1
|
Xu Y, Mao S, Fan H, Wan J, Wang L, Zhang M, Zhu S, Yuan J, Lu Y, Wang Z, Yu B, Jiang Z, Huang Y. LINC MIR503HG Controls SC-β Cell Differentiation and Insulin Production by Targeting CDH1 and HES1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305631. [PMID: 38243869 PMCID: PMC10987150 DOI: 10.1002/advs.202305631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/03/2024] [Indexed: 01/22/2024]
Abstract
Stem cell-derived pancreatic progenitors (SC-PPs), as an unlimited source of SC-derived β (SC-β) cells, offers a robust tool for diabetes treatment in stem cell-based transplantation, disease modeling, and drug screening. Whereas, PDX1+/NKX6.1+ PPs enhances the subsequent endocrine lineage specification and gives rise to glucose-responsive SC-β cells in vivo and in vitro. To identify the regulators that promote induction efficiency and cellular function maturation, single-cell RNA-sequencing is performed to decipher the transcriptional landscape during PPs differentiation. The comprehensive evaluation of functionality demonstrated that manipulating LINC MIR503HG using CRISPR in PP cell fate decision can improve insulin synthesis and secretion in mature SC-β cells, without effects on liver lineage specification. Importantly, transplantation of MIR503HG-/- SC-β cells in recipients significantly restored blood glucose homeostasis, accompanied by serum C-peptide release and an increase in body weight. Mechanistically, by releasing CtBP1 occupying the CDH1 and HES1 promoters, the decrease in MIR503HG expression levels provided an excellent extracellular niche and appropriate Notch signaling activation for PPs following differentiation. Furthermore, this exhibited higher crucial transcription factors and mature epithelial markers in CDH1High expressed clusters. Altogether, these findings highlighted MIR503HG as an essential and exclusive PP cell fate specification regulator with promising therapeutic potential for patients with diabetes.
Collapse
Affiliation(s)
- Yang Xu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Center of Gallbladder DiseaseShanghai East HospitalInstitute of Gallstone DiseaseSchool of MedicineTongji UniversityShanghai200092China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Haowen Fan
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Jian Wan
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Lin Wang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Department of Graduate SchoolDalian Medical UniversityDalianLiaoning116000China
| | - Mingyu Zhang
- Department of Nuclear MedicineBeijing Friendship HospitalAffiliated to Capital Medical UniversityBeijing100050China
| | - Shajun Zhu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Jin Yuan
- Department of Endocrinology and MetabolismAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Zhaoyan Jiang
- Center of Gallbladder DiseaseShanghai East HospitalInstitute of Gallstone DiseaseSchool of MedicineTongji UniversityShanghai200092China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityMedical School of Nantong UniversityNantong226001China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology ProductsCo‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| |
Collapse
|
2
|
Grygoryev D, Ekstrom T, Manalo E, Link JM, Alshaikh A, Keith D, Allen-Petersen BL, Sheppard B, Morgan T, Soufi A, Sears RC, Kim J. Sendai virus is robust and consistent in delivering genes into human pancreatic cancer cells. Heliyon 2024; 10:e27221. [PMID: 38463758 PMCID: PMC10923719 DOI: 10.1016/j.heliyon.2024.e27221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly intratumorally heterogeneous disease that includes several subtypes and is highly plastic. Effective gene delivery to all PDAC cells is essential for modulating gene expression and identifying potential gene-based therapeutic targets in PDAC. Most current gene delivery systems for pancreatic cells are optimized for islet or acinar cells. Lentiviral vectors are the current main gene delivery vectors for PDAC, but their transduction efficiencies vary depending on pancreatic cell type, and are especially poor for the classical subtype of PDAC cells from both primary tumors and cell lines. Methods We systemically compare transduction efficiencies of glycoprotein G of vesicular stomatitis virus (VSV-G)-pseudotyped lentiviral and Sendai viral vectors in human normal pancreatic ductal and PDAC cells. Results We find that the Sendai viral vector gives the most robust gene delivery efficiency regardless of PDAC cell type. Therefore, we propose using Sendai viral vectors to transduce ectopic genes into PDAC cells.
Collapse
Affiliation(s)
- Dmytro Grygoryev
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Taelor Ekstrom
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Elise Manalo
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
| | - Jason M. Link
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Amani Alshaikh
- The University of Edinburgh, Centre for Regenerative Medicine, Institute of Regeneration and Repair, Institute of Stem Cell Research, Edinburgh, UK
- King Abdulaziz City for Science and Technology, Health Sector (KACST), Riyadh, Saudi Arabia
| | - Dove Keith
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Brittany L. Allen-Petersen
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
| | - Brett Sheppard
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
- Department of Surgery, Oregon Health & Science University School of Medicine, USA
| | - Terry Morgan
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
- Department of Pathology, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| | - Abdenour Soufi
- The University of Edinburgh, Centre for Regenerative Medicine, Institute of Regeneration and Repair, Institute of Stem Cell Research, Edinburgh, UK
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| | - Jungsun Kim
- Cancer Early Detection Advanced Research Center at Knight Cancer Institute, Oregon Health & Science University School of Medicine, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University School of Medicine, USA
- Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR, 97201, USA
| |
Collapse
|
3
|
Blanchi B, Taurand M, Colace C, Thomaidou S, Audeoud C, Fantuzzi F, Sawatani T, Gheibi S, Sabadell-Basallote J, Boot FWJ, Chantier T, Piet A, Cavanihac C, Pilette M, Balguerie A, Olleik H, Carlotti F, Ejarque M, Fex M, Mulder H, Cnop M, Eizirik DL, Jouannot O, Gaffuri AL, Czernichow P, Zaldumbide A, Scharfmann R, Ravassard P. EndoC-βH5 cells are storable and ready-to-use human pancreatic beta cells with physiological insulin secretion. Mol Metab 2023; 76:101772. [PMID: 37442376 PMCID: PMC10407753 DOI: 10.1016/j.molmet.2023.101772] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
OBJECTIVES Readily accessible human pancreatic beta cells that are functionally close to primary adult beta cells are a crucial model to better understand human beta cell physiology and develop new treatments for diabetes. We here report the characterization of EndoC-βH5 cells, the latest in the EndoC-βH cell family. METHODS EndoC-βH5 cells were generated by integrative gene transfer of immortalizing transgenes hTERT and SV40 large T along with Herpes Simplex Virus-1 thymidine kinase into human fetal pancreas. Immortalizing transgenes were removed after amplification using CRE activation and remaining non-excized cells eliminated using ganciclovir. Resulting cells were distributed as ready to use EndoC-βH5 cells. We performed transcriptome, immunological and extensive functional assays. RESULTS Ready to use EndoC-βH5 cells display highly efficient glucose dependent insulin secretion. A robust 10-fold insulin secretion index was observed and reproduced in four independent laboratories across Europe. EndoC-βH5 cells secrete insulin in a dynamic manner in response to glucose and secretion is further potentiated by GIP and GLP-1 analogs. RNA-seq confirmed abundant expression of beta cell transcription factors and functional markers, including incretin receptors. Cytokines induce a gene expression signature of inflammatory pathways and antigen processing and presentation. Finally, modified HLA-A2 expressing EndoC-βH5 cells elicit specific A2-alloreactive CD8 T cell activation. CONCLUSIONS EndoC-βH5 cells represent a unique storable and ready to use human pancreatic beta cell model with highly robust and reproducible features. Such cells are thus relevant for the study of beta cell function, screening and validation of new drugs, and development of disease models.
Collapse
Affiliation(s)
| | | | - Claire Colace
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Sofia Thomaidou
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Federica Fantuzzi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium; Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Toshiaki Sawatani
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Sevda Gheibi
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmö, Sweden
| | - Joan Sabadell-Basallote
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain; Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Fransje W J Boot
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | | | - Aline Piet
- Human Cell Design, Canceropole, Toulouse, France
| | | | | | | | - Hamza Olleik
- Human Cell Design, Canceropole, Toulouse, France
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Miriam Ejarque
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Tarragona, Spain
| | - Malin Fex
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmö, Sweden
| | - Hindrik Mulder
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmö, Sweden
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium; Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Raphaël Scharfmann
- Université Paris Cité, Institut Cochin, CNRS, INSERM U1016, Paris, 75014, France
| | - Philippe Ravassard
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Paris, France.
| |
Collapse
|
4
|
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
|
5
|
Cell Replacement Therapy for Type 1 Diabetes Patients: Potential Mechanisms Leading to Stem-Cell-Derived Pancreatic β-Cell Loss upon Transplant. Cells 2023; 12:cells12050698. [PMID: 36899834 PMCID: PMC10000642 DOI: 10.3390/cells12050698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cell replacement therapy using stem-cell-derived insulin-producing β-like cells (sBCs) has been proposed as a practical cure for patients with type one diabetes (T1D). sBCs can correct diabetes in preclinical animal models, demonstrating the promise of this stem cell-based approach. However, in vivo studies have demonstrated that most sBCs, similarly to cadaveric human islets, are lost upon transplantation due to ischemia and other unknown mechanisms. Hence, there is a critical knowledge gap in the current field concerning the fate of sBCs upon engraftment. Here we review, discuss effects, and propose additional potential mechanisms that could contribute toward β-cell loss in vivo. We summarize and highlight some of the literature on phenotypic loss in β-cells under both steady, stressed, and diseased diabetic conditions. Specifically, we focus on β-cell death, dedifferentiation into progenitors, trans-differentiation into other hormone-expressing cells, and/or interconversion into less functional β-cell subtypes as potential mechanisms. While current cell replacement therapy efforts employing sBCs carry great promise as an abundant cell source, addressing the somewhat neglected aspect of β-cell loss in vivo will further accelerate sBC transplantation as a promising therapeutic modality that could significantly enhance the life quality of T1D patients.
Collapse
|
6
|
Wei T, Wei R, Hong T. Regeneration of β cells from cell phenotype conversion among the pancreatic endocrine cells. Chronic Dis Transl Med 2022; 8:1-4. [PMID: 35620156 PMCID: PMC9128562 DOI: 10.1002/cdt3.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/12/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Tianjiao Wei
- Department of Endocrinology and Metabolism Peking University Third Hospital Beijing 100191 China
| | - Rui Wei
- Department of Endocrinology and Metabolism Peking University Third Hospital Beijing 100191 China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism Peking University Third Hospital Beijing 100191 China
| |
Collapse
|
7
|
Namestnikov M, Dekel B. Moving To A New Dimension: 3D Kidney Cultures For Kidney Regeneration. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
8
|
Groen N, Leenders F, Mahfouz A, Munoz-Garcia A, Muraro MJ, de Graaf N, Rabelink TJ, Hoeben R, van Oudenaarden A, Zaldumbide A, Reinders MJT, de Koning EJP, Carlotti F. Single-Cell Transcriptomics Links Loss of Human Pancreatic β-Cell Identity to ER Stress. Cells 2021; 10:3585. [PMID: 34944092 PMCID: PMC8700697 DOI: 10.3390/cells10123585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/25/2021] [Accepted: 12/10/2021] [Indexed: 11/30/2022] Open
Abstract
The maintenance of pancreatic islet architecture is crucial for proper β-cell function. We previously reported that disruption of human islet integrity could result in altered β-cell identity. Here we combine β-cell lineage tracing and single-cell transcriptomics to investigate the mechanisms underlying this process in primary human islet cells. Using drug-induced ER stress and cytoskeleton modification models, we demonstrate that altering the islet structure triggers an unfolding protein response that causes the downregulation of β-cell maturity genes. Collectively, our findings illustrate the close relationship between endoplasmic reticulum homeostasis and β-cell phenotype, and strengthen the concept of altered β-cell identity as a mechanism underlying the loss of functional β-cell mass.
Collapse
Affiliation(s)
- Nathalie Groen
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (N.G.); (F.L.); (A.M.-G.); (N.d.G.); (T.J.R.); (E.J.P.d.K.)
| | - Floris Leenders
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (N.G.); (F.L.); (A.M.-G.); (N.d.G.); (T.J.R.); (E.J.P.d.K.)
| | - Ahmed Mahfouz
- Leiden Computational Biology Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.M.); (M.J.T.R.)
- Delft Bioinformatics Lab, Delft University of Technology, 2628 XE Delft, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Amadeo Munoz-Garcia
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (N.G.); (F.L.); (A.M.-G.); (N.d.G.); (T.J.R.); (E.J.P.d.K.)
| | - Mauro J. Muraro
- Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), 3584 CT Utrecht, The Netherlands; (M.J.M.); (A.v.O.)
| | - Natascha de Graaf
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (N.G.); (F.L.); (A.M.-G.); (N.d.G.); (T.J.R.); (E.J.P.d.K.)
| | - Ton. J. Rabelink
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (N.G.); (F.L.); (A.M.-G.); (N.d.G.); (T.J.R.); (E.J.P.d.K.)
| | - Rob Hoeben
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.H.); (A.Z.)
| | - Alexander van Oudenaarden
- Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), 3584 CT Utrecht, The Netherlands; (M.J.M.); (A.v.O.)
- Molecular Cancer Research, University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.H.); (A.Z.)
| | - Marcel J. T. Reinders
- Leiden Computational Biology Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.M.); (M.J.T.R.)
- Delft Bioinformatics Lab, Delft University of Technology, 2628 XE Delft, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Eelco J. P. de Koning
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (N.G.); (F.L.); (A.M.-G.); (N.d.G.); (T.J.R.); (E.J.P.d.K.)
- Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), 3584 CT Utrecht, The Netherlands; (M.J.M.); (A.v.O.)
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (N.G.); (F.L.); (A.M.-G.); (N.d.G.); (T.J.R.); (E.J.P.d.K.)
| |
Collapse
|
9
|
Title AC, Silva PN, Godbersen S, Hasenöhrl L, Stoffel M. The miR-200-Zeb1 axis regulates key aspects of β-cell function and survival in vivo. Mol Metab 2021; 53:101267. [PMID: 34116231 PMCID: PMC8258987 DOI: 10.1016/j.molmet.2021.101267] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The miR-200-Zeb1 axis regulates the epithelial-to-mesenchymal transition (EMT), differentiation, and resistance to apoptosis. A better understanding of these processes in diabetes is highly relevant, as β-cell dedifferentiation and apoptosis contribute to the loss of functional β-cell mass and diabetes progression. Furthermore, EMT promotes the loss of β-cell identity in the in vitro expansion of human islets. Though the miR-200 family has previously been identified as a regulator of β-cell apoptosis in vivo, studies focusing on Zeb1 are lacking. The aim of this study was thus to investigate the role of Zeb1 in β-cell function and survival in vivo. METHODS miR-200 and Zeb1 are involved in a double-negative feedback loop. We characterized a mouse model in which miR-200 binding sites in the Zeb1 3'UTR are mutated (Zeb1200), leading to a physiologically relevant upregulation of Zeb1 mRNA expression. The role of Zeb1 was investigated in this model via metabolic tests and analysis of isolated islets. Further insights into the distinct contributions of the miR-200 and Zeb1 branches of the feedback loop were obtained by crossing the Zeb1200 allele into a background of miR-141-200c overexpression. RESULTS Mild Zeb1 derepression in vivo led to broad transcriptional changes in islets affecting β-cell identity, EMT, insulin secretion, cell-cell junctions, the unfolded protein response (UPR), and the response to ER stress. The aggregation and insulin secretion of dissociated islets of mice homozygous for the Zeb1200 mutation (Zeb1200M) were impaired, and Zeb1200M islets were resistant to thapsigargin-induced ER stress ex vivo. Zeb1200M mice had increased circulating proinsulin levels but no overt metabolic phenotype, reflecting the strong compensatory ability of islets to maintain glucose homeostasis. CONCLUSIONS This study signifies the importance of the miR-200-Zeb1 axis in regulating key aspects of β-cell function and survival. A better understanding of this axis is highly relevant in developing therapeutic strategies for inducing β-cell redifferentiation and maintaining β-cell identity in in vitro islet expansion.
Collapse
Affiliation(s)
- Alexandra C Title
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland; Competence Center Personalized Medicine, ETH Zürich, Voltastrasse 24, 8044, Zürich, Switzerland
| | - Pamuditha N Silva
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Svenja Godbersen
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Lynn Hasenöhrl
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Markus Stoffel
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland; Competence Center Personalized Medicine, ETH Zürich, Voltastrasse 24, 8044, Zürich, Switzerland; Medical Faculty, University of Zürich, 8091, Zürich, Switzerland.
| |
Collapse
|
10
|
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
|
11
|
A new shortened protocol to obtain islet-like cells from hESC-derived ductal cells. In Vitro Cell Dev Biol Anim 2021; 57:587-597. [PMID: 34212340 DOI: 10.1007/s11626-021-00580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
Conventional methods for obtaining pancreatic β cells are based on simulating the embryonic development phase of endocrine cells via hierarchical differentiation of pluripotent stem cells (PSCs). Accordingly, we attempted to modify the protocols for obtaining insulin-secreting cells (ISCs) by sequential differentiation of a human embryonic stem cell (hESC), using the HS181 cell line. Furthermore, we hypothesize that actual pancreatic endocrine cells may arise from trans-differentiation of mature ductal cells after the embryonic developmental stage and throughout the rest of life. According to the hypothesis, ductal cells are trans-differentiated into endocrine and exocrine cells, undergoing a partial epithelial to mesenchymal transition (EMT). To address this issue, we developed two new protocols based on hESC differentiation to obtain ductal cells and then induce EMT in cells to obtain hormone-secreting islet-like cells (HSCs). The ductal (pre-EMT exocrine) cells were then induced to undergo partial EMT by treating with Wnt3a and activin A, in hypoxia. The cell derived from the latter method significantly expressed the main endocrine cell-specific markers and also β cells, in particular. These experiments not only support our hypothetical model but also offer a promising approach to develop new methods to compensate β cell depletion in patients with type 1 diabetes mellitus (T1DM). Although this protocol of generating islet-like cells from ductal cells has a potential to treat T1DM, this strategy may be exploited to optimize the function of these cells in an animal model and future clinical applications.
Collapse
|
12
|
Namestnikov M, Pleniceanu O, Dekel B. Mixing Cells for Vascularized Kidney Regeneration. Cells 2021; 10:1119. [PMID: 34066487 PMCID: PMC8148539 DOI: 10.3390/cells10051119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [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: 04/26/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
The worldwide rise in prevalence of chronic kidney disease (CKD) demands innovative bio-medical solutions for millions of kidney patients. Kidney regenerative medicine aims to replenish tissue which is lost due to a common pathological pathway of fibrosis/inflammation and rejuvenate remaining tissue to maintain sufficient kidney function. To this end, cellular therapy strategies devised so far utilize kidney tissue-forming cells (KTFCs) from various cell sources, fetal, adult, and pluripotent stem-cells (PSCs). However, to increase engraftment and potency of the transplanted cells in a harsh hypoxic diseased environment, it is of importance to co-transplant KTFCs with vessel forming cells (VFCs). VFCs, consisting of endothelial cells (ECs) and mesenchymal stem-cells (MSCs), synergize to generate stable blood vessels, facilitating the vascularization of self-organizing KTFCs into renovascular units. In this paper, we review the different sources of KTFCs and VFCs which can be mixed, and report recent advances made in the field of kidney regeneration with emphasis on generation of vascularized kidney tissue by cell transplantation.
Collapse
Affiliation(s)
- Michael Namestnikov
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- ediatric Nephrology Division, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
| | - Oren Pleniceanu
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- The Kidney Research Lab, Institute of Nephrology and Hypertension, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
| |
Collapse
|
13
|
Grieco GE, Brusco N, Licata G, Fignani D, Formichi C, Nigi L, Sebastiani G, Dotta F. The Landscape of microRNAs in βCell: Between Phenotype Maintenance and Protection. Int J Mol Sci 2021; 22:ijms22020803. [PMID: 33466949 PMCID: PMC7830142 DOI: 10.3390/ijms22020803] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus is a group of heterogeneous metabolic disorders characterized by chronic hyperglycaemia mainly due to pancreatic β cell death and/or dysfunction, caused by several types of stress such as glucotoxicity, lipotoxicity and inflammation. Different patho-physiological mechanisms driving β cell response to these stresses are tightly regulated by microRNAs (miRNAs), a class of negative regulators of gene expression, involved in pathogenic mechanisms occurring in diabetes and in its complications. In this review, we aim to shed light on the most important miRNAs regulating the maintenance and the robustness of β cell identity, as well as on those miRNAs involved in the pathogenesis of the two main forms of diabetes mellitus, i.e., type 1 and type 2 diabetes. Additionally, we acknowledge that the understanding of miRNAs-regulated molecular mechanisms is fundamental in order to develop specific and effective strategies based on miRNAs as therapeutic targets, employing innovative molecules.
Collapse
Affiliation(s)
- Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Noemi Brusco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Giada Licata
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Daniela Fignani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Caterina Formichi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (G.E.G.); (N.B.); (G.L.); (D.F.); (C.F.); (L.N.); (G.S.)
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, 53100 Siena, Italy
- Tuscany Centre for Precision Medicine (CReMeP), 53100 Siena, Italy
- Correspondence: ; Tel.: +39-0577-231283
| |
Collapse
|
14
|
Shrestha N, De Franco E, Arvan P, Cnop M. Pathological β-Cell Endoplasmic Reticulum Stress in Type 2 Diabetes: Current Evidence. Front Endocrinol (Lausanne) 2021; 12:650158. [PMID: 33967960 PMCID: PMC8101261 DOI: 10.3389/fendo.2021.650158] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
The notion that in diabetes pancreatic β-cells express endoplasmic reticulum (ER) stress markers indicative of increased unfolded protein response (UPR) signaling is no longer in doubt. However, what remains controversial is whether this increase in ER stress response actually contributes importantly to the β-cell failure of type 2 diabetes (akin to 'terminal UPR'), or whether it represents a coping mechanism that represents the best attempt of β-cells to adapt to changes in metabolic demands as presented by disease progression. Here an intercontinental group of experts review evidence for the role of ER stress in monogenic and type 2 diabetes in an attempt to reconcile these disparate views. Current evidence implies that pancreatic β-cells require a regulated UPR for their development, function and survival, as well as to maintain cellular homeostasis in response to protein misfolding stress. Prolonged ER stress signaling, however, can be detrimental to β-cells, highlighting the importance of "optimal" UPR for ER homeostasis, β-cell function and survival.
Collapse
Affiliation(s)
- Neha Shrestha
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, Exeter, United Kingdom
| | - Peter Arvan
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- *Correspondence: Peter Arvan, ; Miriam Cnop,
| | - Miriam Cnop
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
- *Correspondence: Peter Arvan, ; Miriam Cnop,
| |
Collapse
|
15
|
Qadir MMF, Álvarez-Cubela S, Weitz J, Panzer JK, Klein D, Moreno-Hernández Y, Cechin S, Tamayo A, Almaça J, Hiller H, Beery M, Kusmartseva I, Atkinson M, Speier S, Ricordi C, Pugliese A, Caicedo A, Fraker CA, Pastori RL, Domínguez-Bendala J. Long-term culture of human pancreatic slices as a model to study real-time islet regeneration. Nat Commun 2020; 11:3265. [PMID: 32601271 PMCID: PMC7324563 DOI: 10.1038/s41467-020-17040-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 06/04/2020] [Indexed: 01/02/2023] Open
Abstract
The culture of live pancreatic tissue slices is a powerful tool for the interrogation of physiology and pathology in an in vitro setting that retains near-intact cytoarchitecture. However, current culture conditions for human pancreatic slices (HPSs) have only been tested for short-term applications, which are not permissive for the long-term, longitudinal study of pancreatic endocrine regeneration. Using a culture system designed to mimic the physiological oxygenation of the pancreas, we demonstrate high viability and preserved endocrine and exocrine function in HPS for at least 10 days after sectioning. This extended lifespan allowed us to dynamically lineage trace and quantify the formation of insulin-producing cells in HPS from both non-diabetic and type 2 diabetic donors. This technology is expected to be of great impact for the conduct of real-time regeneration/developmental studies in the human pancreas.
Collapse
Affiliation(s)
- Mirza Muhammad Fahd Qadir
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Jonathan Weitz
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Julia K Panzer
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Yaisa Moreno-Hernández
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Universidad Francisco de Vitoria, Madrid, Spain
| | - Sirlene Cechin
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Tamayo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Joana Almaça
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Helmut Hiller
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Beery
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Irina Kusmartseva
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Mark Atkinson
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Institute of Physiology, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alberto Pugliese
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Caicedo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Christopher A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
16
|
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: 1.3] [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
|
17
|
Georgakopoulos N, Prior N, Angres B, Mastrogiovanni G, Cagan A, Harrison D, Hindley CJ, Arnes-Benito R, Liau SS, Curd A, Ivory N, Simons BD, Martincorena I, Wurst H, Saeb-Parsy K, Huch M. Long-term expansion, genomic stability and in vivo safety of adult human pancreas organoids. BMC DEVELOPMENTAL BIOLOGY 2020; 20:4. [PMID: 32098630 PMCID: PMC7043048 DOI: 10.1186/s12861-020-0209-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/03/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Pancreatic organoid systems have recently been described for the in vitro culture of pancreatic ductal cells from mouse and human. Mouse pancreatic organoids exhibit unlimited expansion potential, while previously reported human pancreas organoid (hPO) cultures do not expand efficiently long-term in a chemically defined, serum-free medium. We sought to generate a 3D culture system for long-term expansion of human pancreas ductal cells as hPOs to serve as the basis for studies of human pancreas ductal epithelium, exocrine pancreatic diseases and the development of a genomically stable replacement cell therapy for diabetes mellitus. RESULTS Our chemically defined, serum-free, human pancreas organoid culture medium supports the generation and expansion of hPOs with high efficiency from both fresh and cryopreserved primary tissue. hPOs can be expanded from a single cell, enabling their genetic manipulation and generation of clonal cultures. hPOs expanded for months in vitro maintain their ductal morphology, biomarker expression and chromosomal integrity. Xenografts of hPOs survive long-term in vivo when transplanted into the pancreas of immunodeficient mice. Notably, mouse orthotopic transplants show no signs of tumorigenicity. Crucially, our medium also supports the establishment and expansion of hPOs in a chemically defined, modifiable and scalable, biomimetic hydrogel. CONCLUSIONS hPOs can be expanded long-term, from both fresh and cryopreserved human pancreas tissue in a chemically defined, serum-free medium with no detectable tumorigenicity. hPOs can be clonally expanded, genetically manipulated and are amenable to culture in a chemically defined hydrogel. hPOs therefore represent an abundant source of pancreas ductal cells that retain the characteristics of the tissue-of-origin, which opens up avenues for modelling diseases of the ductal epithelium and increasing understanding of human pancreas exocrine biology as well as for potentially producing insulin-secreting cells for the treatment of diabetes.
Collapse
Affiliation(s)
- Nikitas Georgakopoulos
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
- Cambridge Biorepository for Translational Medicine & Department of Surgery, University o.f Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| | - Nicole Prior
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| | | | - Gianmarco Mastrogiovanni
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Alex Cagan
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Daisy Harrison
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Christopher J Hindley
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
- Department of Physics, The Cavendish Laboratory, University of Cambridge, Thompson Avenue, Cambridge, JJ, CB3 0HE, UK
| | - Robert Arnes-Benito
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| | - Siong-Seng Liau
- Hepatopancreatobiliary Surgical Unit, Addenbrooke's Hospital and MRC Cancer Unit, University of Cambridge, Cambridge, CB2 0XZ, UK
| | - Abbie Curd
- Cambridge Biorepository for Translational Medicine & Department of Surgery, University o.f Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| | - Natasha Ivory
- Cambridge Biorepository for Translational Medicine & Department of Surgery, University o.f Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| | - Benjamin D Simons
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
- Department of Physics, The Cavendish Laboratory, University of Cambridge, Thompson Avenue, Cambridge, JJ, CB3 0HE, UK
| | | | | | - Kourosh Saeb-Parsy
- Cambridge Biorepository for Translational Medicine & Department of Surgery, University o.f Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK.
| | - Meritxell Huch
- The Wellcome Trust/ Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK.
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany.
| |
Collapse
|
18
|
Increased proliferation and altered cell cycle regulation in pancreatic stem cells derived from patients with congenital hyperinsulinism. PLoS One 2019; 14:e0222350. [PMID: 31525223 PMCID: PMC6746350 DOI: 10.1371/journal.pone.0222350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Congenital hyperinsulinism (CHI) is characterised by inappropriate insulin secretion causing profound hypoglycaemia and brain damage if inadequately controlled. Pancreatic tissue isolated from patients with diffuse CHI shows abnormal proliferation rates, the mechanisms of which are not fully resolved. Understanding cell proliferation in CHI may lead to new therapeutic options, alongside opportunities to manipulate β-cell mass in patients with diabetes. We aimed to generate cell-lines from CHI pancreatic tissue to provide in vitro model systems for research. Three pancreatic mesenchymal stem cell-lines (CHIpMSC1-3) were derived from patients with CHI disease variants: focal, atypical and diffuse. All CHIpMSC lines demonstrated increased proliferation compared with control adult-derived pMSCs. Cell cycle alterations including increased CDK1 levels and decreased p27Kip1 nuclear localisation were observed in CHIpMSCs when compared to control pMSCs. In conclusion, CHIpMSCs are a useful in vitro model to further understand the cell cycle alterations leading to increased islet cell proliferation in CHI.
Collapse
|
19
|
Efrat S. Beta-Cell Dedifferentiation in Type 2 Diabetes: Concise Review. Stem Cells 2019; 37:1267-1272. [PMID: 31298804 DOI: 10.1002/stem.3059] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is caused by an inherited predisposition to pancreatic islet β-cell failure, which is manifested under cellular stress induced by metabolic overload. The decrease in the functional β-cell mass associated with T2D has been attributed primarily to β-cell death; however, studies in recent years suggested that β-cell dedifferentiation may contribute to this decline. The mechanisms linking genetic factors and cellular stress to β-cell dedifferentiation remain largely unknown. This study evaluated the evidence for β-cell dedifferentiation in T2D, and T2D and examined experimental systems in which its mechanisms may be studied. Understanding these mechanisms may allow prevention of β-cell dedifferentiation or induction of cell redifferentiation for restoration of the functional β-cell mass. Stem Cells 2019;37:1267-1272.
Collapse
Affiliation(s)
- Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Li Y, Deng S, Peng J, Wang X, Essandoh K, Mu X, Peng T, Meng ZX, Fan GC. MicroRNA-223 is essential for maintaining functional β-cell mass during diabetes through inhibiting both FOXO1 and SOX6 pathways. J Biol Chem 2019; 294:10438-10448. [PMID: 31118273 DOI: 10.1074/jbc.ra119.007755] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/16/2019] [Indexed: 01/26/2023] Open
Abstract
The initiation and development of diabetes are mainly ascribed to the loss of functional β-cells. Therapies designed to regenerate β-cells provide great potential for controlling glucose levels and thereby preventing the devastating complications associated with diabetes. This requires detailed knowledge of the molecular events and underlying mechanisms in this disorder. Here, we report that expression of microRNA-223 (miR-223) is up-regulated in islets from diabetic mice and humans, as well as in murine Min6 β-cells exposed to tumor necrosis factor α (TNFα) or high glucose. Interestingly, miR-223 knockout (KO) mice exhibit impaired glucose tolerance and insulin resistance. Further analysis reveals that miR-223 deficiency dramatically suppresses β-cell proliferation and insulin secretion. Mechanistically, using luciferase reporter gene assays, histological analysis, and immunoblotting, we demonstrate that miR-223 inhibits both forkhead box O1 (FOXO1) and SRY-box 6 (SOX6) signaling, a unique bipartite mechanism that modulates expression of several β-cell markers (pancreatic and duodenal homeobox 1 (PDX1), NK6 homeobox 1 (NKX6.1), and urocortin 3 (UCN3)) and cell cycle-related genes (cyclin D1, cyclin E1, and cyclin-dependent kinase inhibitor P27 (P27)). Importantly, miR-223 overexpression in β-cells could promote β-cell proliferation and improve β-cell function. Taken together, our results suggest that miR-223 is a critical factor for maintaining functional β-cell mass and adaptation during metabolic stress.
Collapse
Affiliation(s)
- Yutian Li
- From the Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Shan Deng
- From the Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267.,Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China 430074
| | - Jiangtong Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China 430074
| | - Xiaohong Wang
- From the Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Kobina Essandoh
- From the Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Xingjiang Mu
- From the Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Tianqing Peng
- Critical Illness Research, Lawson Health Research Institute, Ontario, Canada N6C 2R5, and
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China 310058
| | - Guo-Chang Fan
- From the Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267,
| |
Collapse
|
21
|
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: 19.2] [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
|
22
|
Navarro-Tableros V, Gomez Y, Brizzi MF, Camussi G. Generation of Human Stem Cell-Derived Pancreatic Organoids (POs) for Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:179-220. [PMID: 31025308 DOI: 10.1007/5584_2019_340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-dependent diabetes mellitus or type 1 diabetes mellitus (T1DM) is an auto-immune condition characterized by the loss of pancreatic β-cells. The curative approach for highly selected patients is the pancreas or the pancreatic islet transplantation. Nevertheless, these options are limited by a growing shortage of donor organs and by the requirement of immunosuppression.Xenotransplantation of porcine islets has been extensively investigated. Nevertheless, the strong xenoimmunity and the risk of transmission of porcine endogenous retroviruses, have limited their application in clinic. Generation of β-like cells from stem cells is one of the most promising strategies in regenerative medicine. Embryonic, and more recently, adult stem cells are currently the most promising cell sources exploited to generate functional β-cells in vitro. A number of studies demonstrated that stem cells could generate functional pancreatic organoids (POs), able to restore normoglycemia when implanted in different preclinical diabetic models. Nevertheless, a gradual loss of function and cell dead are commonly detected when POs are transplanted in immunocompetent animals. So far, the main issue to be solved is the post-transplanted islet loss, due to the host immune attack. To avoid this hurdle, nanotechnology has provided a number of polymers currently under investigation for islet micro and macro-encapsulation. These new approaches, besides conferring PO immune protection, are able to supply oxygen and nutrients and to preserve PO morphology and long-term viability.Herein, we summarize the current knowledge on bioengineered POs and the stem cell differentiation platforms. We also discuss the in vitro strategies used to generate functional POs, and the protocols currently used to confer immune-protection against the host immune attack (micro- and macro-encapsulation). In addition, the most relevant ongoing clinical trials, and the most relevant hurdles met to move towards clinical application are revised.
Collapse
Affiliation(s)
- Victor Navarro-Tableros
- 2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Turin, Turin, Italy
| | - Yonathan Gomez
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy.
- Fondazione per la Ricerca Biomedica-ONLUS, Turin, Italy.
| |
Collapse
|
23
|
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: 1.8] [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
|
24
|
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: 1.5] [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
|
25
|
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: 1.0] [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
|
26
|
Roefs MM, Carlotti F, Jones K, Wills H, Hamilton A, Verschoor M, Durkin JMW, Garcia-Perez L, Brereton MF, McCulloch L, Engelse MA, Johnson PRV, Hansen BC, Docherty K, de Koning EJP, Clark A. Increased vimentin in human α- and β-cells in type 2 diabetes. J Endocrinol 2017; 233:217-227. [PMID: 28348116 DOI: 10.1530/joe-16-0588] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022]
Abstract
Type 2 diabetes (T2DM) is associated with pancreatic islet dysfunction. Loss of β-cell identity has been implicated via dedifferentiation or conversion to other pancreatic endocrine cell types. How these transitions contribute to the onset and progression of T2DM in vivo is unknown. The aims of this study were to determine the degree of epithelial-to-mesenchymal transition occurring in α and β cells in vivo and to relate this to diabetes-associated (patho)physiological conditions. The proportion of islet cells expressing the mesenchymal marker vimentin was determined by immunohistochemistry and quantitative morphometry in specimens of pancreas from human donors with T2DM (n = 28) and without diabetes (ND, n = 38) and in non-human primates at different stages of the diabetic syndrome: normoglycaemic (ND, n = 4), obese, hyperinsulinaemic (HI, n = 4) and hyperglycaemic (DM, n = 8). Vimentin co-localised more frequently with glucagon (α-cells) than with insulin (β-cells) in the human ND group (1.43% total α-cells, 0.98% total β-cells, median; P < 0.05); these proportions were higher in T2DM than ND (median 4.53% α-, 2.53% β-cells; P < 0.05). Vimentin-positive β-cells were not apoptotic, had reduced expression of Nkx6.1 and Pdx1, and were not associated with islet amyloidosis or with bihormonal expression (insulin + glucagon). In non-human primates, vimentin-positive β-cell proportion was larger in the diabetic than the ND group (6.85 vs 0.50%, medians respectively, P < 0.05), but was similar in ND and HI groups. In conclusion, islet cell expression of vimentin indicates a degree of plasticity and dedifferentiation with potential loss of cellular identity in diabetes. This could contribute to α- and β-cell dysfunction in T2DM.
Collapse
Affiliation(s)
- Maaike M Roefs
- Department of Internal MedicineLeiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Françoise Carlotti
- Department of Internal MedicineLeiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Katherine Jones
- Oxford Centre for DiabetesEndocrinology and Metabolism (OCDEM), Oxford, UK
| | - Hannah Wills
- Oxford Centre for DiabetesEndocrinology and Metabolism (OCDEM), Oxford, UK
| | - Alexander Hamilton
- Oxford Centre for DiabetesEndocrinology and Metabolism (OCDEM), Oxford, UK
| | - Michael Verschoor
- Department of Internal MedicineLeiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Laura Garcia-Perez
- Department of Internal MedicineLeiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Melissa F Brereton
- Department of PhysiologyAnatomy and Genetics, University of Oxford, Oxford, UK
| | - Laura McCulloch
- Oxford Centre for DiabetesEndocrinology and Metabolism (OCDEM), Oxford, UK
| | - Marten A Engelse
- Department of Internal MedicineLeiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Paul R V Johnson
- Oxford Centre for DiabetesEndocrinology and Metabolism (OCDEM), Oxford, UK
- Nuffield Department of Surgical SciencesJohn Radcliffe Hospital, Oxford, UK
| | - Barbara C Hansen
- Departments of Internal Medicine and PediatricsMorsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | | | - Eelco J P de Koning
- Department of Internal MedicineLeiden University Medical Center (LUMC), Leiden, the Netherlands
- Hubrecht InstituteUtrecht, the Netherlands
| | - Anne Clark
- Oxford Centre for DiabetesEndocrinology and Metabolism (OCDEM), Oxford, UK
| |
Collapse
|
27
|
Westenfelder C, Gooch A, Hu Z, Ahlstrom J, Zhang P. Durable Control of Autoimmune Diabetes in Mice Achieved by Intraperitoneal Transplantation of "Neo-Islets," Three-Dimensional Aggregates of Allogeneic Islet and "Mesenchymal Stem Cells". Stem Cells Transl Med 2017; 6:1631-1643. [PMID: 28467694 PMCID: PMC5689775 DOI: 10.1002/sctm.17-0005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/01/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Novel interventions that reestablish endogenous insulin secretion and thereby halt progressive end-organ damage and prolong survival of patients with autoimmune Type 1 diabetes mellitus (T1DM) are urgently needed. While this is currently accomplished with allogeneic pancreas or islet transplants, their utility is significantly limited by both the scarcity of organ donors and life-long need for often-toxic antirejection drugs. Coadministering islets with bone marrow-derived mesenchymal stem cells (MSCs) that exert robust immune-modulating, anti-inflammatory, anti-apoptotic, and angiogenic actions, improves intrahepatic islet survival and function. Encapsulation of insulin-producing cells to prevent immune destruction has shown both promise and failures. Recently, stem cell-derived insulin secreting β-like cells induced euglycemia in diabetic animals, although their clinical use would still require encapsulation or anti-rejection drugs. Instead of focusing on further improvements in islet transplantation, we demonstrate here that the intraperitoneal administration of islet-sized "Neo-Islets" (NIs), generated by in vitro coaggregation of allogeneic, culture-expanded islet cells with high numbers of immuno-protective and cyto-protective MSCs, resulted in their omental engraftment in immune-competent, spontaneously diabetic nonobese diabetic (NOD) mice. This achieved long-term glycemic control without immunosuppression and without hypoglycemia. In preparation for an Food and Drug Administration-approved clinical trial in dogs with T1DM, we show that treatment of streptozotocin-diabetic NOD/severe combined immunodeficiency mice with identically formed canine NIs produced durable euglycemia, exclusively mediated by dog-specific insulin. We conclude that this novel technology has significant translational relevance for canine and potentially clinical T1DM as it effectively addresses both the organ donor scarcity (>80 therapeutic NI doses/donor pancreas can be generated) and completely eliminates the need for immunosuppression. Stem Cells Translational Medicine 2017;6:1631-1643.
Collapse
Affiliation(s)
- Christof Westenfelder
- Department of Medicine, Division of Nephrology, University of Utah and VA Medical Centers, Salt Lake City, Utah, USA
| | - Anna Gooch
- SymbioCellTech, LLC, Salt Lake City, Utah, USA
| | - Zhuma Hu
- SymbioCellTech, LLC, Salt Lake City, Utah, USA
| | | | - Ping Zhang
- SymbioCellTech, LLC, Salt Lake City, Utah, USA
| |
Collapse
|
28
|
Wang W, Liu C, Jimenez-Gonzalez M, Song WJ, Hussain MA. The undoing and redoing of the diabetic β-cell. J Diabetes Complications 2017; 31:912-917. [PMID: 28242267 PMCID: PMC5450161 DOI: 10.1016/j.jdiacomp.2017.01.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/23/2016] [Accepted: 01/31/2017] [Indexed: 02/08/2023]
Abstract
A hallmark of type 2 diabetes (T2DM) is the reduction in functional β-cell mass, which is considered at least in part to result from an imbalance of β-cell renewal and apoptosis, with the latter being accelerated during metabolic stress. More recent studies, however, suggest that the loss of functional β-cell mass is not as much due to β-cell death but rather to de-differentiation of β-cells when these cells are exposed to metabolic stressors, opening the possibility to re-differentiate and restore functional β-cell mass by therapeutic intervention. In parallel, clinical observations suggest that temporary intensive insulin therapy in early diagnosed humans with T2DM, so as to "rest" endogenous β-cells, allows these patients to regain adequate insulin secretion and to maintain euglycemia for prolonged periods free of continued pharmacotherapy. Whether observations made in (mostly rodent) models of diabetes mellitus and in clinical trials are revealing identical mechanisms and therapeutic opportunities remains a tantalizing possibility. Our intention is for this review to serve as an overview of the field and commentary of this particularly exciting field of research.
Collapse
Affiliation(s)
- Wei Wang
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Chune Liu
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Maria Jimenez-Gonzalez
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Woo-Jin Song
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287
| | - Mehboob A Hussain
- Metabolism Division, Departments of Pediatrics, Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, 600 N Wolfe Street, CMSC 10-113, Baltimore, MD 21287.
| |
Collapse
|
29
|
Joglekar MV, Trivedi PM, Kay TW, Hawthorne WJ, O'Connell PJ, Jenkins AJ, Hardikar AA, Thomas HE. Human islet cells are killed by BID-independent mechanisms in response to FAS ligand. Apoptosis 2016; 21:379-89. [PMID: 26758067 DOI: 10.1007/s10495-016-1212-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cell death via FAS/CD95 can occur either by activation of caspases alone (extrinsic) or by activation of mitochondrial death signalling (intrinsic) depending on the cell type. The BH3-only protein BID is activated in the BCL-2-regulated or mitochondrial apoptosis pathway and acts as a switch between the extrinsic and intrinsic cell death pathways. We have previously demonstrated that islets from BID-deficient mice are protected from FAS ligand-mediated apoptosis in vitro. However, it is not yet known if BID plays a similar role in human beta cell death. We therefore aimed to test the role of BID in human islet cell apoptosis immediately after isolation from human cadaver donors, as well as after de-differentiation in vitro. Freshly isolated human islets or 10-12 day cultured human islet cells exhibited BID transcript knockdown after BID siRNA transfection, however they were not protected from FAS ligand-mediated cell death in vitro as determined by DNA fragmentation analysis using flow cytometry. On the other hand, the same cells transfected with siRNA for FAS-associated via death domain (FADD), a molecule in the extrinsic cell death pathway upstream of BID, showed significant reduction in cell death. De-differentiated islets (human islet-derived progenitor cells) also demonstrated similar results with no difference in cell death after BID knockdown as compared to scramble siRNA transfections. Our results indicate that BID-independent pathways are responsible for FAS-dependent human islet cell death. These results are different from those observed in mouse islets and therefore demonstrate potentially alternate pathways of FAS ligand-induced cell death in human and mouse islet cells.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Prerak M Trivedi
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Thomas W Kay
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Wayne J Hawthorne
- The Centre for Transplant and Renal Research, Westmead Millennium Research Institute, University of Sydney, Westmead, Australia
| | - Philip J O'Connell
- The Centre for Transplant and Renal Research, Westmead Millennium Research Institute, University of Sydney, Westmead, Australia
| | - Alicia J Jenkins
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia. .,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
30
|
Efrat S. Mechanisms of adult human β-cell in vitro dedifferentiation and redifferentiation. Diabetes Obes Metab 2016; 18 Suppl 1:97-101. [PMID: 27615137 DOI: 10.1111/dom.12724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/20/2016] [Indexed: 12/13/2022]
Abstract
Recent studies in animal models and human pathological specimens suggest the involvement of β-cell dedifferentiation in β-cell dysfunction associated with type 2 diabetes. Dedifferentiated β-cells may be exploited for endogenous renewal of the β-cell mass. However, studying human β-cell dedifferentiation in diabetes presents major difficulties. We have analysed mechanisms involved in human β-cell dedifferentiation in vitro, under conditions that allow cell proliferation. Although there are important differences between the two cellular environments, β-cell dedifferentiation in the two conditions is likely to share a number of common pathways. Insights from the in vitro studies may lead to development of approaches for redifferentiation of endogenous dedifferentiated β-cells.
Collapse
Affiliation(s)
- S Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel.
| |
Collapse
|
31
|
Sionov RV, Finesilver G, Sapozhnikov L, Soroker A, Zlotkin-Rivkin E, Saad Y, Kahana M, Bodaker M, Alpert E, Mitrani E. Beta Cells Secrete Significant and Regulated Levels of Insulin for Long Periods when Seeded onto Acellular Micro-Scaffolds. Tissue Eng Part A 2016; 21:2691-702. [PMID: 26416226 DOI: 10.1089/ten.tea.2014.0711] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The aim of this work is to obtain significant and regulated insulin secretion from human beta cells ex vivo. Long-term culture of human pancreatic islets and attempts at expanding human islet cells normally result in loss of beta-cell phenotype. We propose that to obtain proper ex vivo beta cell function, there is a need to develop three-dimensional structures that mimic the natural islet tissue microenvironment. We here describe the preparation of endocrine micro-pancreata (EMPs) that are made up of acellular organ-derived micro-scaffolds seeded with human intact or enzymatically dissociated islets. We show that EMPs constructed by seeding whole islets, freshly enzymatically-dissociated islets or even dissociated islets grown first in standard monolayer cultures express high levels of key beta-cell specific genes and secrete quantities of insulin per cell similar to freshly isolated human islets in a glucose-regulated manner for more than 3 months in vitro.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Gershon Finesilver
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Lena Sapozhnikov
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Avigail Soroker
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Efrat Zlotkin-Rivkin
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Yocheved Saad
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Meygal Kahana
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Matan Bodaker
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Evgenia Alpert
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| | - Eduardo Mitrani
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem , The Alexander Silberman Institute of Life Sciences, Jerusalem, Israel
| |
Collapse
|
32
|
Lima MJ, Muir KR, Docherty HM, McGowan NWA, Forbes S, Heremans Y, Heimberg H, Casey J, Docherty K. Generation of Functional Beta-Like Cells from Human Exocrine Pancreas. PLoS One 2016; 11:e0156204. [PMID: 27243814 PMCID: PMC4887015 DOI: 10.1371/journal.pone.0156204] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/10/2016] [Indexed: 12/24/2022] Open
Abstract
Transcription factor mediated lineage reprogramming of human pancreatic exocrine tissue could conceivably provide an unlimited supply of islets for transplantation in the treatment of diabetes. Exocrine tissue can be efficiently reprogrammed to islet-like cells using a cocktail of transcription factors: Pdx1, Ngn3, MafA and Pax4 in combination with growth factors. We show here that overexpression of exogenous Pax4 in combination with suppression of the endogenous transcription factor ARX considerably enhances the production of functional insulin-secreting β-like cells with concomitant suppression of α-cells. The efficiency was further increased by culture on laminin-coated plates in media containing low glucose concentrations. Immunocytochemistry revealed that reprogrammed cultures were composed of ~45% islet-like clusters comprising >80% monohormonal insulin+ cells. The resultant β-like cells expressed insulin protein levels at ~15–30% of that in adult human islets, efficiently processed proinsulin and packaged insulin into secretory granules, exhibited glucose responsive insulin secretion, and had an immediate and prolonged effect in normalising blood glucose levels upon transplantation into diabetic mice. We estimate that approximately 3 billion of these cells would have an immediate therapeutic effect following engraftment in type 1 diabetes patients and that one pancreas would provide sufficient tissue for numerous transplants.
Collapse
Affiliation(s)
- Maria J. Lima
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
- * E-mail:
| | - Kenneth R. Muir
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Hilary M. Docherty
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Neil W. A. McGowan
- Department of Surgery, University of Edinburgh, Edinburgh Royal Infirmary, Edinburgh, EH16 4SU, United Kingdom
| | - Shareen Forbes
- Endocrinology Unit, University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Yves Heremans
- Diabetes Research Centre, Vrije Universiteit Brussel, B1090 Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Centre, Vrije Universiteit Brussel, B1090 Brussels, Belgium
| | - John Casey
- Department of Surgery, University of Edinburgh, Edinburgh Royal Infirmary, Edinburgh, EH16 4SU, United Kingdom
| | - Kevin Docherty
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| |
Collapse
|
33
|
Domínguez-Bendala J, Lanzoni G, Klein D, Álvarez-Cubela S, Pastori RL. The Human Endocrine Pancreas: New Insights on Replacement and Regeneration. Trends Endocrinol Metab 2016; 27:153-162. [PMID: 26774512 DOI: 10.1016/j.tem.2015.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 12/24/2022]
Abstract
Islet transplantation is an effective cell therapy for type 1 diabetes (T1D) but its clinical application is limited due to shortage of donors. After a decade-long period of exploration of potential alternative cell sources, the field has only recently zeroed in on two of them as the most likely to replace islets. These are pluripotent stem cells (PSCs) (through directed differentiation) and pancreatic non-endocrine cells (through directed differentiation or reprogramming). Here we review progress in both areas, including the initiation of Phase I/II clinical trials using human embryonic stem cell (hESc)-derived progenitors, advances in hESc differentiation in vitro, novel insights on the developmental plasticity of the pancreas, and groundbreaking new approaches to induce β cell conversion from the non-endocrine compartment without genetic manipulation.
Collapse
Affiliation(s)
- Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Giacomo Lanzoni
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
34
|
Hayek A, King CC. Brief review: cell replacement therapies to treat type 1 diabetes mellitus. Clin Diabetes Endocrinol 2016; 2:4. [PMID: 28702240 PMCID: PMC5471705 DOI: 10.1186/s40842-016-0023-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/19/2016] [Indexed: 12/22/2022] Open
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent cells (iPSCs) have the potential to differentiate into any somatic cell, making them ideal candidates for cell replacement therapies to treat a number of human diseases and regenerate damaged or non-functional tissues and organs. Key to the promise of regenerative medicine is developing standardized protocols that can safely be applied in patients. Progress towards this goal has occurred in a number of fields, including type 1 diabetes mellitus (T1D). During the past 10 years, significant technological advances in hESC/iPSC biochemistry have provided a roadmap to generate sufficient quantities of glucose-responsive, insulin-producing cells capable of eliminating diabetes in rodents. Although many of the molecular mechanisms underlying the genesis of these cells remain to be elucidated, the field of cell-based therapeutics to treat T1D has advanced to the point where the first Phase I/II trials in humans have begun. Here, we provide a concise review of the history of cell replacement therapies to treat T1D from islet transplantations and xenotranplantation, to current work in hESC/iPSC. We also highlight the latest advances in efforts to employ insulin-producing, glucose-responsive β-like cells derived from hESC as therapeutics.
Collapse
Affiliation(s)
- Alberto Hayek
- Scripps Whittier Diabetes Institute, La Jolla, CA 92037 USA
| | - Charles C King
- Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093 USA
| |
Collapse
|
35
|
Redifferentiation of expanded human islet β cells by inhibition of ARX. Sci Rep 2016; 6:20698. [PMID: 26856418 PMCID: PMC4746595 DOI: 10.1038/srep20698] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/11/2016] [Indexed: 12/14/2022] Open
Abstract
Ex-vivo expansion of adult human islet β cells has been evaluated for generation of abundant insulin-producing cells for transplantation; however, lineage-tracing has demonstrated that this process results in β-cell dedifferentiation. Redifferentiation of β-cell-derived (BCD) cells can be achieved using a combination of soluble factors termed Redifferentiation Cocktail (RC); however, this treatment leads to redifferentiation of only a fraction of BCD cells. This study aimed at improving redifferentiation efficiency by affecting the balance of islet progenitor-cell transcription factors activated by RC treatment. Specifically, RC treatment induces the transcription factors PAX4 and ARX, which play key roles in directing pancreas endocrine progenitor cells into the β/δ or α/PP developmental pathways, respectively. Misactivation of ARX in RC-treated BCD cells may inhibit their redifferentiation into β cells. Blocking ARX expression by shRNA elevated insulin mRNA levels 12.8-fold, and more than doubled the number of insulin-positive BCD cells. ARX inhibition in expanded α-cell-derived cells treated with RC did not cause their transdifferentiation into insulin-producing cells. The combination of RC and ARX shRNA treatment may facilitate the generation of abundant insulin-producing cells for transplantation into patients with type 1 diabetes.
Collapse
|
36
|
Progenitor cells may aid successful islet compensation in metabolically healthy obese individuals. Med Hypotheses 2016; 86:97-9. [DOI: 10.1016/j.mehy.2015.10.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/26/2015] [Indexed: 12/29/2022]
|
37
|
Klein D, Álvarez-Cubela S, Lanzoni G, Vargas N, Prabakar KR, Boulina M, Ricordi C, Inverardi L, Pastori RL, Domínguez-Bendala J. BMP-7 Induces Adult Human Pancreatic Exocrine-to-Endocrine Conversion. Diabetes 2015; 64:4123-34. [PMID: 26307584 PMCID: PMC4657585 DOI: 10.2337/db15-0688] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/17/2015] [Indexed: 12/30/2022]
Abstract
The exocrine pancreas can give rise to endocrine insulin-producing cells upon ectopic expression of key transcription factors. However, the need for genetic manipulation remains a translational hurdle for diabetes therapy. Here we report the conversion of adult human nonendocrine pancreatic tissue into endocrine cell types by exposure to bone morphogenetic protein 7. The use of this U.S. Food and Drug Administration-approved agent, without any genetic manipulation, results in the neogenesis of clusters that exhibit high insulin content and glucose responsiveness both in vitro and in vivo. In vitro lineage tracing confirmed that BMP-7-induced insulin-expressing cells arise mainly from extrainsular PDX-1(+), carbonic anhydrase II(-) (mature ductal), elastase 3a (acinar)(-) , and insulin(-) subpopulations. The nongenetic conversion of human pancreatic exocrine cells to endocrine cells is novel and represents a safer and simpler alternative to genetic reprogramming.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- Bone Morphogenetic Protein 7/genetics
- Bone Morphogenetic Protein 7/metabolism
- Bone Morphogenetic Protein 7/pharmacology
- C-Peptide/blood
- C-Peptide/metabolism
- Cell Lineage
- Cell Transdifferentiation/drug effects
- Cells, Cultured
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/therapy
- Fluorescent Antibody Technique
- Homeodomain Proteins/metabolism
- Humans
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Insulin-Secreting Cells/transplantation
- Kidney
- Male
- Mice, Nude
- Pancreas, Exocrine/drug effects
- Pancreas, Exocrine/metabolism
- Pancreas, Exocrine/pathology
- Recombinant Proteins/metabolism
- Recombinant Proteins/pharmacology
- Trans-Activators/metabolism
- Transplantation, Heterologous
- Transplantation, Heterotopic
Collapse
Affiliation(s)
- Dagmar Klein
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Giacomo Lanzoni
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Nancy Vargas
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Kamalaveni R Prabakar
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Maria Boulina
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Camillo Ricordi
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL Department of Biomedical Engineering, Miller School of Medicine, University of Miami, Miami, FL
| | - Luca Inverardi
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL
| | - Ricardo L Pastori
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL Department of Cell Biology and Anatomy, Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
38
|
Toren-Haritan G, Efrat S. TGFβ Pathway Inhibition Redifferentiates Human Pancreatic Islet β Cells Expanded In Vitro. PLoS One 2015; 10:e0139168. [PMID: 26418361 PMCID: PMC4587799 DOI: 10.1371/journal.pone.0139168] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/08/2015] [Indexed: 11/18/2022] Open
Abstract
In-vitro expansion of insulin-producing cells from adult human pancreatic islets could provide an abundant cell source for diabetes therapy. However, proliferation of β-cell-derived (BCD) cells is associated with loss of phenotype and epithelial-mesenchymal transition (EMT). Nevertheless, BCD cells maintain open chromatin structure at β-cell genes, suggesting that they could be readily redifferentiated. The transforming growth factor β (TGFβ) pathway has been implicated in EMT in a range of cell types. Here we show that human islet cell expansion in vitro involves upregulation of the TGFβ pathway. Blocking TGFβ pathway activation using short hairpin RNA (shRNA) against TGFβ Receptor 1 (TGFBR1, ALK5) transcripts inhibits BCD cell proliferation and dedifferentiation. Treatment of expanded BCD cells with ALK5 shRNA results in their redifferentiation, as judged by expression of β-cell genes and decreased cell proliferation. These effects, which are reproducible in cells from multiple human donors, are mediated, at least in part, by AKT-FOXO1 signaling. ALK5 inhibition synergizes with a soluble factor cocktail to promote BCD cell redifferentiation. The combined treatment may offer a therapeutically applicable way for generating an abundant source of functional insulin-producing cells following ex-vivo expansion.
Collapse
Affiliation(s)
- Ginat Toren-Haritan
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
39
|
Sintov E, Nathan G, Knoller S, Pasmanik-Chor M, Russ HA, Efrat S. Inhibition of ZEB1 expression induces redifferentiation of adult human β cells expanded in vitro. Sci Rep 2015; 5:13024. [PMID: 26264186 PMCID: PMC4532995 DOI: 10.1038/srep13024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/15/2015] [Indexed: 01/20/2023] Open
Abstract
In-vitro expansion of functional adult human β-cells is an attractive approach for generating insulin-producing cells for transplantation. However, human islet cell expansion in culture results in loss of β-cell phenotype and epithelial-mesenchymal transition (EMT). This process activates expression of ZEB1 and ZEB2, two members of the zinc-finger homeobox family of E-cadherin repressors, which play key roles in EMT. Downregulation of ZEB1 using shRNA in expanded β-cell-derived (BCD) cells induced mesenchymal-epithelial transition (MET), β-cell gene expression, and proliferation attenuation. In addition, inhibition of ZEB1 expression potentiated redifferentiation induced by a combination of soluble factors, as judged by an improved response to glucose stimulation and a 3-fold increase in the fraction of C-peptide-positive cells to 60% of BCD cells. Furthermore, ZEB1 shRNA led to increased insulin secretion in cells transplanted in vivo. Our findings suggest that the effects of ZEB1 inhibition are mediated by attenuation of the miR-200c target genes SOX6 and SOX2. These findings, which were reproducible in cells derived from multiple human donors, emphasize the key role of ZEB1 in EMT in cultured BCD cells and support the value of ZEB1 inhibition for BCD cell redifferentiation and generation of functional human β-like cells for cell therapy of diabetes.
Collapse
Affiliation(s)
- Elad Sintov
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gili Nathan
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarah Knoller
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Holger A Russ
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Corritore E, Dugnani E, Pasquale V, Misawa R, Witkowski P, Lei J, Markmann J, Piemonti L, Sokal EM, Bonner-Weir S, Lysy PA. β-Cell differentiation of human pancreatic duct-derived cells after in vitro expansion. Cell Reprogram 2015; 16:456-66. [PMID: 25437872 DOI: 10.1089/cell.2014.0025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
β-Cell replacement therapy is a promising field of research that is currently evaluating new sources of cells for clinical use. Pancreatic epithelial cells are potent candidates for β-cell engineering, but their large-scale expansion has not been evidenced yet. Here we describe the efficient expansion and β-cell differentiation of purified human pancreatic duct cells (DCs). When cultured in endothelial growth-promoting media, purified CA19-9(+) cells proliferated extensively and achieved up to 22 population doublings over nine passages. While proliferating, human pancreatic duct-derived cells (HDDCs) downregulated most DC markers, but they retained low CK19 and SOX9 gene expression. HDDCs acquired mesenchymal features but differed from fibroblasts or pancreatic stromal cells. Coexpression of duct and mesenchymal markers suggested that HDDCs were derived from DCs via a partial epithelial-to-mesenchymal transition (EMT). This was supported by the blockade of HDDC appearance in CA19-9(+) cell cultures after incubation with the EMT inhibitor A83-01. After a differentiation protocol mimicking pancreatic development, HDDC populations contained about 2% of immature insulin-producing cells and showed glucose-unresponsive insulin secretion. Downregulation of the mesenchymal phenotype improved β-cell gene expression profile of differentiated HDDCs without affecting insulin protein expression and secretion. We show that pancreatic ducts represent a new source for engineering large amounts of β-like-cells with potential for treating diabetes.
Collapse
Affiliation(s)
- Elisa Corritore
- 1 Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain , B-1200, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cheng Y, Kang H, Shen J, Hao H, Liu J, Guo Y, Mu Y, Han W. Beta-cell regeneration from vimentin+/MafB+ cells after STZ-induced extreme beta-cell ablation. Sci Rep 2015; 5:11703. [PMID: 26129776 PMCID: PMC4486952 DOI: 10.1038/srep11703] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 06/02/2015] [Indexed: 12/16/2022] Open
Abstract
Loss of functional beta-cells is fundamental in both type 1 and type 2 diabetes. In situ beta-cell regeneration therefore has garnered great interest as an approach to diabetes therapy. Here, after elimination of pre-existing beta cells by a single high-dose of streptozotocin (STZ), we demonstrated that a considerable amount of beta-like-cells was generated within 48 hrs. But the newly formed insulin producing cells failed to respond to glucose challenge at this time and diminished afterwards. Insulin treatment to normalize the glucose level protected the neogenic beta-like cells and the islet function was also gradually matured. Strikingly, intermediate cells lacking epithelial marker E-cadherin but expressing mesenchymal cell-specific marker vimentin appeared within 16 hrs following STZ exposure, which served as the major source of insulin-producing cells observed at 24 hrs. Moreover, these intermediate cells strongly expressed alpha-cell-specific marker MafB. In summary, the data presented here identified a novel intermediate cell type as beta-cell progenitors, showing mesenchymal cell feature as well as alpha-cell marker MafB. Our results might have important implications for efforts to stimulate beta-cell regeneration.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Hongjun Kang
- Department of Critical Care Medicine, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Jing Shen
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
- Department of Endocrinology, Chinese PLA 309 Hospital, 17 Heishanhu Road, Beijing 100091, China
| | - Haojie Hao
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Jiejie Liu
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Yelei Guo
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Weidong Han
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| |
Collapse
|
42
|
Nathan G, Kredo-Russo S, Geiger T, Lenz A, Kaspi H, Hornstein E, Efrat S. MiR-375 promotes redifferentiation of adult human β cells expanded in vitro. PLoS One 2015; 10:e0122108. [PMID: 25875172 PMCID: PMC4395232 DOI: 10.1371/journal.pone.0122108] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/17/2015] [Indexed: 11/26/2022] Open
Abstract
In-vitro expansion of β cells from adult human pancreatic islets could provide abundant cells for cell replacement therapy of diabetes. However, proliferation of β-cell-derived (BCD) cells is associated with dedifferentiation. Here we analyzed changes in microRNAs (miRNAs) during BCD cell dedifferentiation and identified miR-375 as one of the miRNAs greatly downregulated. We hypothesized that restoration of miR-375 expression in expanded BCD cells may contribute to their redifferentiation. Our findings demonstrate that overexpression of miR-375 alone leads to activation of β-cell gene expression, reduced cell proliferation, and a switch from N-cadherin to E-cadherin expression, which characterizes mesenchymal-epithelial transition. These effects, which are reproducible in cells derived from multiple human donors, are likely mediated by repression of PDPK1 transcripts and indirect downregulation of GSK3 activity. These findings support an important role of miR-375 in regulation of human β-cell phenotype, and suggest that miR-375 upregulation may facilitate the generation of functional insulin-producing cells following ex-vivo expansion of human islet cells.
Collapse
Affiliation(s)
- Gili Nathan
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Kredo-Russo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ayelet Lenz
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Haggai Kaspi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Hornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
43
|
van der Meulen T, Huising MO. Role of transcription factors in the transdifferentiation of pancreatic islet cells. J Mol Endocrinol 2015; 54:R103-17. [PMID: 25791577 PMCID: PMC4373662 DOI: 10.1530/jme-14-0290] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The α and β cells act in concert to maintain blood glucose. The α cells release glucagon in response to low levels of glucose to stimulate glycogenolysis in the liver. In contrast, β cells release insulin in response to elevated levels of glucose to stimulate peripheral glucose disposal. Despite these opposing roles in glucose homeostasis, α and β cells are derived from a common progenitor and share many proteins important for glucose sensing and hormone secretion. Results from recent work have underlined these similarities between the two cell types by revealing that β-to-α as well as α-to-β transdifferentiation can take place under certain experimental circumstances. These exciting findings highlight unexpected plasticity of adult islets and offer hope of novel therapeutic paths to replenish β cells in diabetes. In this review, we focus on the transcription factor networks that establish and maintain pancreatic endocrine cell identity and how they may be perturbed to facilitate transdifferentiation.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| | - Mark O Huising
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| |
Collapse
|
44
|
Muir KR, Lima MJ, Docherty HM, McGowan NWA, Forbes S, Heremans Y, Forbes SJ, Heimberg H, Casey J, Docherty K. Krüppel-Like Factor 4 Overexpression Initiates a Mesenchymal-to-Epithelial Transition and Redifferentiation of Human Pancreatic Cells following Expansion in Long Term Adherent Culture. PLoS One 2015; 10:e0140352. [PMID: 26457418 PMCID: PMC4601732 DOI: 10.1371/journal.pone.0140352] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/24/2015] [Indexed: 02/01/2023] Open
Abstract
A replenishable source of insulin-producing cells has the potential to cure type 1 diabetes. Attempts to culture and expand pancreatic β-cells in vitro have resulted in their transition from insulin-producing epithelial cells to mesenchymal stromal cells (MSCs) with high proliferative capacity but devoid of any hormone production. The aim of this study was to determine whether the transcription factor Krüppel-like factor 4 (KLF4), could induce a mesenchymal-to-epithelial transition (MET) of the cultured cells. Islet-enriched pancreatic cells, allowed to dedifferentiate and expand in adherent cell culture, were transduced with an adenovirus containing KLF4 (Ad-Klf4). Cells were subsequently analysed for changes in cell morphology by light microscopy, and for the presence of epithelial and pancreatic markers by immunocytochemistry and quantitative RT/PCR. Infection with Ad-Klf4 resulted in morphological changes, down-regulation of mesenchymal markers, and re-expression of both epithelial and pancreatic cell markers including insulin and transcription factors specific to β-cells. This effect was further enhanced by culturing cells in suspension. However, the effects of Ad-KLf4 were transient and this was shown to be due to increased apoptosis in Klf4-expressing cells. Klf4 has been recently identified as a pioneer factor with the ability to modulate the structure of chromatin and enhance reprogramming/transdifferentiation. Our results show that Klf4 may have a role in the redifferentiation of expanded pancreatic cells in culture, but before this can be achieved the off-target effects that result in increased apoptosis would need to be overcome.
Collapse
Affiliation(s)
- Kenneth R. Muir
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | - Maria João Lima
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | - Hilary M. Docherty
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | - Neil W. A. McGowan
- Department of Surgery, University of Edinburgh, Edinburgh Royal Infirmary, Edinburgh, United Kingdom
| | - Shareen Forbes
- Endocrinology Unit, University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stuart J. Forbes
- MRC Centre for Regenerative Medicine, SCRM Building, The University of Edinburgh, Edinburgh, United Kingdom
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - John Casey
- Department of Surgery, University of Edinburgh, Edinburgh Royal Infirmary, Edinburgh, United Kingdom
| | - Kevin Docherty
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
- * E-mail:
| |
Collapse
|
45
|
Redifferentiation of adult human β cells expanded in vitro by inhibition of the WNT pathway. PLoS One 2014; 9:e112914. [PMID: 25393025 PMCID: PMC4231080 DOI: 10.1371/journal.pone.0112914] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/16/2014] [Indexed: 12/20/2022] Open
Abstract
In vitro expansion of adult human islet β cells is an attractive solution for the shortage of tissue for cell replacement therapy of type 1 diabetes. Using a lineage tracing approach we have demonstrated that β-cell-derived (BCD) cells rapidly dedifferentiate in culture and can proliferate for up to 16 population doublings. Dedifferentiation is associated with changes resembling epithelial-mesenchymal transition (EMT). The WNT pathway has been shown to induce EMT and plays key roles in regulating replication and differentiation in many cell types. Here we show that BCD cell dedifferentiation is associated with β-catenin translocation into the nucleus and activation of the WNT pathway. Inhibition of β-catenin expression in expanded BCD cells using short hairpin RNA resulted in growth arrest, mesenchymal-epithelial transition, and redifferentiation, as judged by activation of β-cell gene expression. Furthermore, inhibition of β-catenin expression synergized with redifferentiation induced by a combination of soluble factors, as judged by an increase in the number of C-peptide-positive cells. Simultaneous inhibition of the WNT and NOTCH pathways also resulted in a synergistic effect on redifferentiation. These findings, which were reproducible in cells derived from multiple human donors, suggest that inhibition of the WNT pathway may contribute to a therapeutically applicable way for generation of functional insulin-producing cells following ex-vivo expansion.
Collapse
|
46
|
Filios SR, Xu G, Chen J, Hong K, Jing G, Shalev A. MicroRNA-200 is induced by thioredoxin-interacting protein and regulates Zeb1 protein signaling and beta cell apoptosis. J Biol Chem 2014; 289:36275-83. [PMID: 25391656 DOI: 10.1074/jbc.m114.592360] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Small noncoding microRNAs have emerged as important regulators of cellular processes, but their role in pancreatic beta cells has only started to be elucidated. Loss of pancreatic beta cells is a key factor in the pathogenesis of diabetes, and we have demonstrated that beta cell expression of thioredoxin-interacting protein (TXNIP) is increased in diabetes and causes beta cell apoptosis, whereas TXNIP deficiency is protective against diabetes. Recently, we found that TXNIP also impairs beta cell function by inducing microRNA (miR)-204. Interestingly, using INS-1 beta cells and primary islets, we have now discovered that expression of another microRNA, miR-200, is induced by TXNIP and by diabetes. Furthermore, we found that miR-200 targeted and decreased Zeb1 (zinc finger E-box-binding homeobox 1) and promoted beta cell apoptosis as measured by cleaved caspase-3 levels, Bax/Bcl2 ratio, and TUNEL. In addition, Zeb1 knockdown mimicked the miR-200 effects on beta cell apoptosis, suggesting that Zeb1 plays an important role in mediating miR-200 effects. Moreover, miR-200 increased beta cell expression of the epithelial marker E-cadherin, consistent with inhibition of epithelial-mesenchymal transition, a process thought to be involved in beta cell expansion. Thus, we have identified a novel TXNIP/miR-200/Zeb1/E-cadherin signaling pathway that, for the first time, links miR-200 to beta cell apoptosis and diabetes and also beta cell TXNIP to epithelial-mesenchymal transition. In addition, our results shed new light on the regulation and function of miR-200 in beta cells and show that TXNIP-induced microRNAs control various processes of beta cell biology.
Collapse
Affiliation(s)
- Stephen R Filios
- From the Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-2182
| | - Guanlan Xu
- From the Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-2182
| | - Junqin Chen
- From the Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-2182
| | - Kyunghee Hong
- From the Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-2182
| | - Gu Jing
- From the Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-2182
| | - Anath Shalev
- From the Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-2182
| |
Collapse
|
47
|
Minami K, Seino S. Current status of regeneration of pancreatic β-cells. J Diabetes Investig 2014; 4:131-41. [PMID: 24843642 PMCID: PMC4019265 DOI: 10.1111/jdi.12062] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 01/21/2013] [Indexed: 12/13/2022] Open
Abstract
Newly generated insulin‐secreting cells for use in cell therapy for insulin‐deficient diabetes mellitus require properties similar to those of native pancreatic β‐cells. Pancreatic β‐cells are highly specialized cells that produce a large amount of insulin, and secrete insulin in a regulated manner in response to glucose and other stimuli. It is not yet explained how the β‐cells acquire this complex function during normal differentiation. So far, in vitro generation of insulin‐secreting cells from embryonic stem cells, induced‐pluripotent stem cells and adult stem/progenitor‐like cells has been reported. However, most of these cells are functionally immature and show poor glucose‐responsive insulin secretion compared to that of native pancreatic β‐cells (or islets). Strategies to generate functional β‐cells or a whole organ in vivo have also recently been proposed. Establishing a protocol to generate fully functional insulin‐secreting cells that closely resemble native β‐cells is a critical matter in regenerative medicine for diabetes. Understanding the physiological processes of differentiation, proliferation and regeneration of pancreatic β‐cells might open the path to cell therapy to cure patients with absolute insulin deficiency.
Collapse
Affiliation(s)
- Kohtaro Minami
- Division of Cellular and Molecular Medicine Department of Physiology and Cell Biology Kobe University Graduate School of Medicine Kobe Japan
| | - Susumu Seino
- Division of Cellular and Molecular Medicine Department of Physiology and Cell Biology Kobe University Graduate School of Medicine Kobe Japan ; Division of Diabetes and Endocrinology Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan ; Core Research for Evolutional Science and Technology (CREST) Japan Science and Technology Corp. Kawaguchi Saitama Japan
| |
Collapse
|
48
|
Shimoda M, Chen S, Noguchi H, Takita M, Sugimoto K, Itoh T, Chujo D, Iwahashi S, Naziruddin B, Levy MF, Matsumoto S, Grayburn PA. A new method for generating insulin-secreting cells from human pancreatic epithelial cells after islet isolation transformed by NeuroD1. Hum Gene Ther Methods 2014; 25:206-19. [PMID: 24845703 DOI: 10.1089/hgtb.2013.122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The generation of insulin-secreting cells from nonendocrine pancreatic epithelial cells (NEPEC) has been demonstrated for potential clinical use in the treatment of diabetes. However, previous methods either had limited efficacy or required viral vectors, which hinder clinical application. In this study, we aimed to establish an efficient method of insulin-secreting cell generation from NEPEC without viral vectors. We used nonislet fractions from both research-grade human pancreata from brain-dead donors and clinical pancreata after total pancreatectomy with autologous islet transplantation to treat chronic pancreatitis. It is of note that a few islets could be mingled in the nonislet fractions, but their influence could be limited. The NeuroD1 gene was induced into NEPEC using an effective triple lipofection method without viral vectors to generate insulin-secreting cells. The differentiation was promoted by adding a growth factor cocktail into the culture medium. Using the research-grade human pancreata, the effective method showed high efficacy in the differentiation of NEPEC into insulin-positive cells that secreted insulin in response to a glucose challenge and improved diabetes after being transplanted into diabetic athymic mice. Using the clinical pancreata, similar efficacy was obtained, even though those pancreata suffered chronic pancreatitis. In conclusion, our effective differentiation protocol with triple lipofection method enabled us to achieve very efficient insulin-secreting cell generation from human NEPEC without viral vectors. This method offers the potential for supplemental insulin-secreting cell transplantation for both allogeneic and autologous islet transplantation.
Collapse
|
49
|
Lee SJ, Kim KH, Park KK. Mechanisms of fibrogenesis in liver cirrhosis: The molecular aspects of epithelial-mesenchymal transition. World J Hepatol 2014; 6:207-216. [PMID: 24799989 PMCID: PMC4009476 DOI: 10.4254/wjh.v6.i4.207] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/09/2014] [Accepted: 03/04/2014] [Indexed: 02/06/2023] Open
Abstract
Liver injuries are repaired by fibrosis and regeneration. The cause of fibrosis and diminished regeneration, especially in liver cirrhosis, is still unknown. Epithelial-mesenchymal transition (EMT) has been found to be associated with liver fibrosis. The possibility that EMT could contribute to hepatic fibrogenesis reinforced the concept that activated hepatic stellate cells are not the only key players in the hepatic fibrogenic process and that other cell types, either hepatic or bone marrow-derived cells could contribute to this process. Following an initial enthusiasm for the discovery of this novel pathway in fibrogenesis, more recent research has started to cast serious doubts upon the real relevance of this phenomenon in human fibrogenetic disorders. The debate on the authenticity of EMT or on its contribution to the fibrogenic process has become very animated. The overall result is a general confusion on the meaning and on the definition of several key aspects. The aim of this article is to describe how EMT participates to hepatic fibrosis and discuss the evidence of supporting this possibility in order to reach reasonable and useful conclusions.
Collapse
|
50
|
Seeberger KL, Anderson SJ, Ellis CE, Yeung TY, Korbutt GS. Identification and differentiation of PDX1 β-cell progenitors within the human pancreatic epithelium. World J Diabetes 2014; 5:59-68. [PMID: 24567802 PMCID: PMC3932428 DOI: 10.4239/wjd.v5.i1.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 11/27/2013] [Accepted: 12/16/2013] [Indexed: 02/05/2023] Open
Abstract
AIM: To minimize the expansion of pancreatic mesenchymal cells in vitro and confirm that β-cell progenitors reside within the pancreatic epithelium.
METHODS: Due to mesenchymal stem cell (MSC) expansion and overgrowth, progenitor cells within the pancreatic epithelium cannot be characterized in vitro, though β-cell dedifferentiation and expansion of MSC intermediates via epithelial-mesenchymal transition (EMT) may generate β-cell progenitors. Pancreatic epithelial cells from endocrine and non-endocrine tissue were expanded and differentiated in a novel pancreatic epithelial expansion medium supplemented with growth factors known to support epithelial cell growth (dexamethasone, epidermal growth factor, 3,5,3’-triiodo-l-thyronine, bovine brain extract). Cells were also infected with a single and dual lentiviral reporter prior to cell differentiation. Enhanced green fluorescent protein was controlled by the rat Insulin 1 promoter and the monomeric red fluorescent protein was controlled by the mouse PDX1 promoter. In combination with lentiviral tracing, cells expanded and differentiated in the pancreatic medium were characterized by flow cytometry (BD fluorescence activated cell sorting), immunostaining and real-time polymerase chain reaction (PCR) (7900HT Fast Realtime PCR System).
RESULTS: In the presence of 10% serum MSCs rapidly expand in vitro while the epithelial cell population declines. The percentage of vimentin+ cells increased from 22% ± 5.83% to 80.43% ± 3.24% (14 d) and 99.00% ± 0.0% (21 d), and the percentage of epithelial cells decreased from 74.71% ± 8.34% to 26.57% ± 9.75% (14 d) and 4.00% ± 1.53% (21 d), P < 0.01 for all time points. Our novel pancreatic epithelial expansion medium preserved the epithelial cell phenotype and minimized epithelial cell dedifferentiation and EMT. Cells expanded in our epithelial medium contained significantly less mesenchymal cells (vimentin+) compared to controls (44.87% ± 4.93% vs 95.67% ± 1.36%; P < 0.01). During cell differentiation lentiviral reporting demonstrated that, PDX1+ and insulin+ cells were localized within adherent epithelial cell aggregates compared to controls. Compared to starting islets differentiated cells had at least two fold higher gene expression of PDX1, insulin, PAX4 and RFX (P < 0.05).
CONCLUSION: PDX1+ cells were confined to adherent epithelial cell aggregates and not vimentin+ cells (mesenchymal), suggesting that EMT is not a mechanism for generating pancreatic progenitor cells.
Collapse
|