1
|
Haq N, Toczyska KW, Wilson ME, Jacobs M, Zhao M, Lei Y, Shen Z, Pearson JA, Persaud SJ, Pullen TJ, Bewick GA. Reformed islets: a long-term primary cell platform for exploring mouse and human islet biology. Cell Death Discov 2024; 10:480. [PMID: 39580467 PMCID: PMC11585622 DOI: 10.1038/s41420-024-02234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 10/19/2024] [Accepted: 10/30/2024] [Indexed: 11/25/2024] Open
Abstract
Pancreatic islets are 3D micro-organs that maintain β-cell functionality through cell-cell and cell-matrix communication. While primary islets, the gold standard for in vitro models, have a short culture life of approximately 1-2 weeks, we developed a novel protocol that employs reformed islets following dispersion coupled with a fine-tuned culture environment. Reformed islets exhibit physiological characteristics similar to primary islets, enabling high-resolution imaging and repeated functional assessment. Unlike other in vitro platforms, reformed islets retain an immune population, allowing the study of interactions between β cells and resident and infiltrating immune cells. Analyses showed that reformed islets have a similar composition and cytoarchitecture to primary islets, including macrophages and T cells, and can secrete insulin in response to glucose. Reformed islets exhibited partial dedifferentiation compared to native islets but were otherwise transcriptionally similar. The reformed islets offer a useful platform for studying diabetes pathology and can recapitulate both T1DM and T2DM disease milieus, providing an advantage over other models, such as mouse and human β-cell lines, which lack the input of non-β-endocrine cells and immune cell crosstalk.
Collapse
Affiliation(s)
- N Haq
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - K W Toczyska
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - M E Wilson
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - M Jacobs
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - Min Zhao
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - Y Lei
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - Z Shen
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - J A Pearson
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - S J Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - T J Pullen
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK
| | - G A Bewick
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, Diabetes Endocrinology and Obesity Clinical Academic Partnership, King's College London and King's Health Partners, Guy's Campus, London, UK.
| |
Collapse
|
2
|
Rohban R, Martins CP, Esni F. Advanced therapy to cure diabetes: mission impossible is now possible? Front Cell Dev Biol 2024; 12:1484859. [PMID: 39629270 PMCID: PMC11611888 DOI: 10.3389/fcell.2024.1484859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Cell and Gene therapy are referred to as advanced therapies that represent overlapping fields of regenerative medicine. They have similar therapeutic goals such as to modify cellular identity, improve cell function, or fight a disease. These two therapeutic avenues, however, possess major differences. While cell therapy involves introduction of new cells, gene therapy entails introduction or modification of genes. Furthermore, the aim of cell therapy is often to replace, or repair damaged tissue, whereas gene therapy is used typically as a preventive approach. Diabetes mellitus severely affects the quality of life of afflicted individuals and has various side effects including cardiovascular, ophthalmic disorders, and neuropathy while putting enormous economic pressure on both the healthcare system and the patient. In recent years, great effort has been made to develop cutting-edge therapeutic interventions for diabetes treatment, among which cell and gene therapies stand out. This review aims to highlight various cell- and gene-based therapeutic approaches leading to the generation of new insulin-producing cells as a topmost "panacea" for treating diabetes, while deliberately avoiding a detailed molecular description of these approaches. By doing so, we aim to target readers who are new to the field and wish to get a broad helicopter overview of the historical and current trends of cell- and gene-based approaches in β-cell regeneration.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Christina P. Martins
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- McGowan Institute for regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
3
|
Tanday N, Tarasov AI, Moffett RC, Flatt PR, Irwin N. Pancreatic islet cell plasticity: Pathogenic or therapeutically exploitable? Diabetes Obes Metab 2024; 26:16-31. [PMID: 37845573 DOI: 10.1111/dom.15300] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 10/18/2023]
Abstract
The development of pancreatic islet endocrine cells is a tightly regulated process leading to the generation of distinct cell types harbouring different hormones in response to small changes in environmental stimuli. Cell differentiation is driven by transcription factors that are also critical for the maintenance of the mature islet cell phenotype. Alteration of the insulin-secreting β-cell transcription factor set by prolonged metabolic stress, associated with the pathogenesis of diabetes, obesity or pregnancy, results in the loss of β-cell identity through de- or transdifferentiation. Importantly, the glucose-lowering effects of approved and experimental antidiabetic agents, including glucagon-like peptide-1 mimetics, novel peptides and small molecules, have been associated with preventing or reversing β-cell dedifferentiation or promoting the transdifferentiation of non-β-cells towards an insulin-positive β-cell-like phenotype. Therefore, we review the manifestations of islet cell plasticity in various experimental settings and discuss the physiological and therapeutic sides of this phenomenon, focusing on strategies for preventing β-cell loss or generating new β-cells in diabetes. A better understanding of the molecular mechanisms underpinning islet cell plasticity is a prerequisite for more targeted therapies to help prevent β-cell decline in diabetes.
Collapse
Affiliation(s)
- Neil Tanday
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Andrei I Tarasov
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| | - R Charlotte Moffett
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| | - Nigel Irwin
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| |
Collapse
|
4
|
Morisseau L, Tokito F, Poulain S, Plaisance V, Pawlowski V, Kim SH, Legallais C, Jellali R, Sakai Y, Abderrahmani A, Leclerc E. Generation of β-like cell subtypes from differentiated human induced pluripotent stem cells in 3D spheroids. Mol Omics 2023; 19:810-822. [PMID: 37698079 DOI: 10.1039/d3mo00050h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Since the identification of four different pancreatic β-cell subtypes and bi-hormomal cells playing a role in the diabetes pathogenesis, the search for in vitro models that mimics such cells heterogeneity became a key priority in experimental and clinical diabetology. We investigated the potential of human induced pluripotent stem cells to lead to the development of the different β-cells subtypes in honeycomb microwell-based 3D spheroids. The glucose-stimulated insulin secretion confirmed the spheroids functionality. Then, we performed a single cell RNA sequencing of the spheroids. Using a knowledge-based analysis with a stringency on the pancreatic markers, we extracted the β-cells INS+/UCN3+ subtype (11%; β1-like cells), the INS+/ST8SIA1+/CD9- subtype (3%, β3-like cells) and INS+/CD9+/ST8SIA1-subtype (1%; β2-like cells) consistently with literature findings. We did not detect the INS+/ST8SIA1+/CD9+ cells (β4-like cells). Then, we also identified four bi-hormonal cells subpopulations including δ-like cells (INS+/SST+, 6%), γ-like cells (INS+/PPY+, 3%), α-like-cells (INS+/GCG+, 6%) and ε-like-cells (INS+/GHRL+, 2%). Using data-driven clustering, we extracted four progenitors' subpopulations (with the lower level of INS gene) that included one population highly expressing inhibin genes (INHBA+/INHBB+), one population highly expressing KCNJ3+/TPH1+, one population expressing hepatocyte-like lineage markers (HNF1A+/AFP+), and one population expressing stem-like cell pancreatic progenitor markers (SOX2+/NEUROG3+). Furthermore, among the cycling population we found a large number of REST+ cells and CD9+ cells (CD9+/SPARC+/REST+). Our data confirm that our differentiation leads to large β-cell heterogeneity, which can be used for investigating β-cells plasticity under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Lisa Morisseau
- Biomechanics and Bioengineering UMR 7338, Université de technologie de Compiègne, CNRS, Centre de Recherche Royallieu CS 60319, Compiègne, 60203 Cedex, France
| | - Fumiya Tokito
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Stéphane Poulain
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| | - Valerie Plaisance
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Valerie Pawlowski
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Soo Hyeon Kim
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| | - Cécile Legallais
- Biomechanics and Bioengineering UMR 7338, Université de technologie de Compiègne, CNRS, Centre de Recherche Royallieu CS 60319, Compiègne, 60203 Cedex, France
| | - Rachid Jellali
- Biomechanics and Bioengineering UMR 7338, Université de technologie de Compiègne, CNRS, Centre de Recherche Royallieu CS 60319, Compiègne, 60203 Cedex, France
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
- Laboratory for Integrated Micro Mechatronic Systems, CNRS/IIS IRL 2820, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| | - Amar Abderrahmani
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Eric Leclerc
- Laboratory for Integrated Micro Mechatronic Systems, CNRS/IIS IRL 2820, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| |
Collapse
|
5
|
Mathisen AF, Abadpour S, Legøy TA, Paulo JA, Ghila L, Scholz H, Chera S. Global proteomics reveals insulin abundance as a marker of human islet homeostasis alterations. Acta Physiol (Oxf) 2023; 239:e14037. [PMID: 37621186 PMCID: PMC10592125 DOI: 10.1111/apha.14037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
AIM The variation in quality between the human islet samples represents a major problem for research, especially when used as control material. The assays assessing the quality of human islets used in research are non-standardized and limited, with many important parameters not being consistently assessed. High-throughput studies aimed at characterizing the diversity and segregation markers among apparently functionally healthy islet preps are thus a requirement. Here, we designed a pilot study to comprehensively identify the diversity of global proteome signatures and the deviation from normal homeostasis in randomly selected human-isolated islet samples. METHODS By using Tandem Mass Tag 16-plex proteomics, we focused on the recurrently observed disparity in the detected insulin abundance between the samples, used it as a segregating parameter, and analyzed the correlated changes in the proteome signature and homeostasis by pathway analysis. RESULTS In this pilot study, we showed that insulin protein abundance is a predictor of human islet homeostasis and quality. This parameter is independent of other quality predictors within their acceptable range, thus being able to further stratify islets samples of apparent good quality. Human islets with low amounts of insulin displayed changes in their metabolic and signaling profile, especially in regard to energy homeostasis and cell identity maintenance. We further showed that xenotransplantation into diabetic hosts is not expected to improve the pre-transplantation signature, as it has a negative effect on energy balance, antioxidant activity, and islet cell identity. CONCLUSIONS Insulin protein abundance predicts significant changes in human islet homeostasis among random samples of apparently good quality.
Collapse
Affiliation(s)
- Andreas F. Mathisen
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Shadab Abadpour
- Hybrid Technology Hub-Centre of Excellence, Faculty of Medicine, University of Oslo, Norway
- Institute for Surgical Research and Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway
| | - Thomas Aga Legøy
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Luiza Ghila
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hanne Scholz
- Hybrid Technology Hub-Centre of Excellence, Faculty of Medicine, University of Oslo, Norway
- Institute for Surgical Research and Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway
| | - Simona Chera
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
6
|
Brennecke BR, Yang US, Liu S, Ilerisoy FS, Ilerisoy BN, Joglekar A, Kim LB, Peachee SJ, Richtsmeier SL, Stephens SB, Sander EA, Strack S, Moninger TO, Ankrum JA, Imai Y. Utilization of commercial collagens for preparing well-differentiated human beta cells for confocal microscopy. Front Endocrinol (Lausanne) 2023; 14:1187216. [PMID: 37305047 PMCID: PMC10248405 DOI: 10.3389/fendo.2023.1187216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction With technical advances, confocal and super-resolution microscopy have become powerful tools to dissect cellular pathophysiology. Cell attachment to glass surfaces compatible with advanced imaging is critical prerequisite but remains a considerable challenge for human beta cells. Recently, Phelps et al. reported that human beta cells plated on type IV collagen (Col IV) and cultured in neuronal medium preserve beta cell characteristics. Methods We examined human islet cells plated on two commercial sources of Col IV (C6745 and C5533) and type V collagen (Col V) for differences in cell morphology by confocal microscopy and secretory function by glucose-stimulated insulin secretion (GSIS). Collagens were authenticated by mass spectrometry and fluorescent collagen-binding adhesion protein CNA35. Results All three preparations allowed attachment of beta cells with high nuclear localization of NKX6.1, indicating a well-differentiated status. All collagen preparations supported robust GSIS. However, the morphology of islet cells differed between the 3 preparations. C5533 showed preferable features as an imaging platform with the greatest cell spread and limited stacking of cells followed by Col V and C6745. A significant difference in attachment behavior of C6745 was attributed to the low collagen contents of this preparation indicating importance of authentication of coating material. Human islet cells plated on C5533 showed dynamic changes in mitochondria and lipid droplets (LDs) in response to an uncoupling agent 2-[2-[4-(trifluoromethoxy)phenyl]hydrazinylidene]-propanedinitrile (FCCP) or high glucose + oleic acid. Discussion An authenticated preparation of Col IV provides a simple platform to apply advanced imaging for studies of human islet cell function and morphology.
Collapse
Affiliation(s)
- Brianna R. Brennecke
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - USeong Yang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Siming Liu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Fatma S. Ilerisoy
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Beyza N. Ilerisoy
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Aditya Joglekar
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Lucy B. Kim
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Spencer J. Peachee
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Syreine L. Richtsmeier
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Samuel B. Stephens
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Edward A. Sander
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
| | - Thomas O. Moninger
- Central Microscopy Research Facility, Roy G. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - James A. Ankrum
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Yumi Imai
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Medical Service, Endocrinology Section, Iowa City Veterans Affairs Medical Center, Iowa City, IA, United States
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Ji Z, Lu M, Xie H, Yuan H, Chen Q. β cell regeneration and novel strategies for treatment of diabetes (Review). Biomed Rep 2022; 17:72. [DOI: 10.3892/br.2022.1555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Zengyang Ji
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Min Lu
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Huanhuan Xie
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Honggang Yuan
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Qing Chen
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| |
Collapse
|
10
|
Mohan S, Lafferty RA, Flatt PR, Moffett RC, Irwin N. Ac3IV, a V1a and V1b receptor selective vasopressin analogue, protects against hydrocortisone-induced changes in pancreatic islet cell lineage. Peptides 2022; 152:170772. [PMID: 35202749 DOI: 10.1016/j.peptides.2022.170772] [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: 12/07/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 11/25/2022]
Abstract
The Avpr1a (V1a) and Avpr1b (V1b) receptor selective, vasopressin (AVP) analogue, Ac3IV has been shown to improve metabolism and pancreatic islet structure in diabetes and insulin resistance. The present study further investigates these actions by assessing the ability of Ac3IV to protect against pancreatic islet architectural disturbances induced by hydrocortisone (HC) treatment in transgenic Ins1Cre/+;Rosa26-eYFP mice, that possess beta-cell lineage tracing capabilities. HC intervention increased (p < 0.001) energy intake but reduced (p < 0.01) body weight gain, with no impact of Ac3IV. All HC mice had reduced (p < 0.05) circulating glucose, but plasma insulin and glucagon concentrations remained unchanged. However, HC mice presented with increased (p < 0.001) pancreatic insulin content, which was further augmented by Ac3IV. In addition, Ac3IV treatment countered HC-induced increases in islet-, beta- and alpha-cell areas (p < 0.01), as well as promoting islet number towards control levels. This was accompanied by reduced (p < 0.05) beta-cell growth, but enhanced (p < 0.001) alpha-cell proliferation. There were no changes in islet cell apoptotic rates in any of the groups of HC mice, but co-expression of CK19 with insulin in pancreatic ductal cells was reduced by Ac3IV. Assessment of beta-cell lineage revealed that Ac3IV partially protected against HC-mediated de-differentiation of mature beta-cells, whilst also decreasing (p < 0.01) beta- to alpha-cell transdifferentiation. Our data indicate that sustained activation of V1a and V1b receptors exerts positive islet cell transition effects to help retain beta-cell identity in HC mice.
Collapse
Affiliation(s)
- Shruti Mohan
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Ryan A Lafferty
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - R Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK.
| |
Collapse
|
11
|
Álvarez-Nava F, Soto-Quintana M. The Hypothesis of the Prolonged Cell Cycle in Turner Syndrome. J Dev Biol 2022; 10:16. [PMID: 35645292 PMCID: PMC9149809 DOI: 10.3390/jdb10020016] [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: 02/28/2022] [Accepted: 03/13/2022] [Indexed: 01/27/2023] Open
Abstract
Turner syndrome (TS) is a chromosomal disorder that is caused by a missing or structurally abnormal second sex chromosome. Subjects with TS are at an increased risk of developing intrauterine growth retardation, low birth weight, short stature, congenital heart diseases, infertility, obesity, dyslipidemia, hypertension, insulin resistance, type 2 diabetes mellitus, metabolic syndrome, and cardiovascular diseases (stroke and myocardial infarction). The underlying pathogenetic mechanism of TS is unknown. The assumption that X chromosome-linked gene haploinsufficiency is associated with the TS phenotype is questioned since such genes have not been identified. Thus, other pathogenic mechanisms have been suggested to explain this phenotype. Morphogenesis encompasses a series of events that includes cell division, the production of migratory precursors and their progeny, differentiation, programmed cell death, and integration into organs and systems. The precise control of the growth and differentiation of cells is essential for normal development. The cell cycle frequency and the number of proliferating cells are essential in cell growth. 45,X cells have a failure to proliferate at a normal rate, leading to a decreased cell number in a given tissue during organogenesis. A convergence of data indicates an association between a prolonged cell cycle and the phenotypical features in Turner syndrome. This review aims to examine old and new findings concerning the relationship between a prolonged cell cycle and TS phenotype. These studies reveal a diversity of phenotypic features in TS that could be explained by reduced cell proliferation. The implications of this hypothesis for our understanding of the TS phenotype and its pathogenesis are discussed. It is not surprising that 45,X monosomy leads to cellular growth pathway dysregulation with profound deleterious effects on both embryonic and later stages of development. The prolonged cell cycle could represent the beginning of the pathogenesis of TS, leading to a series of phenotypic consequences in embryonic/fetal, neonatal, pediatric, adolescence, and adulthood life.
Collapse
Affiliation(s)
- Francisco Álvarez-Nava
- Biological Sciences School, Faculty of Biological Sciences, Central University of Ecuador, Quito 170113, Ecuador
| | | |
Collapse
|
12
|
Joglekar MV, Sahu S, Wong WKM, Satoor SN, Dong CX, Farr RJ, Williams MD, Pandya P, Jhala G, Yang SNY, Chew YV, Hetherington N, Thiruchevlam D, Mitnala S, Rao GV, Reddy DN, Loudovaris T, Hawthorne WJ, Elefanty AG, Joglekar VM, Stanley EG, Martin D, Thomas HE, Tosh D, Dalgaard LT, Hardikar AA. A Pro-Endocrine Pancreatic Islet Transcriptional Program Established During Development Is Retained in Human Gallbladder Epithelial Cells. Cell Mol Gastroenterol Hepatol 2022; 13:1530-1553.e4. [PMID: 35032693 PMCID: PMC9043310 DOI: 10.1016/j.jcmgh.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS Pancreatic islet β-cells are factories for insulin production; however, ectopic expression of insulin also is well recognized. The gallbladder is a next-door neighbor to the developing pancreas. Here, we wanted to understand if gallbladders contain functional insulin-producing cells. METHODS We compared developing and adult mouse as well as human gallbladder epithelial cells and islets using immunohistochemistry, flow cytometry, enzyme-linked immunosorbent assays, RNA sequencing, real-time polymerase chain reaction, chromatin immunoprecipitation, and functional studies. RESULTS We show that the epithelial lining of developing, as well as adult, mouse and human gallbladders naturally contain interspersed cells that retain the capacity to actively transcribe, translate, package, and release insulin. We show that human gallbladders also contain functional insulin-secreting cells with the potential to naturally respond to glucose in vitro and in situ. Notably, in a non-obese diabetic (NOD) mouse model of type 1 diabetes, we observed that insulin-producing cells in the gallbladder are not targeted by autoimmune cells. Interestingly, in human gallbladders, insulin splice variants are absent, although insulin splice forms are observed in human islets. CONCLUSIONS In summary, our biochemical, transcriptomic, and functional data in mouse and human gallbladder epithelial cells collectively show the evolutionary and developmental similarities between gallbladder and the pancreas that allow gallbladder epithelial cells to continue insulin production in adult life. Understanding the mechanisms regulating insulin transcription and translation in gallbladder epithelial cells would help guide future studies in type 1 diabetes therapy.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Subhshri Sahu
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Wilson K M Wong
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Sarang N Satoor
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Charlotte X Dong
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Ryan J Farr
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Michael D Williams
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Prapti Pandya
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Gaurang Jhala
- Immunology and Diabetes Group, St. Vincent's Institute for Medical Research, Victoria, Australia
| | - Sundy N Y Yang
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Yi Vee Chew
- The Westmead Institute for Medical Research, Westmead Millenium Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Nicola Hetherington
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Dhan Thiruchevlam
- Department of Gastroenterology, St. Vincent's Hospital, Melbourne, Victoria, Australia
| | - Sasikala Mitnala
- Surgical Gastroenterology Research, Asian Institute of Gastroenterology, Hyderabad, India
| | - Guduru V Rao
- Surgical Gastroenterology Research, Asian Institute of Gastroenterology, Hyderabad, India
| | | | - Thomas Loudovaris
- Immunology and Diabetes Group, St. Vincent's Institute for Medical Research, Victoria, Australia
| | - Wayne J Hawthorne
- The Westmead Institute for Medical Research, Westmead Millenium Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Andrew G Elefanty
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | | | - Edouard G Stanley
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - David Martin
- Upper Gastrointestinal Surgery, Strathfield Hospital, Strathfield, New South Wales, Australia
| | - Helen E Thomas
- Immunology and Diabetes Group, St. Vincent's Institute for Medical Research, Victoria, Australia
| | - David Tosh
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Louise T Dalgaard
- Section of Eukaryotic Cell Biology, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia.
| |
Collapse
|
13
|
Parte S, Nimmakayala RK, Batra SK, Ponnusamy MP. Acinar to ductal cell trans-differentiation: A prelude to dysplasia and pancreatic ductal adenocarcinoma. Biochim Biophys Acta Rev Cancer 2022; 1877:188669. [PMID: 34915061 DOI: 10.1016/j.bbcan.2021.188669] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer (PC) is the deadliest neoplastic epithelial malignancies and is projected to be the second leading cause of cancer-related mortality by 2024. Five years overall survival being ~10%, mortality and incidence rates are disturbing. Acinar to ductal cell metaplasia (ADM) encompasses cellular reprogramming and phenotypic switch-over, making it a cardinal event in tumor initiation. Differential cues and varied regulatory factors drive synchronous functions of metaplastic cell populations leading to multiple cell fates and physiological outcomes. ADM is a precursor for developing early pre-neoplastic lesions further progressing into PC due to oncogenic signaling. Hence delineating molecular events guiding tumor initiation may provide cues for regenerative medicine and precision onco-medicine. Therefore, understanding PC pathogenesis and early diagnosis are crucial. We hereby provide a timely overview of the current progress in this direction and future perspectives we foresee unfolding in the best interest of patient well-being and better clinical management of PC.
Collapse
Affiliation(s)
- Seema Parte
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
14
|
Manipulating cellular microRNAs and analyzing high-dimensional gene expression data using machine learning workflows. STAR Protoc 2021; 2:100910. [PMID: 34746868 PMCID: PMC8554629 DOI: 10.1016/j.xpro.2021.100910] [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: 11/29/2022] Open
Abstract
MicroRNAs (miRNAs) are elements of the gene regulatory network and manipulating their abundance is essential toward elucidating their role in patho-physiological conditions. We present a detailed workflow that identifies important miRNAs using a machine learning algorithm. We then provide optimized techniques to validate the identified miRNAs through over-expression/loss-of-function studies. Overall, these protocols apply to any field in biology where high-dimensional data are produced. For complete details on the use and execution of this protocol, please refer to Wong et al. (2021a). LASSO and bootstrapping identify important miRNAs associated with gene of interest Generating puromycin resistant, doxycycline inducible, miRNA overexpressing cells Transient miRNA knockdown using LNA inhibitors in human primary cells Optimized reagent concentrations and cell densities for miRNA manipulation
Collapse
|
15
|
Lien YC, Lu XM, Won KJ, Wang PZ, Osei-Bonsu W, Simmons RA. The Transcriptome and Epigenome Reveal Novel Changes in Transcription Regulation During Pancreatic Rat Islet Maturation. Endocrinology 2021; 162:6360893. [PMID: 34467975 PMCID: PMC8455347 DOI: 10.1210/endocr/bqab181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Indexed: 01/03/2023]
Abstract
Islet function is critical for normal glucose homeostasis. Unlike adult β cells, fetal and neonatal islets are more proliferative and have decreased insulin secretion in response to stimuli. However, the underlying mechanisms governing functional maturity of islets have not been completely elucidated. Pancreatic islets comprise different cell types. The microenvironment of islets and interactions between these cell types are critical for β-cell development and maturation. Thus, the study of intact islets is optimal to identify novel molecular mechanisms controlling islet functional development. Transcriptomes and genome-wide histone landscapes of H3K4me3, H3K27me3, and H3K27Ac from intact islets isolated from 2- and 10-week-old Sprague-Dawley rats were integrated to elucidate genes and pathways modulating islet development, as well as the contribution of epigenetic regulation. A total of 4489 differentially expressed genes were identified; 2289 and 2200 of them were up- and down-regulated in 10-week islets, respectively. Ingenuity Pathway Analysis revealed critical pathways regulating functional maturation of islets, including nutrient sensing, neuronal function, immune function, cell replication, and extracellular matrix. Furthermore, we identified significant changes in enrichment of H3K4me3, H3K27me3, and H3K27Ac marks, which correlated with expression changes of genes critical for islet function. These histone marks were enriched at critical transcription factor-binding motifs, such as Hoxa9, C/EBP-β, Gata1, Foxo1, E2f1, E2f3, and Mafb. In addition, our chromatin immunoprecipitation sequencing data revealed multiple potential bivalent genes whose poised states changed with maturation. Collectively, our current study identified critical novel pathways for mature islet function and suggested a role for histone modifications in regulating islet development and maturation.
Collapse
Affiliation(s)
- Yu-Chin Lien
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Xueqing Maggie Lu
- Institute for Biomedical Informatics, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyoung-Jae Won
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Paul Zhiping Wang
- Institute for Biomedical Informatics, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wendy Osei-Bonsu
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Correspondence: Rebecca A. Simmons, MD, BRB II/III, 13th Floor, Rm 1308, 421 Curie Blvd, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Joglekar MV, Dong CX, Wong WKM, Dalgaard LT, Hardikar AA. A bird's eye view of the dynamics of pancreatic β-cell heterogeneity. Acta Physiol (Oxf) 2021; 233:e13664. [PMID: 33884752 DOI: 10.1111/apha.13664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Mugdha V. Joglekar
- Diabetes and Islet Biology Group, School of Medicine Western Sydney University Campbelltown NSW Australia
| | - Charlotte X. Dong
- Diabetes and Islet Biology Group, School of Medicine Western Sydney University Campbelltown NSW Australia
| | - Wilson K. M. Wong
- Diabetes and Islet Biology Group, School of Medicine Western Sydney University Campbelltown NSW Australia
| | | | - Anandwardhan A. Hardikar
- Diabetes and Islet Biology Group, School of Medicine Western Sydney University Campbelltown NSW Australia
- Department of Science and Environment Roskilde University Roskilde Denmark
| |
Collapse
|
17
|
Pnueli L, Shalev D, Refael T, David C, Boehm U, Melamed P. Proliferating primary pituitary cells as a model for studying regulation of gonadotrope chromatin and gene expression. Mol Cell Endocrinol 2021; 533:111349. [PMID: 34090968 DOI: 10.1016/j.mce.2021.111349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/20/2021] [Accepted: 05/29/2021] [Indexed: 12/27/2022]
Abstract
The chromatin organization of the gonadotropin gene promoters in the pituitary gonadotropes plays a major role in determining how these gene are activated, but is difficult to study because of the low numbers of these cells in the pituitary gland. Here, we set out to create a cell model to study gonadotropin chromatin, and found that by optimizing cell culture conditions, we can maintain stable proliferating cultures of primary non-transformed gonadotrope cells over weeks to months. Although expression of the gonadotropin genes drops very low, these cells are enriched in gonadotrope markers and respond to GnRH. Furthermore, >85% of the cells contained Lhb and/or Fshb mature transcripts; though these were virtually restricted to the nuclei. The gonadotropes were harvested initially due to expression of dTOMATO, following activation of Cre recombinase by the Gnrhr promoter. Over 6 mo in culture, a similar proportion of the recombined DNA was maintained (i.e. cells derived from the original gonadotropes or having acquired Gnrhr-promoter activity), together with cells of a distinct origin. The cells are enriched with markers of proliferating pituitary and stem cells, including Sox2, suggesting that multipotent precursor cells might have proliferated and differentiated into gonadotrope-like cells. These cell cultures offer a new and versatile methodology for research in gonadotrope differentiation and function, and can provide enough primary cells for chromatin immunoprecipitation and epigenetic analysis, while our initial studies also indicate a possible regulatory mechanism that might be involved in the nuclear export of gonadotropin gene mRNAs.
Collapse
Affiliation(s)
- Lilach Pnueli
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Dor Shalev
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Tal Refael
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Cfir David
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg, Germany
| | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| |
Collapse
|
18
|
Wong WK, Joglekar MV, Saini V, Jiang G, Dong CX, Chaitarvornkit A, Maciag GJ, Gerace D, Farr RJ, Satoor SN, Sahu S, Sharangdhar T, Ahmed AS, Chew YV, Liuwantara D, Heng B, Lim CK, Hunter J, Januszewski AS, Sørensen AE, Akil AS, Gamble JR, Loudovaris T, Kay TW, Thomas HE, O'Connell PJ, Guillemin GJ, Martin D, Simpson AM, Hawthorne WJ, Dalgaard LT, Ma RC, Hardikar AA. Machine learning workflows identify a microRNA signature of insulin transcription in human tissues. iScience 2021; 24:102379. [PMID: 33981968 PMCID: PMC8082091 DOI: 10.1016/j.isci.2021.102379] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/19/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Dicer knockout mouse models demonstrated a key role for microRNAs in pancreatic β-cell function. Studies to identify specific microRNA(s) associated with human (pro-)endocrine gene expression are needed. We profiled microRNAs and key pancreatic genes in 353 human tissue samples. Machine learning workflows identified microRNAs associated with (pro-)insulin transcripts in a discovery set of islets (n = 30) and insulin-negative tissues (n = 62). This microRNA signature was validated in remaining 261 tissues that include nine islet samples from individuals with type 2 diabetes. Top eight microRNAs (miR-183-5p, -375-3p, 216b-5p, 183-3p, -7-5p, -217-5p, -7-2-3p, and -429-3p) were confirmed to be associated with and predictive of (pro-)insulin transcript levels. Use of doxycycline-inducible microRNA-overexpressing human pancreatic duct cell lines confirmed the regulatory roles of these microRNAs in (pro-)endocrine gene expression. Knockdown of these microRNAs in human islet cells reduced (pro-)insulin transcript abundance. Our data provide specific microRNAs to further study microRNA-mRNA interactions in regulating insulin transcription.
Collapse
Affiliation(s)
- Wilson K.M. Wong
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Mugdha V. Joglekar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Vijit Saini
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- School of Life Sciences and the Centre for Health Technologies, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Guozhi Jiang
- Department of Medicine and Therapeutics, and Hong Kong Institute of Diabetes and Obesity, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Special Administrative Region, China
| | - Charlotte X. Dong
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Alissa Chaitarvornkit
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Grzegorz J. Maciag
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Dario Gerace
- School of Life Sciences and the Centre for Health Technologies, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Ryan J. Farr
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Sarang N. Satoor
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Subhshri Sahu
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Tejaswini Sharangdhar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Asma S. Ahmed
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Yi Vee Chew
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Road, Westmead, NSW 2145, Australia
| | - David Liuwantara
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Benjamin Heng
- Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2019, Australia
| | - Chai K. Lim
- Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2019, Australia
| | - Julie Hunter
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, University of Sydney Medical School, Locked Bag #6, Newtown, NSW 2042, Australia
| | - Andrzej S. Januszewski
- NHMRC Clinical Trials Centre, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Anja E. Sørensen
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Ammira S.A. Akil
- Department of Human Genetics-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Jennifer R. Gamble
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, University of Sydney Medical School, Locked Bag #6, Newtown, NSW 2042, Australia
| | - Thomas Loudovaris
- St Vincent's Institute and The University of Melbourne Department of Medicine, 9 Princes Street, Fitzroy, VIC, Australia
| | - Thomas W. Kay
- St Vincent's Institute and The University of Melbourne Department of Medicine, 9 Princes Street, Fitzroy, VIC, Australia
| | - Helen E. Thomas
- St Vincent's Institute and The University of Melbourne Department of Medicine, 9 Princes Street, Fitzroy, VIC, Australia
| | - Philip J. O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Gilles J. Guillemin
- Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2019, Australia
| | - David Martin
- Upper GI Surgery, Strathfield Hospital, 2/3 Everton Road, Strathfield, NSW 2135, Australia
| | - Ann M. Simpson
- School of Life Sciences and the Centre for Health Technologies, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Wayne J. Hawthorne
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Louise T. Dalgaard
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Ronald C.W. Ma
- Department of Medicine and Therapeutics, and Hong Kong Institute of Diabetes and Obesity, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Special Administrative Region, China
| | - Anandwardhan A. Hardikar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW 2560, Australia
- Diabetes and Islet Biology group, Faculty of Medicine and Health, University of Sydney, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| |
Collapse
|
19
|
Lorenzo PI, Cobo-Vuilleumier N, Martín-Vázquez E, López-Noriega L, Gauthier BR. Harnessing the Endogenous Plasticity of Pancreatic Islets: A Feasible Regenerative Medicine Therapy for Diabetes? Int J Mol Sci 2021; 22:4239. [PMID: 33921851 PMCID: PMC8073058 DOI: 10.3390/ijms22084239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes is a chronic metabolic disease caused by an absolute or relative deficiency in functional pancreatic β-cells that leads to defective control of blood glucose. Current treatments for diabetes, despite their great beneficial effects on clinical symptoms, are not curative treatments, leading to a chronic dependence on insulin throughout life that does not prevent the secondary complications associated with diabetes. The overwhelming increase in DM incidence has led to a search for novel antidiabetic therapies aiming at the regeneration of the lost functional β-cells to allow the re-establishment of the endogenous glucose homeostasis. Here we review several aspects that must be considered for the development of novel and successful regenerative therapies for diabetes: first, the need to maintain the heterogeneity of islet β-cells with several subpopulations of β-cells characterized by different transcriptomic profiles correlating with differences in functionality and in resistance/behavior under stress conditions; second, the existence of an intrinsic islet plasticity that allows stimulus-mediated transcriptome alterations that trigger the transdifferentiation of islet non-β-cells into β-cells; and finally, the possibility of using agents that promote a fully functional/mature β-cell phenotype to reduce and reverse the process of dedifferentiation of β-cells during diabetes.
Collapse
Affiliation(s)
- Petra I. Lorenzo
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Nadia Cobo-Vuilleumier
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Eugenia Martín-Vázquez
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Livia López-Noriega
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
| | - Benoit R. Gauthier
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain; (N.C.-V.); (E.M.-V.); (L.L.-N.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 028029 Madrid, Spain
| |
Collapse
|
20
|
Honzawa N, Fujimoto K. The Plasticity of Pancreatic β-Cells. Metabolites 2021; 11:metabo11040218. [PMID: 33918379 PMCID: PMC8065544 DOI: 10.3390/metabo11040218] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes is caused by impaired insulin secretion and/or insulin resistance. Loss of pancreatic β-cell mass detected in human diabetic patients has been considered to be a major cause of impaired insulin secretion. Additionally, apoptosis is found in pancreatic β-cells; β-cell mass loss is induced when cell death exceeds proliferation. Recently, however, β-cell dedifferentiation to pancreatic endocrine progenitor cells and β-cell transdifferentiation to α-cell was reported in human islets, which led to a new underlying molecular mechanism. Hyperglycemia inhibits nuclear translocation and expression of forkhead box-O1 (FoxO1) and induces the expression of neurogenin-3 (Ngn3), which is required for the development and maintenance of pancreatic endocrine progenitor cells. This new hypothesis (Foxology) is attracting attention because it explains molecular mechanism(s) underlying β-cell plasticity. The lineage tracing technique revealed that the contribution of dedifferentiation is higher than that of β-cell apoptosis retaining to β-cell mass loss. In addition, islet cells transdifferentiate each other, such as transdifferentiation of pancreatic β-cell to α-cell and vice versa. Islet cells can exhibit plasticity, and they may have the ability to redifferentiate into any cell type. This review describes recent findings in the dedifferentiation and transdifferentiation of β-cells. We outline novel treatment(s) for diabetes targeting islet cell plasticity.
Collapse
Affiliation(s)
- Norikiyo Honzawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8, Nishishinbashi, Minato-ku, Tokyo 105-8461, Japan;
| | - Kei Fujimoto
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University Kashiwa Hospital, 163-1, Kashiwashita, Kshiwa-shi, Chiba 277-8567, Japan
- Correspondence: ; Tel.: +81-04-7164-1111
| |
Collapse
|
21
|
Lafferty RA, Tanday N, Moffett RC, Reimann F, Gribble FM, Flatt PR, Irwin N. Positive Effects of NPY1 Receptor Activation on Islet Structure Are Driven by Pancreatic Alpha- and Beta-Cell Transdifferentiation in Diabetic Mice. Front Endocrinol (Lausanne) 2021; 12:633625. [PMID: 33716983 PMCID: PMC7949013 DOI: 10.3389/fendo.2021.633625] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/20/2021] [Indexed: 02/03/2023] Open
Abstract
Enzymatically stable and specific neuropeptide Y1 receptor (NPYR1) agonists, such as sea lamprey PYY(1-36) (SL-PYY(1-36)), are believed to improve glucose regulation in diabetes by targeting pancreatic islets. In this study, streptozotocin (STZ) diabetic transgenic GluCreERT2 ;ROSA26-eYFP and Ins1Cre/+;Rosa26-eYFP mouse models have been used to study effects of sustained NPYR1 activation on islet cell composition and alpha- and beta-cell lineage transitioning. STZ induced a particularly severe form of diabetes in Ins1Cre/+;Rosa26-eYFP mice, but twice-daily administration (25 nmol/kg) of SL-PYY(1-36) for 11 days consistently improved metabolic status. Blood glucose was decreased (p < 0.05 - p < 0.001) and both fasted plasma and pancreatic insulin significantly increased by SL-PYY(1-36). In both GluCreERT2 ;ROSA26-eYFP and Ins1Cre/+; Rosa26-eYFP mice, STZ provoked characteristic losses (p < 0.05 - p < 0.001) of islet numbers, beta-cell and pancreatic islet areas together with increases in area and central islet location of alpha-cells. With exception of alpha-cell area, these morphological changes were fully, or partially, returned to non-diabetic control levels by SL-PYY(1-36). Interestingly, STZ apparently triggered decreased (p < 0.001) alpha- to beta-cell transition in GluCreERT2 ;ROSA26-eYFP mice, together with increased loss of beta-cell identity in Ins1Cre/+;Rosa26-eYFP mice, but both effects were significantly (p < 0.001) reversed by SL-PYY(1-36). SL-PYY(1-36) also apparently reduced (p < 0.05) beta- to alpha-cell conversion in Ins1Cre/+;Rosa26-eYFP mice and glucagon expressing alpha-cells in GluCreERT2 ;ROSA26-eYFP mice. These data indicate that islet benefits of prolonged NPY1R activation, and especially restoration of beta-cell mass, are observed irrespective of diabetes status, being linked to cell lineage alterations including transdifferentiation of alpha- to beta-cells.
Collapse
Affiliation(s)
- Ryan A. Lafferty
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Neil Tanday
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - R. Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Fiona M. Gribble
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Peter R. Flatt
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| |
Collapse
|
22
|
Bilekova S, Sachs S, Lickert H. Pharmacological Targeting of Endoplasmic Reticulum Stress in Pancreatic Beta Cells. Trends Pharmacol Sci 2020; 42:85-95. [PMID: 33353789 DOI: 10.1016/j.tips.2020.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Diabetes is a disease with pandemic dimensions and no pharmacological treatment prevents disease progression. Dedifferentiation has been proposed to be a driver of beta-cell dysfunction in both type 1 and type 2 diabetes. Regenerative therapies aim to re-establish function in dysfunctional or dedifferentiated beta cells and restore the defective insulin secretion. Unsustainable levels of insulin production, with increased demand at disease onset, strain the beta-cell secretory machinery, leading to endoplasmic reticulum (ER) stress. Unresolved chronic ER stress is a major contributor to beta-cell loss of function and identity. Restoring ER homeostasis, enhancing ER-associated degradation of misfolded protein, and boosting chaperoning activity, are emerging therapeutic approaches for diabetes treatment.
Collapse
Affiliation(s)
- Sara Bilekova
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Technical University of Munich, Medical Faculty, Munich, Germany
| | - Stephan Sachs
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Technical University of Munich, Medical Faculty, Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Technical University of Munich, Medical Faculty, Munich, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
23
|
Abadpour S, Aizenshtadt A, Olsen PA, Shoji K, Wilson SR, Krauss S, Scholz H. Pancreas-on-a-Chip Technology for Transplantation Applications. Curr Diab Rep 2020; 20:72. [PMID: 33206261 PMCID: PMC7674381 DOI: 10.1007/s11892-020-01357-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Human pancreas-on-a-chip (PoC) technology is quickly advancing as a platform for complex in vitro modeling of islet physiology. This review summarizes the current progress and evaluates the possibility of using this technology for clinical islet transplantation. RECENT FINDINGS PoC microfluidic platforms have mainly shown proof of principle for long-term culturing of islets to study islet function in a standardized format. Advancement in microfluidic design by using imaging-compatible biomaterials and biosensor technology might provide a novel future tool for predicting islet transplantation outcome. Progress in combining islets with other tissue types gives a possibility to study diabetic interventions in a minimal equivalent in vitro environment. Although the field of PoC is still in its infancy, considerable progress in the development of functional systems has brought the technology on the verge of a general applicable tool that may be used to study islet quality and to replace animal testing in the development of diabetes interventions.
Collapse
Affiliation(s)
- Shadab Abadpour
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Post Box 4950, Nydalen, N-0424 Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Aleksandra Aizenshtadt
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Petter Angell Olsen
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kayoko Shoji
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Steven Ray Wilson
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Chemistry, University of Oslo, Oslo, Norway
| | - Stefan Krauss
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital, Oslo, Norway
| | - Hanne Scholz
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Post Box 4950, Nydalen, N-0424 Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
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
|
25
|
Direct suppression of human islet dedifferentiation, progenitor genes, but not epithelial to mesenchymal transition by liraglutide. Heliyon 2020; 6:e04951. [PMID: 32995630 PMCID: PMC7501427 DOI: 10.1016/j.heliyon.2020.e04951] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/03/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
β-cell dedifferentiation has been accounted as one of the major mechanisms for β-cell failure; thus, is a cause to diabetes. We study direct impacts of liraglutide treatment on ex vivo human dedifferentiated islets, and its effects on genes important in endocrine function, progenitor states, and epithelial mesenchymal transition (EMT). Human islets from non-diabetic donors, were purified and incubated until day 1 and day 4, and were determined insulin contents, numbers of insulin (INS+) and glucagon (GCG+) cells. The islets from day 3 to day 7 were treated with diabetic drugs, the long acting GLP-1 receptor agonist, liraglutide. As observed in pancreatic islets of type 2 diabetic patients, ex vivo dedifferentiated islets showed more than 50% reduced insulin contents while number of glucagon increased from 10% to about 20%. β-cell specific genes: PDX1, MAFA, as well as β-cell functional markers: GLUT1 and SUR1, were significantly depleted more than 40%. Notably, we found increased levels of glucagon regulator, ARX and pre-glucagon transcripts, and remarkably upregulated progenitor expressions: NEUROG3 and ALDH1A identified as β-cell dysfunction markers in diabetic models. Hyperglucagonemia was often observed in type 2 patients that could lead to over production of gluconeogenesis by the liver. Liraglutide treatments resulted in decreased number of GCG+ cells, increased numbers of GLP-1 positive cells but did not alter elevated levels of EMT marker genes: ACTA2, CDH-2, SNAIL2, and VIM. These effects of liraglutide were blunted when FOXO1 transcripts were depleted. This work illustrates that ex vivo human isolated islets can be used as a tool to study different aspects of β-cell dedifferentiation. Our novel finding suggests a role of GLP-1 pathway in beta-cell maintenance in FOXO1-dependent manner. Importantly, dedifferentiated islets ex vivo is a useful model that can be utilized to verify the actions of potential drugs to diabetic β-cell failure.
Collapse
|
26
|
Thirlwell KL, Colligan D, Mountford JC, Samuel K, Bailey L, Cuesta-Gomez N, Hewit KD, Kelly CJ, West CC, McGowan NWA, Casey JJ, Graham GJ, Turner ML, Forbes S, Campbell JDM. Pancreas-derived mesenchymal stromal cells share immune response-modulating and angiogenic potential with bone marrow mesenchymal stromal cells and can be grown to therapeutic scale under Good Manufacturing Practice conditions. Cytotherapy 2020; 22:762-771. [PMID: 32828673 DOI: 10.1016/j.jcyt.2020.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) isolated from various tissues are under investigation as cellular therapeutics in a wide range of diseases. It is appreciated that the basic biological functions of MSCs vary depending on tissue source. However, in-depth comparative analyses between MSCs isolated from different tissue sources under Good Manufacturing Practice (GMP) conditions are lacking. Human clinical-grade low-purity islet (LPI) fractions are generated as a byproduct of islet isolation for transplantation. MSC isolates were derived from LPI fractions with the aim of performing a systematic, standardized comparative analysis of these cells with clinically relevant bone marrow-derived MSCs (BM MSCs). METHODS MSC isolates were derived from LPI fractions and expanded in platelet lysate-supplemented medium or in commercially available xenogeneic-free medium. Doubling rate, phenotype, differentiation potential, gene expression, protein production and immunomodulatory capacity of LPIs were compared with those of BM MSCs. RESULTS MSCs can be readily derived in vitro from non-transplanted fractions resulting from islet cell processing (i.e., LPI MSCs). LPI MSCs grow stably in serum-free or platelet lysate-supplemented media and demonstrate in vitro self-renewal, as measured by colony-forming unit assay. LPI MSCs express patterns of chemokines and pro-regenerative factors similar to those of BM MSCs and, importantly, are equally able to attract immune cells in vitro and in vivo and suppress T-cell proliferation in vitro. Additionally, LPI MSCs can be expanded to therapeutically relevant doses at low passage under GMP conditions. CONCLUSIONS LPI MSCs represent an alternative source of GMP MSCs with functions comparable to BM MSCs.
Collapse
Affiliation(s)
- Kayleigh L Thirlwell
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK; Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - David Colligan
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Joanne C Mountford
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Kay Samuel
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Laura Bailey
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Nerea Cuesta-Gomez
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Kay D Hewit
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK; Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Christopher J Kelly
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | | - Neil W A McGowan
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK
| | - John J Casey
- Transplant Unit, National Islet Transplant Programme, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Marc L Turner
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Shareen Forbes
- University/British Heart Foundation Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK; Transplant Unit, National Islet Transplant Programme, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - John D M Campbell
- Tissues, Cells and Advanced Therapeutics, The Jack Copland Centre, Scottish National Blood Transfusion Service, Edinburgh, UK; Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
27
|
Williams MD, Joglekar MV, Hardikar AA, Wong WKM. Directed differentiation into insulin-producing cells using microRNA manipulation. Open Med (Wars) 2020; 15:567-570. [PMID: 33336012 PMCID: PMC7711856 DOI: 10.1515/med-2020-0170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/28/2019] [Indexed: 02/04/2023] Open
Abstract
Our commentary is focused on three studies that used microRNA overexpression methods for directed differentiation of stem cells into insulin-producing cells. Islet transplantation is the only cell-based therapy used to treat type 1 diabetes mellitus. However, due to the scarcity of cadaveric donors and limited availability of good quality and quantity of islets for transplant, alternate sources of insulin-producing cells are being studied and used by researchers. This commentary provides an overview of distinct studies focused on manipulating microRNA expression to optimize differentiation of embryonic stem cells or induced pluripotent stem cells into insulin-producing cells. These studies have used different approaches to overexpress microRNAs that are highly abundant in human islets (such as miR-375 and miR-7) in their differentiation protocol to achieve better differentiation into functional islet beta (β)-cells.
Collapse
Affiliation(s)
- Michael D Williams
- Diabetes and Islet biology Group, NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Mugdha V Joglekar
- Diabetes and Islet biology Group, NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet biology Group, NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| | - Wilson K M Wong
- Diabetes and Islet biology Group, NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Level 6, Medical Foundation Building, 92-94 Parramatta Road, Camperdown, NSW 2050, Australia
| |
Collapse
|
28
|
Barraclough JY, Joglekar MV, Januszewski AS, Martínez G, Celermajer DS, Keech AC, Hardikar AA, Patel S. A MicroRNA Signature in Acute Coronary Syndrome Patients and Modulation by Colchicine. J Cardiovasc Pharmacol Ther 2020; 25:444-455. [PMID: 32356454 DOI: 10.1177/1074248420922793] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Circulating microRNAs (miRNAs) may play a pathogenic role in acute coronary syndromes (ACS). It is not yet known if miRNAs dysregulated in ACS are modulated by colchicine. We profiled miRNAs in plasma samples simultaneously collected from the aorta, coronary sinus, and right atrium in patients with ACS. METHODS A total of 396 of 754 miRNAs were detected by TaqMan real-time polymerase chain reaction from EDTA-plasma in a discovery cohort of 15 patients (n = 3 controls, n = 6 ACS standard therapy, n = 6 ACS standard therapy plus colchicine). Fifty-one significantly different miRNAs were then measured in a verification cohort of 92 patients (n = 13 controls, n = 40 ACS standard therapy, n = 39 ACS standard therapy plus colchicine). Samples were simultaneously obtained from the coronary sinus, aortic root, and right atrium. RESULTS Circulating levels of 30 of 51 measured miRNAs were higher in ACS standard therapy patients compared to controls. In patients with ACS, levels of 12 miRNAs (miR-17, -106b-3p, -191, -106a, -146a, -130a, -223, -484, -889, -425-3p, -629, -142-5p) were lower with colchicine treatment. Levels of 7 of these 12 miRNA were higher in ACS standard therapy patients compared to controls and returned to levels seen in control individuals after colchicine treatment. Three miRNAs suppressed by colchicine (miR-146a, miR-17, miR-130a) were identified as regulators of inflammatory pathways. MicroRNAs were comparable across sampling sites with select differences in the transcoronary gradient of 4 miRNA. CONCLUSION The levels of specific miRNAs elevated in ACS returned to levels similar to control individuals following colchicine. These miRNAs may mediate ACS (via inflammatory pathways) or increase post-ACS risk, and could be potentially used as biomarkers of treatment efficacy.
Collapse
Affiliation(s)
- Jennifer Y Barraclough
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.,Sydney Medical School, The University of Sydney, Australia.,Heart Research Institute Sydney, Australia
| | - Mugdha V Joglekar
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Andrzej S Januszewski
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Gonzalo Martínez
- Heart Research Institute Sydney, Australia.,Division of Cardiovascular Diseases, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David S Celermajer
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.,Sydney Medical School, The University of Sydney, Australia.,Heart Research Institute Sydney, Australia
| | - Anthony C Keech
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.,NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Anandwardhan A Hardikar
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Sanjay Patel
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.,Sydney Medical School, The University of Sydney, Australia.,Heart Research Institute Sydney, Australia
| |
Collapse
|
29
|
Tanday N, Irwin N, Flatt PR, Moffett RC. Dapagliflozin exerts positive effects on beta cells, decreases glucagon and does not alter beta- to alpha-cell transdifferentiation in mouse models of diabetes and insulin resistance. Biochem Pharmacol 2020; 177:114009. [PMID: 32360307 DOI: 10.1016/j.bcp.2020.114009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022]
Abstract
Loss of beta cell identity and subsequent transdifferentiation of beta-to-alpha cells is implicated in the pathogenesis of diabetes. In addition, sodium-glucose transport protein 2 (SGLT2) inhibition has been linked to altered alpha-cell function. To investigate these phenomenon, lineage tracing of beta-cells was examined following 10-12 days dapagliflozin (1 or 5 mg/kg, once daily, as appropriate) treatment in multiple low-dose streptozotocin (STZ), high fat fed (HFF) or hydrocortisone (HC) transgenic Ins1Cre/+/Rosa26-eYFP mouse models of diabetes and insulin resistance. As anticipated, STZ, HFF and HC treated mice developed characteristic features of insulin deficiency or resistance. Dapagliflozin elicited differing beneficial effects depending on the aetiology of syndrome studied. The SGLT2 inhibitor efficiently promoted (P < 0.001) weight loss in HFF and STZ mice, whilst in HC mice it reduced (P < 0.001) energy intake, without an impact on body weight. Despite lacking significant effects on glycaemia, 1 mg/kg dapagliflozin consistently decreased both plasma and pancreatic glucagon. This was associated with increased pancreatic insulin in STZ and HFF mice. In STZ and HFF mice, beta cell proliferation and Pdx1 expression were enhanced by dapagliflozin, with a further increase in overall glucagon staining in HFF islets. Islet, beta- and alpha-cell areas were increased in dapagliflozin treated HC mice, which appeared to be linked to decreased alpha- and beta-cell apoptosis. Although the diabetes-like syndromes induced clear alterations in islet cell transdifferentiation, treatment with dapagliflozin (1 mg/kg) had no significant impact on this process, with 5 mg/kg marginally decreasing loss of beta-cells identity in STZ mice. These data suggest that SGLT2 inhibitors have positive effects on beta cells and decrease plasma and pancreatic glucagon, independent of changes in ambient glucose levels. Our combined data indicate that SGLT2 inhibitors do not directly induce hyperglucagonaemia.
Collapse
Affiliation(s)
- Neil Tanday
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK.
| | - Peter R Flatt
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - R Charlotte Moffett
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
30
|
Villard O, Armanet M, Couderc G, Bony C, Moreaux J, Noël D, De Vos J, Klein B, Veyrune JL, Wojtusciszyn A. Characterization of immortalized human islet stromal cells reveals a MSC-like profile with pancreatic features. Stem Cell Res Ther 2020; 11:158. [PMID: 32303252 PMCID: PMC7165390 DOI: 10.1186/s13287-020-01649-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) represent an interesting tool to improve pancreatic islet transplantation. They have immunomodulatory properties and secrete supportive proteins. However, the functional properties of MSCs vary according to many factors such as donor characteristics, tissue origin, or isolation methods. To counteract this heterogeneity, we aimed to immortalize and characterize adherent cells derived from human pancreatic islets (hISCs), using phenotypic, transcriptomic, and functional analysis. METHODS Adherent cells derived from human islets in culture were infected with a hTERT retrovirus vector and then characterized by microarray hybridization, flow cytometry analysis, and immunofluorescence assays. Osteogenic, adipogenic, and chondrogenic differentiation as well as PBMC proliferation suppression assays were used to compare the functional abilities of hISCs and MSCs. Extracellular matrix (ECM) gene expression profile analysis was performed using the SAM (Significance Analysis of Microarrays) software, and protein expression was confirmed by western blotting. RESULTS hISCs kept an unlimited proliferative potential. They exhibited several properties of MSCs such as CD73, CD90, and CD105 expression and differentiation capacity. From a functional point of view, hISCs inhibited the proliferation of activated peripheral blood mononuclear cells. The transcriptomic profile of hISCs highly clusterized with bone marrow (BM)-MSCs and revealed a differential enrichment of genes involved in the organization of the ECM. Indeed, the expression and secretion profiles of ECM proteins including collagens I, IV, and VI, fibronectin, and laminins, known to be expressed in abundance around and within the islets, were different between hISCs and BM-MSCs. CONCLUSION We generated a new human cell line from pancreatic islets, with MSCs properties and retaining some pancreatic specificities related to the production of ECM proteins. hISCs appear as a very promising tool in islet transplantation by their availability (as a source of inexhaustible source of cells) and ability to secrete a supportive "pancreatic" microenvironment.
Collapse
Affiliation(s)
- Orianne Villard
- Laboratory of Cell Therapy for Diabetes, Institute of Regenerative Medicine and Biotherapy, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Endocrinology, Diabetes, and Nutrition, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Mathieu Armanet
- Laboratory of Cell Therapy for Diabetes, Institute of Regenerative Medicine and Biotherapy, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Cell Therapy Unit, Hospital Saint- Louis, AP-HP, Paris, France
- Department of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, 8 avenue de la Sallaz - 1011, Lausanne, Switzerland
| | - Guilhem Couderc
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Claire Bony
- IRMB, INSERM U 1183, Univ Montpellier, INSERM, Montpellier, France
| | - Jerome Moreaux
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- IGH, Univ Montpellier, CNRS, Montpellier, France
| | - Daniele Noël
- IRMB, INSERM U 1183, Univ Montpellier, INSERM, Montpellier, France
| | - John De Vos
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
- IRMB, INSERM U 1183, Univ Montpellier, INSERM, Montpellier, France
| | - Bernard Klein
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Jean-Luc Veyrune
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Anne Wojtusciszyn
- Laboratory of Cell Therapy for Diabetes, Institute of Regenerative Medicine and Biotherapy, Univ. Montpellier, CHU Montpellier, Montpellier, France.
- Department of Endocrinology, Diabetes, and Nutrition, Univ. Montpellier, CHU Montpellier, Montpellier, France.
- Department of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, 8 avenue de la Sallaz - 1011, Lausanne, Switzerland.
| |
Collapse
|
31
|
Lee H, Lee YS, Harenda Q, Pietrzak S, Oktay HZ, Schreiber S, Liao Y, Sonthalia S, Ciecko AE, Chen YG, Keles S, Sridharan R, Engin F. Beta Cell Dedifferentiation Induced by IRE1α Deletion Prevents Type 1 Diabetes. Cell Metab 2020; 31:822-836.e5. [PMID: 32220307 PMCID: PMC7346095 DOI: 10.1016/j.cmet.2020.03.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 01/12/2020] [Accepted: 02/29/2020] [Indexed: 01/15/2023]
Abstract
Immune-mediated destruction of insulin-producing β cells causes type 1 diabetes (T1D). However, how β cells participate in their own destruction during the disease process is poorly understood. Here, we report that modulating the unfolded protein response (UPR) in β cells of non-obese diabetic (NOD) mice by deleting the UPR sensor IRE1α prior to insulitis induced a transient dedifferentiation of β cells, resulting in substantially reduced islet immune cell infiltration and β cell apoptosis. Single-cell and whole-islet transcriptomics analyses of immature β cells revealed remarkably diminished expression of β cell autoantigens and MHC class I components, and upregulation of immune inhibitory markers. IRE1α-deficient mice exhibited significantly fewer cytotoxic CD8+ T cells in their pancreata, and adoptive transfer of their total T cells did not induce diabetes in Rag1-/- mice. Our results indicate that inducing β cell dedifferentiation, prior to insulitis, allows these cells to escape immune-mediated destruction and may be used as a novel preventive strategy for T1D in high-risk individuals.
Collapse
Affiliation(s)
- Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Yong-Syu Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Quincy Harenda
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Stefan Pietrzak
- Department of Cell and Regenerative Biology, Wisconsin Institute for Discovery, Madison, WI 53706, USA
| | - Hülya Zeynep Oktay
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Sierra Schreiber
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Yian Liao
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Shreyash Sonthalia
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Ashley E Ciecko
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yi-Guang Chen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sunduz Keles
- Department of Biostatistics & Medical Informatics and Department of Statistics, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53705, USA
| | - Rupa Sridharan
- Department of Cell and Regenerative Biology, Wisconsin Institute for Discovery, Madison, WI 53706, USA
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
32
|
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
|
33
|
EMT and Stemness-Key Players in Pancreatic Cancer Stem Cells. Cancers (Basel) 2019; 11:cancers11081136. [PMID: 31398893 PMCID: PMC6721598 DOI: 10.3390/cancers11081136] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/01/2019] [Accepted: 08/06/2019] [Indexed: 12/15/2022] Open
Abstract
Metastasis and tumor progression are the major cause of death in patients suffering from pancreatic ductal adenocarcinoma. Tumor growth and especially dissemination are typically associated with activation of an epithelial-to-mesenchymal transition (EMT) program. This phenotypic transition from an epithelial to a mesenchymal state promotes migration and survival both during development and in cancer progression. When re-activated in pathological contexts such as cancer, this type of developmental process confers additional stemness properties to specific subsets of cells. Cancer stem cells (CSCs) are a subpopulation of cancer cells with stem-like features that are responsible for the propagation of the tumor as well as therapy resistance and cancer relapse, but also for circulating tumor cell release and metastasis. In support of this concept, EMT transcription factors generate cells with stem cell properties and mediate chemoresistance. However, their role in pancreatic ductal adenocarcinoma metastasis remains controversial. As such, a better characterization of CSC populations will be crucial in future development of therapies targeting these cells. In this review, we will discuss the latest updates on the mechanisms common to pancreas development and CSC-mediated tumor progression.
Collapse
|
34
|
Enhanced PDGF signaling in gestational diabetes mellitus is involved in pancreatic β-cell dysfunction. Biochem Biophys Res Commun 2019; 516:402-407. [PMID: 31217075 DOI: 10.1016/j.bbrc.2019.06.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
Abstract
Gestational diabetes mellitus (GDM) is often accompanied by the development of hyperinsulinemia as an adaptation to increased insulin demand, but this subsequently causes insulin resistance. Loss of function in pancreatic β-cells further aggravates the development of GDM. The level of serum platelet-derived growth factor (PDGF) reportedly increases in GDM patients. The present study investigated whether enhanced PDGF signaling directly causes β-cell dysfunction during gestation. Serum PDGF levels were negatively correlated with β-cell function in GDM patients. Administration of PDGF-BB disrupted glucose tolerance and β-cell function without inducing apoptosis in gestational mice but had no similar effect in non-gestational mice. The β-cell-specific genes encoding insulin synthesis proteins were decreased in the islets of PDGF-BB-treated gestational mice. In vitro experiments using INS1 insulinoma cells showed that PDGF-BB promoted cell proliferation, whereas it downregulated β-cell-specific genes. Taken together, these findings suggested that PDGF reduces β-cell function during gestation possibly through β-cell dedifferentiation.
Collapse
|
35
|
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
|
36
|
Wu MR, Hsiao JK, Liu HM, Huang YY, Tseng YJ, Chou PT, Weng TI, Yang CY. In vivo imaging of insulin-secreting human pancreatic ductal cells using MRI reporter gene technique: A feasibility study. Magn Reson Med 2019; 82:763-774. [PMID: 30957300 DOI: 10.1002/mrm.27749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE The purpose of this study was to investigate the feasibility of in vivo imaging of human pancreatic ductal cells by OATP1B3 reporter gene under MRI. METHODS A human cell line (PANC-1) derived from the pancreatic ductal epithelium was used in this study. After transduction of OATP1B3, the cellular physiological functions and the ability of intracellular uptake of the MRI contrast medium (Gd-EOB-DTPA) were examined. Induced differentiation of the PANC-1 cells into hormone-secreting cells were performed to simulate pancreatic β-like cells. The hormone-secreting cells were implanted into rats and in vivo MRI was evaluated. RESULTS The mRNA and proteins of OATP1B3 were highly expressed. No significant change of cellular physiological functions was found after the expression. After induced differentiation, the hormone secretion capacities of the OATP1B3-expressing PANC-1 cells were confirmed. Intra-cellular uptake of Gd-EOB-DTPA was determined in vitro by inductively coupled plasma mass spectrometry and MRI. In vivo MRI of the OATP1B3-expressing xenograft revealed an increased signal intensity after contrast enhancement. CONCLUSION OATP1B3 can be used as a safe and feasible in vivo MRI gene reporter for human pancreatic ductal cells.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hon-Man Liu
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan.,Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Radiology and Medical Imaging, Fu-Jen Catholic University and Hospital, New Taipei City, Taiwan
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Jui Tseng
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Pi-Tai Chou
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Te-I Weng
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Yi Yang
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medical Imaging, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan.,School of Medicine for International Students, I-Shou University, Kaohsiung City, Taiwan
| |
Collapse
|
37
|
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: 96] [Impact Index Per Article: 16.0] [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
|
38
|
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
|
39
|
Williams MD, Joglekar MV, Satoor SN, Wong W, Keramidaris E, Rixon A, O'Connell P, Hawthorne WJ, Mitchell GM, Hardikar AA. Epigenetic and Transcriptome Profiling Identifies a Population of Visceral Adipose-Derived Progenitor Cells with the Potential to Differentiate into an Endocrine Pancreatic Lineage. Cell Transplant 2018; 28:89-104. [PMID: 30376726 PMCID: PMC6322142 DOI: 10.1177/0963689718808472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by the loss of insulin-producing β-cells in the pancreas. T1D can be treated using cadaveric islet transplantation, but this therapy is severely limited by a lack of pancreas donors. To develop an alternative cell source for transplantation therapy, we carried out the epigenetic characterization in nine different adult mouse tissues and identified visceral adipose-derived progenitors as a candidate cell population. Chromatin conformation, assessed using chromatin immunoprecipitation (ChIP) sequencing and validated by ChIP-polymerase chain reaction (PCR) at key endocrine pancreatic gene promoters, revealed similarities between visceral fat and endocrine pancreas. Multiple techniques involving quantitative PCR, in-situ PCR, confocal microscopy, and flow cytometry confirmed the presence of measurable (2-1000-fold over detectable limits) pancreatic gene transcripts and mesenchymal progenitor cell markers (CD73, CD90 and CD105; >98%) in visceral adipose tissue-derived mesenchymal cells (AMCs). The differentiation potential of AMCs was explored in transgenic reporter mice expressing green fluorescent protein (GFP) under the regulation of the Pdx1 (pancreatic and duodenal homeobox-1) gene promoter. GFP expression was measured as an index of Pdx1 promoter activity to optimize culture conditions for endocrine pancreatic differentiation. Differentiated AMCs demonstrated their capacity to induce pancreatic endocrine genes as evidenced by increased GFP expression and validated using TaqMan real-time PCR (at least 2-200-fold relative to undifferentiated AMCs). Human AMCs differentiated using optimized protocols continued to produce insulin following transplantation in NOD/SCID mice. Our studies provide a systematic analysis of potential islet progenitor populations using genome-wide profiling studies and characterize visceral adipose-derived cells for replacement therapy in diabetes.
Collapse
Affiliation(s)
- Michael D Williams
- 1 NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia.,2 Department of Surgery, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.,3 O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Mugdha V Joglekar
- 1 NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Sarang N Satoor
- 1 NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Wilson Wong
- 1 NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Effie Keramidaris
- 3 O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Amanda Rixon
- 3 O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,4 Experimental Medical and Surgical Unit (EMSU), St Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Philip O'Connell
- 5 The Center for Transplant and Renal Research, Westmead Institute of Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Wayne J Hawthorne
- 5 The Center for Transplant and Renal Research, Westmead Institute of Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Geraldine M Mitchell
- 2 Department of Surgery, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.,3 O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,6 Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| | - Anandwardhan A Hardikar
- 1 NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
40
|
Cluster-assembled zirconia substrates promote long-term differentiation and functioning of human islets of Langerhans. Sci Rep 2018; 8:9979. [PMID: 29967323 PMCID: PMC6028636 DOI: 10.1038/s41598-018-28019-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/07/2018] [Indexed: 12/19/2022] Open
Abstract
Ex vivo expansion and differentiation of human pancreatic β-cell are enabling steps of paramount importance for accelerating the development of therapies for diabetes. The success of regenerative strategies depends on their ability to reproduce the chemical and biophysical properties of the microenvironment in which β-cells develop, proliferate and function. In this paper we focus on the biophysical properties of the extracellular environment and exploit the cluster-assembled zirconia substrates with tailored roughness to mimic the nanotopography of the extracellular matrix. We demonstrate that β-cells can perceive nanoscale features of the substrate and can convert these stimuli into mechanotransductive processes which promote long-term in vitro human islet culture, thus preserving β-cell differentiation and function. Proteomic and quantitative immunofluorescence analyses demonstrate that the process is driven by nanoscale topography, via remodelling of the actin cytoskeleton and nuclear architecture. These modifications activate a transcriptional program which stimulates an adaptive metabolic glucose response. Engineered cluster-assembled substrates coupled with proteomic approaches may provide a useful strategy for identifying novel molecular targets for treating diabetes mellitus and for enhancing tissue engineering in order to improve the efficacy of islet cell transplantation therapies.
Collapse
|
41
|
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
|
42
|
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
|
43
|
Characterization and Differentiation of Sorted Human Fetal Pancreatic ALDHhi and ALDHhi/CD133+ Cells Toward Insulin-Expressing Cells. Stem Cells Dev 2018; 27:275-286. [DOI: 10.1089/scd.2017.0135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
44
|
Abstract
INTRODUCTION The etiology of diabetes is mainly attributed to insulin deficiency due to the lack of β cells (type 1), or to insulin resistance that eventually results in β cell dysfunction (type 2). Therefore, an ultimate cure for diabetes requires the ability to replace the lost insulin-secreting β cells. Strategies for regenerating β cells are under extensive investigation. AREAS COVERED Herein, the authors first summarize the mechanisms underlying embryonic β cell development and spontaneous adult β cell regeneration, which forms the basis for developing β cell regeneration strategies. Then the rationale and progress of each β cell regeneration strategy is reviewed. Current β cell regeneration strategies can be classified into two main categories: in vitro β cell regeneration using pluripotent stem cells and in vivo reprogramming of non-β cells into β cells. Each has its own advantages and disadvantages. EXPERT OPINION Regenerating β cells has shown its potential as a cure for the treatment of insulin-deficient diabetes. Much progress has been made, and β cell regeneration therapy is getting closer to a clinical reality. Nevertheless, more hurdles need to be overcome before any of the strategies suggested can be fully translated from bench to bedside.
Collapse
Affiliation(s)
- Shengli Dong
- Department of Biochemistry & Molecular Biology, Louisiana State University Health Science Center, New Orleans, Louisiana
| | - Hongju Wu
- Department of Medicine, Tulane University Health Science Center, New Orleans, Louisiana
| |
Collapse
|
45
|
Krivova YS, Proshchina AE, Barabanov VM, Barinova IV, Saveliev SV. Immunohistochemical detection of vimentin in pancreatic islet β- and α-cells of macrosomic infants of diabetic and nondiabetic mothers. Early Hum Dev 2018; 117:44-49. [PMID: 29275072 DOI: 10.1016/j.earlhumdev.2017.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/08/2017] [Accepted: 12/10/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND Expression of the intermediate filament protein vimentin has been recently observed in the pancreatic islet β- and α-cells of humans with type 2 diabetes mellitus. It was suggested that the presence of vimentin in endocrine cells may indicate islet tissue renewal, or potentially represent the dedifferentiation of endocrine cells, which could contribute to the onset of type 2 diabetes or islet cell dysfunction. AIM To analyze the expression of vimentin in pancreatic β- and α-cells of macrosomic infants of diabetic and nondiabetic mothers. SUBJECTS Pancreatic samples of five macrosomic infants (gestational age 34-40weeks) from three diabetic and two nondiabetic mothers were compared to six control infants (32-40weeks, weight appropriate for gestational age) from normoglycemic mothers. METHODS Pancreatic autopsy samples were examined by double immunofluorescent labeling with antibodies against vimentin and either insulin or glucagon. Alterations in the endocrine pancreas were measured using morphometric methods, then data were statistically analyzed. RESULTS In the pancreatic islets of macrosomic infants from diabetic and nondiabetic mothers, we observed vimentin-positive cells, some of which simultaneously contained insulin or glucagon. We also quantitatively showed that the presence of such cells was associated with hypertrophy and hyperplasia of the islets, and with an increase in β- and α-cell density. CONCLUSIONS We speculate that the appearance of vimentin-positive islet cells may reflect induction of differentiation in response to the increased insulin demand, and vimentin may serve as an early marker of endocrine pancreas disorders.
Collapse
Affiliation(s)
- Yuliya S Krivova
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia.
| | - Alexandra E Proshchina
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia
| | - Valeriy M Barabanov
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia
| | - Irina V Barinova
- Moscow Regional Research Institute of Obstetrics and Gynecology, Ministry of Health of the Moscow Region, 22a Pokrovka St., Moscow 101000, Russia
| | - Sergey V Saveliev
- Laboratory of Nervous System Development, Research Institute of Human Morphology, 3 Tsurupy St., Moscow 117418, Russia
| |
Collapse
|
46
|
Barui A, Chowdhury F, Pandit A, Datta P. Rerouting mesenchymal stem cell trajectory towards epithelial lineage by engineering cellular niche. Biomaterials 2018; 156:28-44. [DOI: 10.1016/j.biomaterials.2017.11.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/22/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
|
47
|
Moreno-Amador JL, Téllez N, Marin S, Aloy-Reverté C, Semino C, Nacher M, Montanya E. Epithelial to mesenchymal transition in human endocrine islet cells. PLoS One 2018; 13:e0191104. [PMID: 29360826 PMCID: PMC5779658 DOI: 10.1371/journal.pone.0191104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022] Open
Abstract
Background β-cells undergo an epithelial to mesenchymal transition (EMT) when expanded in monolayer culture and give rise to highly proliferative mesenchymal cells that retain the potential to re-differentiate into insulin-producing cells. Objective To investigate whether EMT takes place in the endocrine non-β cells of human islets. Methodology Human islets isolated from 12 multiorgan donors were dissociated into single cells, purified by magnetic cell sorting, and cultured in monolayer. Results Co-expression of insulin and the mesenchymal marker vimentin was identified within the first passage (p1) and increased subsequently (insulin+vimentin+ 7.2±6% at p1; 43±15% at p4). The endocrine non-β-cells did also co-express vimentin (glucagon+vimentin+ 59±1.5% and 93±6%, somatostatin+vimentin+ 16±9.4% and 90±10% at p1 and p4 respectively; PP+vimentin+ 74±14% at p1; 88±12% at p2). The percentage of cells expressing only endocrine markers was progressively reduced (0.6±0.2% insulin+, 0.2±0.1% glucagon+, and 0.3±0.2% somatostatin+ cells at p4, and 0.7±0.3% PP+ cells at p2. Changes in gene expression were also indicated of EMT, with reduced expression of endocrine markers and the epithelial marker CDH-1 (p<0.01), and increased expression of mesenchymal markers (CDH-2, SNAI2, ZEB1, ZEB2, VIM, NT5E and ACTA2; p<0.05). Treatment with the EMT inhibitor A83-01 significantly reduced the percentage of co-expressing cells and preserved the expression of endocrine markers. Conclusions In adult human islets, all four endocrine islet cell types undergo EMT when islet cells are expanded in monolayer conditions. The presence of EMT in all islet endocrine cells could be relevant to design of strategies aiming to re-differentiate the expanded islet cells towards a β-cell phenotype.
Collapse
Affiliation(s)
- José Luis Moreno-Amador
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Noèlia Téllez
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Sandra Marin
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | | | - Carlos Semino
- IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - Montserrat Nacher
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Hospital Universitari Bellvitge, Hospitalet de Llobregat, Spain
- * E-mail: (MN); (ED)
| | - Eduard Montanya
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Hospital Universitari Bellvitge, Hospitalet de Llobregat, Spain
- University of Barcelona, Barcelona, Spain
- * E-mail: (MN); (ED)
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Inadequate insulin-producing pancreatic β-cell mass is a key feature of both type 1 and type 2 diabetes. Efforts to regenerate β-cell mass from pancreatic precursors may thus ameliorate absolute or relative insulin deficiency, thereby improving glucose homeostasis. A clear understanding of the processes that govern the generation of new β-cells in the mature pancreas will be fundamental to success in this effort. This review discusses the current state of knowledge regarding β-cell regeneration and emphasizes recent studies of significance. RECENT FINDINGS Recent reports demonstrate regenerative potential in the adult human pancreas. Further, they build on the strong existing evidence that proliferation of preexisting β-cells is the predominant source of new β-cells in adulthood by dissecting the cell cycle machinery components and critical signaling pathways required for β-cell proliferation. Finally, β-cell trophic peptides have demonstrated preclinical potential as pharmacologic regenerative agents and may form the basis for clinical interventions in the future. SUMMARY Efforts to augment β-cell regeneration by enhancing β-cell viability and proliferation may lead to novel therapeutic approaches for type 1 and type 2 diabetes. An intimate understanding of the molecular mechanisms underlying the regulation of β-cell proliferation and survival will be fundamental to the optimization of endogenous β-cell regeneration.
Collapse
|
49
|
Apostolou G, Apostolou N, Moulos P, Chatzipantelis P. Endometrial cytopathology. An image analysis approach using the Ki-67 biomarker. Cytopathology 2017; 28:385-390. [PMID: 28737230 DOI: 10.1111/cyt.12435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2017] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To investigate the different identity and biological behaviour of endometrial benign epithelial and endometrial adenocarcinoma cell categories. METHODS For this study, the imprint smears from three groups, 10 cases of disordered proliferative/benign hyperplastic endometrium, 21 cases of low-grade and eight cases of high-grade endometrial adenocarcinoma, were examined using image analysis and the Ki-67 biomarker. The plastic stem cell model was also applied. RESULTS Among the examined groups, the nuclear area major axis ranged statistically different in the digitally measured Ki-67 positive endometrial epithelial and adenocarcinoma cells (P<.0001). Moreover, higher values of the cycling nuclear area major axis were observed in high-grade, as compared with the low-grade endometrial adenocarcinomas (P<.0001) and the cases of disordered/benign hyperplastic endometrium (P<.0001). Additionally, a Ki-67 increase pathway was observed in the benign endometrial lesions, and a relatively stable pathway was noticed in low- and high-grade endometrial adenocarcinomas. CONCLUSIONS The different range of the nuclear area major axis among cycling endometrial epithelial and adenocarcinoma cells may correlate with their specific identity and biological behaviour. The different values of the cycling nuclear area major dimension may also be connected with the biological behaviour of the three examined groups. Moreover, the endometrial epithelial cells may follow a Ki-67 increase pathway, instead of the relatively stable pathway which the rapidly proliferating adenocarcinoma cells may use. Finally, the studied cell categories may exhibit different biology, because their stem cells may reside in different states of stemness.
Collapse
Affiliation(s)
- G Apostolou
- Department of Cytopathology, Anti-cancer Oncological Hospital St. Savvas, Athens, Greece
| | - N Apostolou
- Department of Biomedical Engineering, Olympic Polyclinic, Athens, Greece
| | - P Moulos
- HybridStat Predictive Analytics, Athens, Greece
| | - P Chatzipantelis
- Department of Cellular Pathology, Royal Devon and Exeter Hospital, Exeter, UK
| |
Collapse
|
50
|
Atouf F, Choi Y, Fowler MJ, Poffenberger G, Vobecky J, Ta M, Chapman GB, Powers AC, Lumelsky NL. Generation of Islet-Like Hormone-Producing Cells In Vitro from Adult Human Pancreas. Cell Transplant 2017; 14:735-48. [PMID: 16454348 DOI: 10.3727/000000005783982602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transplantation of pancreatic islets can provide long-lasting insulin independence for diabetic patients, but the current islet supply is limited. Here we describe a new in vitro system that utilizes adult human pancreatic islet-enriched fractions to generate hormone-producing cells over 3–4 weeks of culture. By labeling proliferating cells with a retrovirus-expressing green fluorescent protein, we show that in this system hormone-producing cells are generated de novo. These hormone-producing cells aggregate to form islet-like cell clusters. The cell clusters, when tested in vitro, release insulin in response to glucose and other secretagogues. After transplantation into immunodeficient, nondiabetic mice, the islet-like cell clusters survive and release human insulin. We propose that this system will be useful as an experimental tool for investigating mechanisms for generating new islet cells from the postnatal pancreas, and for designing strategies to generate physiologically competent pancreatic islet cells ex vivo.
Collapse
Affiliation(s)
- Fouad Atouf
- Islet and Autoimmunity Branch, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1453, USA
| | | | | | | | | | | | | | | | | |
Collapse
|