis, with the latter being accelerated during metabolic stress. More recent studies, however, suggest that the loss of functional β-cell mass is not as much due to β-cell death but rather to de-differentiation of β-cells when these cells are exposed to metabolic stressors, opening the possibility to re-differentiate and restore functional β-cell mass by therapeutic intervention. In parallel, clinical observations suggest that temporary intensive insulin therapy in early diagnosed humans with T2DM, so as to "rest" endogenous β-cells, allows these patients to regain adequate insulin secretion and to maintain euglycemia for prolonged periods free of continued pharmacotherapy. Whether observations made in (mostly rodent) models of diabetes mellitus and in clinical trials are revealing identical mechanisms and therapeutic opportunities remains a tantalizing possibility. Our intention is for this review to serve as an overview of the field and commentary of this particularly exciting field of research.
Collapse
Affiliation(s)
- Wei Wang
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Chune Liu
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Maria Jimenez-Gonzalez
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Woo-Jin Song
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Mehboob A Hussain
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287.
| |
Collapse
|
35
|
Phelps EA, Cianciaruso C, Santo-Domingo J, Pasquier M, Galliverti G, Piemonti L, Berishvili E, Burri O, Wiederkehr A, Hubbell JA, Baekkeskov S. Advances in pancreatic islet monolayer culture on glass surfaces enable super-resolution microscopy and insights into beta cell ciliogenesis and proliferation. Sci Rep 2017; 7:45961. [PMID: 28401888 PMCID: PMC5388888 DOI: 10.1038/srep45961] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 03/08/2017] [Indexed: 12/16/2022] Open
Abstract
A robust and reproducible method for culturing monolayers of adherent and well-spread primary islet cells on glass coverslips is required for detailed imaging studies by super-resolution and live-cell microscopy. Guided by an observation that dispersed islet cells spread and adhere well on glass surfaces in neuronal co-culture and form a monolayer of connected cells, we demonstrate that in the absence of neurons, well-defined surface coatings combined with components of neuronal culture media collectively support robust attachment and growth of primary human or rat islet cells as monolayers on glass surfaces. The islet cell monolayer cultures on glass stably maintain distinct mono-hormonal insulin+, glucagon+, somatostatin+ and PP+ cells and glucose-responsive synchronized calcium signaling as well as expression of the transcription factors Pdx-1 and NKX-6.1 in beta cells. This technical advance enabled detailed observation of sub-cellular processes in primary human and rat beta cells by super-resolution microscopy. The protocol is envisaged to have broad applicability to sophisticated analyses of pancreatic islet cells that reveal new biological insights, as demonstrated by the identification of an in vitro protocol that markedly increases proliferation of primary beta cells and is associated with a reduction in ciliated, ostensibly proliferation-suppressed beta cells.
Collapse
Affiliation(s)
- Edward A Phelps
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Chiara Cianciaruso
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.,Graduate Program in Biotechnology and Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jaime Santo-Domingo
- Nestlé Institute of Health Sciences S.A., EPFL Innovation Park, CH-1015 Lausanne, Switzerland
| | - Miriella Pasquier
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Gabriele Galliverti
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.,Graduate Program in Biotechnology and Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Lorenzo Piemonti
- Pancreatic Islet Processing Facility, Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Faculty of Medicine, Department of Surgery, Geneva University Hospitals and University of Geneva, CH-1211 Geneva, Switzerland
| | - Olivier Burri
- BioImaging and Optics Core Facility, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Andreas Wiederkehr
- Nestlé Institute of Health Sciences S.A., EPFL Innovation Park, CH-1015 Lausanne, Switzerland
| | - Jeffrey A Hubbell
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.,Graduate Program in Biotechnology and Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.,Institute for Molecular Engineering, University of Chicago, Chicago, IL 60615, USA
| | - Steinunn Baekkeskov
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.,Graduate Program in Biotechnology and Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
36
|
Joglekar MV, Trivedi PM, Kay TW, Hawthorne WJ, O'Connell PJ, Jenkins AJ, Hardikar AA, Thomas HE. Human islet cells are killed by BID-independent mechanisms in response to FAS ligand. Apoptosis 2016; 21:379-89. [PMID: 26758067 DOI: 10.1007/s10495-016-1212-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cell death via FAS/CD95 can occur either by activation of caspases alone (extrinsic) or by activation of mitochondrial death signalling (intrinsic) depending on the cell type. The BH3-only protein BID is activated in the BCL-2-regulated or mitochondrial apoptosis pathway and acts as a switch between the extrinsic and intrinsic cell death pathways. We have previously demonstrated that islets from BID-deficient mice are protected from FAS ligand-mediated apoptosis in vitro. However, it is not yet known if BID plays a similar role in human beta cell death. We therefore aimed to test the role of BID in human islet cell apoptosis immediately after isolation from human cadaver donors, as well as after de-differentiation in vitro. Freshly isolated human islets or 10-12 day cultured human islet cells exhibited BID transcript knockdown after BID siRNA transfection, however they were not protected from FAS ligand-mediated cell death in vitro as determined by DNA fragmentation analysis using flow cytometry. On the other hand, the same cells transfected with siRNA for FAS-associated via death domain (FADD), a molecule in the extrinsic cell death pathway upstream of BID, showed significant reduction in cell death. De-differentiated islets (human islet-derived progenitor cells) also demonstrated similar results with no difference in cell death after BID knockdown as compared to scramble siRNA transfections. Our results indicate that BID-independent pathways are responsible for FAS-dependent human islet cell death. These results are different from those observed in mouse islets and therefore demonstrate potentially alternate pathways of FAS ligand-induced cell death in human and mouse islet cells.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Prerak M Trivedi
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Thomas W Kay
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Wayne J Hawthorne
- The Centre for Transplant and Renal Research, Westmead Millennium Research Institute, University of Sydney, Westmead, Australia
| | - Philip J O'Connell
- The Centre for Transplant and Renal Research, Westmead Millennium Research Institute, University of Sydney, Westmead, Australia
| | - Alicia J Jenkins
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia. .,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
37
|
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: 31] [Impact Index Per Article: 3.4] [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
|
38
|
Efrat S. Mechanisms of adult human β-cell in vitro dedifferentiation and redifferentiation. Diabetes Obes Metab 2016; 18 Suppl 1:97-101. [PMID: 27615137 DOI: 10.1111/dom.12724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/20/2016] [Indexed: 12/13/2022]
Abstract
Recent studies in animal models and human pathological specimens suggest the involvement of β-cell dedifferentiation in β-cell dysfunction associated with type 2 diabetes. Dedifferentiated β-cells may be exploited for endogenous renewal of the β-cell mass. However, studying human β-cell dedifferentiation in diabetes presents major difficulties. We have analysed mechanisms involved in human β-cell dedifferentiation in vitro, under conditions that allow cell proliferation. Although there are important differences between the two cellular environments, β-cell dedifferentiation in the two conditions is likely to share a number of common pathways. Insights from the in vitro studies may lead to development of approaches for redifferentiation of endogenous dedifferentiated β-cells.
Collapse
Affiliation(s)
- S Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel.
| |
Collapse
|
39
|
Corritore E, Lee YS, Sokal EM, Lysy PA. β-cell replacement sources for type 1 diabetes: a focus on pancreatic ductal cells. Ther Adv Endocrinol Metab 2016; 7:182-99. [PMID: 27540464 PMCID: PMC4973405 DOI: 10.1177/2042018816652059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thorough research on the capacity of human islet transplantation to cure type 1 diabetes led to the achievement of 3- to 5-year-long insulin independence in nearly half of transplanted patients. Yet, translation of this technique to clinical routine is limited by organ shortage and the need for long-term immunosuppression, restricting its use to adults with unstable disease. The production of new bona fide β cells in vitro was thus investigated and finally achieved with human pluripotent stem cells (PSCs). Besides ethical concerns about the use of human embryos, studies are now evaluating the possibility of circumventing the spontaneous tumor formation associated with transplantation of PSCs. These issues fueled the search for cell candidates for β-cell engineering with safe profiles for clinical translation. In vivo studies revealed the regeneration capacity of the exocrine pancreas after injury that depends at least partially on facultative progenitors in the ductal compartment. These stimulated subpopulations of pancreatic ductal cells (PDCs) underwent β-cell transdifferentiation through reactivation of embryonic signaling pathways. In vitro models for expansion and differentiation of purified PDCs toward insulin-producing cells were described using cocktails of growth factors, extracellular-matrix proteins and transcription factor overexpression. In this review, we will describe the latest findings in pancreatic β-cell mass regeneration due to adult ductal progenitor cells. We will further describe recent advances in human PDC transdifferentiation to insulin-producing cells with potential for clinical translational studies.
Collapse
Affiliation(s)
- Elisa Corritore
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Yong-Syu Lee
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne M. Sokal
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
40
|
Dirice E, Walpita D, Vetere A, Meier BC, Kahraman S, Hu J, Dančík V, Burns SM, Gilbert TJ, Olson DE, Clemons PA, Kulkarni RN, Wagner BK. Inhibition of DYRK1A Stimulates Human β-Cell Proliferation. Diabetes 2016; 65:1660-71. [PMID: 26953159 PMCID: PMC4878416 DOI: 10.2337/db15-1127] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 02/22/2016] [Indexed: 12/15/2022]
Abstract
Restoring functional β-cell mass is an important therapeutic goal for both type 1 and type 2 diabetes (1). While proliferation of existing β-cells is the primary means of β-cell replacement in rodents (2), it is unclear whether a similar principle applies to humans, as human β-cells are remarkably resistant to stimulation of division (3,4). Here, we show that 5-iodotubercidin (5-IT), an annotated adenosine kinase inhibitor previously reported to increase proliferation in rodent and porcine islets (5), strongly and selectively increases human β-cell proliferation in vitro and in vivo. Remarkably, 5-IT also increased glucose-dependent insulin secretion after prolonged treatment. Kinome profiling revealed 5-IT to be a potent and selective inhibitor of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) and cell division cycle-like kinase families. Induction of β-cell proliferation by either 5-IT or harmine, another natural product DYRK1A inhibitor, was suppressed by coincubation with the calcineurin inhibitor FK506, suggesting involvement of DYRK1A and nuclear factor of activated T cells signaling. Gene expression profiling in whole islets treated with 5-IT revealed induction of proliferation- and cell cycle-related genes, suggesting that true proliferation is induced by 5-IT. Furthermore, 5-IT promotes β-cell proliferation in human islets grafted under the kidney capsule of NOD-scid IL2Rg(null) mice. These results point to inhibition of DYRK1A as a therapeutic strategy to increase human β-cell proliferation.
Collapse
Affiliation(s)
- Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Deepika Walpita
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Amedeo Vetere
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Bennett C Meier
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
| | - Sevim Kahraman
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Vlado Dančík
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Sean M Burns
- Chemical Biology Program, Harvard Medical School, Boston, MA Diabetes Unit, Departments of Medicine and Molecular Biology, Massachusetts General Hospital, Boston, MA
| | - Tamara J Gilbert
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - David E Olson
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | - Paul A Clemons
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Bridget K Wagner
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA
| |
Collapse
|
41
|
Abstract
Since insulin discovery, islet transplantation was the first protocol to show the possibility to cure patients with type 1 diabetes using low-risk procedures. The scarcity of pancreas donors triggered a burst of studies focused on the production of new β cells in vitro. These were rapidly dominated by pluripotent stem cells (PSCs) demonstrating diabetes-reversal potential in diabetic mice. Subsequent enthusiasm fostered a clinical trial with immunoisolated embryonic-derived pancreatic progenitors. Yet safety is the Achilles' heel of PSCs, and a whole branch of β cell engineering medicine focuses on transdifferentiation of adult pancreatic cells. New data showed the possibility to chemically stimulate acinar or α cells to undergo β cell neogenesis and provide opportunities to intervene in situ without the need for a transplant, at least after weighing benefits against systemic adverse effects. The current studies suggested the pancreas as a reservoir of facultative progenitors (e.g., in the duct lining) could be exploited ex vivo for expansion and β cell differentiation in timely fashion and without the hurdles of PSC use. Diabetes cell therapy is thus a growing field not only with great potential but also with many pitfalls to overcome for becoming fully envisioned as a competitor to the current treatment standards.
Collapse
Affiliation(s)
- Philippe A Lysy
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium.
- Pediatric Endocrinology Unit, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium.
| | - Elisa Corritore
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne M Sokal
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
42
|
Beamish CA, Strutt BJ, Arany EJ, Hill DJ. Insulin-positive, Glut2-low cells present within mouse pancreas exhibit lineage plasticity and are enriched within extra-islet endocrine cell clusters. Islets 2016; 8:65-82. [PMID: 27010375 PMCID: PMC4987018 DOI: 10.1080/19382014.2016.1162367] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 01/01/2023] Open
Abstract
Regeneration of insulin-producing β-cells from resident pancreas progenitors requires an understanding of both progenitor identity and lineage plasticity. One model suggested that a rare β-cell sub-population within islets demonstrated multi-lineage plasticity. We hypothesized that β-cells from young mice (postnatal day 7, P7) exhibit such plasticity and used a model of islet dedifferentiation toward a ductal epithelial-cell phenotype to test this theory. RIPCre;Z/AP(+/+) mice were used to lineage trace the fate of β-cells during dedifferentiation culture by a human placental alkaline phosphatase (HPAP) reporter. There was a significant loss of HPAP-expressing β-cells in culture, but remaining HPAP(+) cells lost insulin expression while gaining expression of the epithelial duct cell marker cytokeratin-19 (Ck19). Flow cytometry and recovery of β-cell subpopulations from whole pancreas vs. islets suggest that the HPAP(+)Ck19(+) cells had derived from insulin-positive, glucose-transporter-2-low (Ins(+)Glut2(LO)) cells, representing 3.5% of all insulin-expressing cells. The majority of these cells were found outside of islets within clusters of <5 β-cells. These insulin(+)Glut2(LO) cells demonstrated a greater proliferation rate in vivo and in vitro as compared to insulin(+)Glut2(+) cells at P7, were retained into adulthood, and a subset differentiated into endocrine, ductal, and neural lineages, illustrating substantial plasticity. Results were confirmed using RIPCre;ROSA- eYFP mice. Quantitative PCR data indicated these cells possess an immature β-cell phenotype. These Ins(+)Glut2(LO) cells may represent a resident population of cells capable of forming new, functional β-cells, and which may be potentially exploited for regenerative therapies in the future.
Collapse
Affiliation(s)
- Christine A. Beamish
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - Brenda J. Strutt
- Department of Medicine, Western University, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - Edith J. Arany
- Department of Medicine, Western University, London, ON, Canada
- Department of Pathology, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - David J. Hill
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| |
Collapse
|
43
|
Kim HS, Lee MK. β-Cell regeneration through the transdifferentiation of pancreatic cells: Pancreatic progenitor cells in the pancreas. J Diabetes Investig 2016; 7:286-96. [PMID: 27330712 PMCID: PMC4847880 DOI: 10.1111/jdi.12475] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/27/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022] Open
Abstract
Pancreatic progenitor cell research has been in the spotlight, as these cells have the potential to replace pancreatic β‐cells for the treatment of type 1 and 2 diabetic patients with the absence or reduction of pancreatic β‐cells. During the past few decades, the successful treatment of diabetes through transplantation of the whole pancreas or isolated islets has nearly been achieved. However, novel sources of pancreatic islets or insulin‐producing cells are required to provide sufficient amounts of donor tissues. To overcome this limitation, the use of pancreatic progenitor cells is gaining more attention. In particular, pancreatic exocrine cells, such as duct epithelial cells and acinar cells, are attractive candidates for β‐cell regeneration because of their differentiation potential and pancreatic lineage characteristics. It has been assumed that β‐cell neogenesis from pancreatic progenitor cells could occur in pancreatic ducts in the postnatal stage. Several studies have shown that insulin‐producing cells can arise in the duct tissue of the adult pancreas. Acinar cells also might have the potential to differentiate into insulin‐producing cells. The present review summarizes recent progress in research on the transdifferentiation of pancreatic exocrine cells into insulin‐producing cells, especially duct and acinar cells.
Collapse
Affiliation(s)
- Hyo-Sup Kim
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| | - Moon-Kyu Lee
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| |
Collapse
|
44
|
Redifferentiation of expanded human islet β cells by inhibition of ARX. Sci Rep 2016; 6:20698. [PMID: 26856418 PMCID: PMC4746595 DOI: 10.1038/srep20698] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/11/2016] [Indexed: 12/14/2022] Open
Abstract
Ex-vivo expansion of adult human islet β cells has been evaluated for generation of abundant insulin-producing cells for transplantation; however, lineage-tracing has demonstrated that this process results in β-cell dedifferentiation. Redifferentiation of β-cell-derived (BCD) cells can be achieved using a combination of soluble factors termed Redifferentiation Cocktail (RC); however, this treatment leads to redifferentiation of only a fraction of BCD cells. This study aimed at improving redifferentiation efficiency by affecting the balance of islet progenitor-cell transcription factors activated by RC treatment. Specifically, RC treatment induces the transcription factors PAX4 and ARX, which play key roles in directing pancreas endocrine progenitor cells into the β/δ or α/PP developmental pathways, respectively. Misactivation of ARX in RC-treated BCD cells may inhibit their redifferentiation into β cells. Blocking ARX expression by shRNA elevated insulin mRNA levels 12.8-fold, and more than doubled the number of insulin-positive BCD cells. ARX inhibition in expanded α-cell-derived cells treated with RC did not cause their transdifferentiation into insulin-producing cells. The combination of RC and ARX shRNA treatment may facilitate the generation of abundant insulin-producing cells for transplantation into patients with type 1 diabetes.
Collapse
|
45
|
Niu G, McQuilling JP, Zhou Y, Opara EC, Orlando G, Soker S. In Vitro Proliferation of Porcine Pancreatic Islet Cells for β-Cell Therapy Applications. J Diabetes Res 2016; 2016:5807876. [PMID: 28050568 PMCID: PMC5168554 DOI: 10.1155/2016/5807876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/04/2016] [Accepted: 11/13/2016] [Indexed: 11/29/2022] Open
Abstract
β-Cell replacement through transplantation is the only curative treatment to establish a long-term stable euglycemia in diabetic patients. Owing to the shortage of donor tissue, attempts are being made to develop alternative sources of insulin-secreting cells. Stem cells differentiation and reprograming as well as isolating pancreatic progenitors from different sources are some examples; however, no approach has yet yielded a clinically relevant solution. Dissociated islet cells that are cultured in cell numbers by in vitro proliferation provide a promising platform for redifferentiation towards β-cells phenotype. In this study, we cultured islet-derived cells in vitro and examined the expression of β-cell genes during the proliferation. Islets were isolated from porcine pancreases and enzymatically digested to dissociate the component cells. The cells proliferated well in tissue culture plates and were subcultured for no more than 5 passages. Only 10% of insulin expression, as measured by PCR, was preserved in each passage. High glucose media enhanced insulin expression by about 4-18 fold, suggesting a glucose-dependent effect in the proliferated islet-derived cells. The islet-derived cells also expressed other pancreatic genes such as Pdx1, NeuroD, glucagon, and somatostatin. Taken together, these results indicate that pancreatic islet-derived cells, proliferated in vitro, retained the expression capacity for key pancreatic genes, thus suggesting that the cells may be redifferentiated into insulin-secreting β-like cells.
Collapse
Affiliation(s)
- Guoguang Niu
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - John P. McQuilling
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Yu Zhou
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Emmanuel C. Opara
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Giuseppe Orlando
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Virginia Tech, Wake Forest University School of Biomedical Engineering and Sciences, 320 ICTAS, Stanger St., Virginia Tech, Blacksburg, VA 24060, USA
- *Shay Soker:
| |
Collapse
|
46
|
ActivinB Is Induced in Insulinoma To Promote Tumor Plasticity through a β-Cell-Induced Dedifferentiation. Mol Cell Biol 2015; 36:756-64. [PMID: 26711255 DOI: 10.1128/mcb.00930-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/13/2015] [Indexed: 12/19/2022] Open
Abstract
Loss of pancreatic β-cell maturity occurs in diabetes and insulinomas. Although both physiological and pathological stresses are known to promote β-cell dedifferentiation, little is known about the molecules involved in this process. Here we demonstrate that activinB, a transforming growth factor β (TGF-β)-related ligand, is upregulated during tumorigenesis and drives the loss of insulin expression and β-cell maturity in a mouse insulinoma model. Our data further identify Pax4 as a previously unknown activinB target and potent contributor to the observed β-cell dedifferentiation. More importantly, using compound mutant mice, we found that deleting activinB expression abolishes tumor β-cell dedifferentiation and, surprisingly, increases survival without significantly affecting tumor growth. Hence, this work reveals an unexpected role for activinB in the loss of β-cell maturity, islet plasticity, and progression of insulinoma through its participation in β-cell dedifferentiation.
Collapse
|
47
|
Jiang FX, Morahan G. Multipotent pancreas progenitors: Inconclusive but pivotal topic. World J Stem Cells 2015; 7:1251-1261. [PMID: 26730269 PMCID: PMC4691693 DOI: 10.4252/wjsc.v7.i11.1251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/20/2015] [Accepted: 11/11/2015] [Indexed: 02/07/2023] Open
Abstract
The establishment of multipotent pancreas progenitors (MPP) should have a significant impact not only on the ontology of the pancreas, but also for the translational research of glucose-responding endocrine β-cells. Deficiency of the latter may lead to the pandemic type 1 or type 2 diabetes mellitus, a metabolic disorder. An ideal treatment of which would potentially be the replacement of destroyed or failed β-cells, by restoring function of endogenous pancreatic endocrine cells or by transplantation of donor islets or in vitro generated insulin-secreting cells. Thus, considerable research efforts have been devoted to identify MPP candidates in the pre- and post-natal pancreas for the endogenous neogenesis or regeneration of endocrine insulin-secreting cells. In order to advance this inconclusive but critical field, we here review the emerging concepts, recent literature and newest developments of potential MPP and propose measures that would assist its forward progression.
Collapse
|
48
|
Epigenomic Reprogramming of Adult Cardiomyocyte-Derived Cardiac Progenitor Cells. Sci Rep 2015; 5:17686. [PMID: 26657817 PMCID: PMC4677315 DOI: 10.1038/srep17686] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 10/14/2015] [Indexed: 01/01/2023] Open
Abstract
It has been believed that mammalian adult cardiomyocytes (ACMs) are terminally-differentiated and are unable to proliferate. Recently, using a bi-transgenic ACM fate mapping mouse model and an in vitro culture system, we demonstrated that adult mouse cardiomyocytes were able to dedifferentiate into cardiac progenitor-like cells (CPCs). However, little is known about the molecular basis of their intrinsic cellular plasticity. Here we integrate single-cell transcriptome and whole-genome DNA methylation analyses to unravel the molecular mechanisms underlying the dedifferentiation and cell cycle reentry of mouse ACMs. Compared to parental cardiomyocytes, dedifferentiated mouse cardiomyocyte-derived CPCs (mCPCs) display epigenomic reprogramming with many differentially-methylated regions, both hypermethylated and hypomethylated, across the entire genome. Correlated well with the methylome, our transcriptomic data showed that the genes encoding cardiac structure and function proteins are remarkably down-regulated in mCPCs, while those for cell cycle, proliferation, and stemness are significantly up-regulated. In addition, implantation of mCPCs into infarcted mouse myocardium improves cardiac function with augmented left ventricular ejection fraction. Our study demonstrates that the cellular plasticity of mammalian cardiomyocytes is the result of a well-orchestrated epigenomic reprogramming and a subsequent global transcriptomic alteration.
Collapse
|
49
|
Klein D, Álvarez-Cubela S, Lanzoni G, Vargas N, Prabakar KR, Boulina M, Ricordi C, Inverardi L, Pastori RL, Domínguez-Bendala J. BMP-7 Induces Adult Human Pancreatic Exocrine-to-Endocrine Conversion. Diabetes 2015; 64:4123-34. [PMID: 26307584 PMCID: PMC4657585 DOI: 10.2337/db15-0688] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/17/2015] [Indexed: 12/30/2022]
Abstract
The exocrine pancreas can give rise to endocrine insulin-producing cells upon ectopic expression of key transcription factors. However, the need for genetic manipulation remains a translational hurdle for diabetes therapy. Here we report the conversion of adult human nonendocrine pancreatic tissue into endocrine cell types by exposure to bone morphogenetic protein 7. The use of this U.S. Food and Drug Administration-approved agent, without any genetic manipulation, results in the neogenesis of clusters that exhibit high insulin content and glucose responsiveness both in vitro and in vivo. In vitro lineage tracing confirmed that BMP-7-induced insulin-expressing cells arise mainly from extrainsular PDX-1(+), carbonic anhydrase II(-) (mature ductal), elastase 3a (acinar)(-) , and insulin(-) subpopulations. The nongenetic conversion of human pancreatic exocrine cells to endocrine cells is novel and represents a safer and simpler alternative to genetic reprogramming.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- Bone Morphogenetic Protein 7/genetics
- Bone Morphogenetic Protein 7/metabolism
- Bone Morphogenetic Protein 7/pharmacology
- C-Peptide/blood
- C-Peptide/metabolism
- Cell Lineage
- Cell Transdifferentiation/drug effects
- Cells, Cultured
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/therapy
- Fluorescent Antibody Technique
- Homeodomain Proteins/metabolism
- Humans
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Insulin-Secreting Cells/transplantation
- Kidney
- Male
- Mice, Nude
- Pancreas, Exocrine/drug effects
- Pancreas, Exocrine/metabolism
- Pancreas, Exocrine/pathology
- Recombinant Proteins/metabolism
- Recombinant Proteins/pharmacology
- Trans-Activators/metabolism
- Transplantation, Heterologous
- Transplantation, Heterotopic
Collapse
Affiliation(s)
- Dagmar Klein
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Giacomo Lanzoni
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Nancy Vargas
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Kamalaveni R Prabakar
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Maria Boulina
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Camillo Ricordi
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL Department of Biomedical Engineering, Miller School of Medicine, University of Miami, Miami, FL
| | - Luca Inverardi
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL
| | - Ricardo L Pastori
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL Department of Cell Biology and Anatomy, Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
50
|
Benazra M, Lecomte MJ, Colace C, Müller A, Machado C, Pechberty S, Bricout-Neveu E, Grenier-Godard M, Solimena M, Scharfmann R, Czernichow P, Ravassard P. A human beta cell line with drug inducible excision of immortalizing transgenes. Mol Metab 2015; 4:916-25. [PMID: 26909308 PMCID: PMC4731729 DOI: 10.1016/j.molmet.2015.09.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/17/2015] [Accepted: 09/22/2015] [Indexed: 12/30/2022] Open
Abstract
Objectives Access to immortalized human pancreatic beta cell lines that are phenotypically close to genuine adult beta cells, represent a major tool to better understand human beta cell physiology and develop new therapeutics for Diabetes. Here we derived a new conditionally immortalized human beta cell line, EndoC-βH3 in which immortalizing transgene can be efficiently removed by simple addition of tamoxifen. Methods We used lentiviral mediated gene transfer to stably integrate a tamoxifen inducible form of CRE (CRE-ERT2) into the recently developed conditionally immortalized EndoC βH2 line. The resulting EndoC-βH3 line was characterized before and after tamoxifen treatment for cell proliferation, insulin content and insulin secretion. Results We showed that EndoC-βH3 expressing CRE-ERT2 can be massively amplified in culture. We established an optimized tamoxifen treatment to efficiently excise the immortalizing transgenes resulting in proliferation arrest. In addition, insulin expression raised by 12 fold and insulin content increased by 23 fold reaching 2 μg of insulin per million cells. Such massive increase was accompanied by enhanced insulin secretion upon glucose stimulation. We further observed that tamoxifen treated cells maintained a stable function for 5 weeks in culture. Conclusions EndoC βH3 cell line represents a powerful tool that allows, using a simple and efficient procedure, the massive production of functional non-proliferative human beta cells. Such cells are close to genuine human beta cells and maintain a stable phenotype for 5 weeks in culture. EndoC-βH3: a conditionally immortalized human pancreatic beta cell line. Proliferation arrest upon removal of immortalizing transgenes with Tamoxifen. Enhancement of beta cell function upon removal of immortalizing transgenes. Tamoxifen-treated EndoC-βH3 maintain a stable phenotype for 5 weeks in culture. EndoC-βH3: a unique tool for large-scale drug discovery and proliferation studies.
Collapse
Affiliation(s)
- Marion Benazra
- Institut du cerveau et de la moelle (ICM), Biotechnology & Biotherapy Team, 75013 Paris, France
- CNRS UMR7225, 75013 Paris, France
- INSERM U1127, 75013 Paris, France
- Université Pierre et Marie Curie, 75013 Paris, France
| | - Marie-José Lecomte
- Endocells, Pépinière d'entreprises Institut du Cerveau et de la Moelle, 75007 Paris, France
| | - Claire Colace
- Institut du cerveau et de la moelle (ICM), Biotechnology & Biotherapy Team, 75013 Paris, France
- CNRS UMR7225, 75013 Paris, France
- INSERM U1127, 75013 Paris, France
- Université Pierre et Marie Curie, 75013 Paris, France
| | - Andreas Müller
- Paul Langerhans Institute of the Helmholtz Center Munich at the University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
- German Center for Diabetes Research (DZD e.V), 85764 Neuherberg, Germany
| | - Cécile Machado
- Endocells, Pépinière d'entreprises Institut du Cerveau et de la Moelle, 75007 Paris, France
| | - Severine Pechberty
- Endocells, Pépinière d'entreprises Institut du Cerveau et de la Moelle, 75007 Paris, France
| | - Emilie Bricout-Neveu
- Endocells, Pépinière d'entreprises Institut du Cerveau et de la Moelle, 75007 Paris, France
| | - Maud Grenier-Godard
- Endocells, Pépinière d'entreprises Institut du Cerveau et de la Moelle, 75007 Paris, France
| | - Michele Solimena
- Paul Langerhans Institute of the Helmholtz Center Munich at the University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
- German Center for Diabetes Research (DZD e.V), 85764 Neuherberg, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Raphaël Scharfmann
- INSERM, U1016, Institut Cochin, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Paul Czernichow
- Endocells, Pépinière d'entreprises Institut du Cerveau et de la Moelle, 75007 Paris, France
| | - Philippe Ravassard
- Institut du cerveau et de la moelle (ICM), Biotechnology & Biotherapy Team, 75013 Paris, France
- CNRS UMR7225, 75013 Paris, France
- INSERM U1127, 75013 Paris, France
- Université Pierre et Marie Curie, 75013 Paris, France
- Corresponding author. ICM Biotechnology & Biotherapy Team, Hôpital Pitié Salpêtrière, 47 Bd. De l'Hôpital, 75013 Paris, France. Tel./fax: +33 157274575.
| |
Collapse
|