101
|
Gerace D, Zhou Q, Kenty JHR, Veres A, Sintov E, Wang X, Boulanger KR, Li H, Melton DA. Engineering human stem cell-derived islets to evade immune rejection and promote localized immune tolerance. Cell Rep Med 2023; 4:100879. [PMID: 36599351 PMCID: PMC9873825 DOI: 10.1016/j.xcrm.2022.100879] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/02/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023]
Abstract
Immunological protection of transplanted stem cell-derived islet (SC-islet) cells is yet to be achieved without chronic immunosuppression or encapsulation. Existing genetic engineering approaches to produce immune-evasive SC-islet cells have so far shown variable results. Here, we show that targeting human leukocyte antigens (HLAs) and PD-L1 alone does not sufficiently protect SC-islet cells from xenograft (xeno)- or allograft (allo)-rejection. As an addition to these approaches, we genetically engineer SC-islet cells to secrete the cytokines interleukin-10 (IL-10), transforming growth factor β (TGF-β), and modified IL-2 such that they promote a tolerogenic local microenvironment by recruiting regulatory T cells (Tregs) to the islet grafts. Cytokine-secreting human SC-β cells resist xeno-rejection and correct diabetes for up to 8 weeks post-transplantation in non-obese diabetic (NOD) mice. Thus, genetically engineering human embryonic SCs (hESCs) to induce a tolerogenic local microenvironment represents a promising approach to provide SC-islet cells as a cell replacement therapy for diabetes without the requirement for encapsulation or immunosuppression.
Collapse
Affiliation(s)
- Dario Gerace
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Quan Zhou
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Jennifer Hyoje-Ryu Kenty
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Adrian Veres
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Elad Sintov
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Xi Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Kyle R Boulanger
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Hongfei Li
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard University, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
102
|
Halliez C, Ibrahim H, Otonkoski T, Mallone R. In vitro beta-cell killing models using immune cells and human pluripotent stem cell-derived islets: Challenges and opportunities. Front Endocrinol (Lausanne) 2023; 13:1076683. [PMID: 36726462 PMCID: PMC9885197 DOI: 10.3389/fendo.2022.1076683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Type 1 diabetes (T1D) is a disease of both autoimmunity and β-cells. The β-cells play an active role in their own demise by mounting defense mechanisms that are insufficient at best, and that can become even deleterious in the long term. This complex crosstalk is important to understanding the physiological defense mechanisms at play in healthy conditions, their alterations in the T1D setting, and therapeutic agents that may boost such mechanisms. Robust protocols to develop stem-cell-derived islets (SC-islets) from human pluripotent stem cells (hPSCs), and islet-reactive cytotoxic CD8+ T-cells from peripheral blood mononuclear cells offer unprecedented opportunities to study this crosstalk. Challenges to develop in vitro β-cell killing models include the cluster morphology of SC-islets, the relatively weak cytotoxicity of most autoimmune T-cells and the variable behavior of in vitro expanded CD8+ T-cells. These challenges may however be highly rewarding in light of the opportunities offered by such models. Herein, we discuss these opportunities including: the β-cell/immune crosstalk in an islet microenvironment; the features that make β-cells more sensitive to autoimmunity; therapeutic agents that may modulate β-cell vulnerability; and the possibility to perform analyses in an autologous setting, i.e., by generating T-cell effectors and SC-islets from the same donor.
Collapse
Affiliation(s)
- Clémentine Halliez
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
- Department of Pediatrics, Helsinki University Hospital, Helsinki, Finland
| | - Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| |
Collapse
|
103
|
Sakuma K, Tsubooka-Yamazoe N, Hashimoto K, Sakai N, Asano S, Watanabe-Matsumoto S, Watanabe T, Saito B, Matsumoto H, Ueno H, Ito R, Toyoda T. CDK8/19 inhibition plays an important role in pancreatic β-cell induction from human iPSCs. Stem Cell Res Ther 2023; 14:1. [PMID: 36600289 PMCID: PMC9814340 DOI: 10.1186/s13287-022-03220-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Transplantation of differentiated cells from human-induced pluripotent stem cells (hiPSCs) holds great promise for clinical treatments. Eliminating the risk factor of malignant cell transformation is essential for ensuring the safety of such cells. This study was aimed at assessing and mitigating mutagenicity that may arise during the cell culture process in the protocol of pancreatic islet cell (iPIC) differentiation from hiPSCs. METHODS We evaluated the mutagenicity of differentiation factors used for hiPSC-derived pancreatic islet-like cells (iPICs). We employed Ames mutagenicity assay, flow cytometry analysis, immunostaining, time-resolved fluorescence resonance energy transfer-based (TR-FRET) cell-free dose-response assays, single-cell RNA-sequencing and in vivo efficacy study. RESULTS We observed a mutagenic effect of activin receptor-like kinase 5 inhibitor II (ALK5iII). ALK5iII is a widely used β-cell inducer but no other tested ALK5 inhibitors induced β-cells. We obtained kinase inhibition profiles and found that only ALK5iII inhibited cyclin-dependent kinases 8 and 19 (CDK8/19) among all ALK5 inhibitors tested. Consistently, CDK8/19 inhibitors efficiently induced β-cells in the absence of ALK5iII. A combination treatment with non-mutagenic ALK5 inhibitor SB431542 and CDK8/19 inhibitor senexin B afforded generation of iPICs with in vitro cellular composition and in vivo efficacy comparable to those observed with ALK5iII. CONCLUSION Our findings suggest a new risk mitigation approach for cell therapy and advance our understanding of the β-cell differentiation mechanism.
Collapse
Affiliation(s)
- Kensuke Sakuma
- iPSC-Derived Pancreatic Islet Cell (iPIC) Therapy Department, Orizuru Therapeutics Inc., Fujisawa, Kanagawa, 251-8555, Japan. .,Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Kanagawa, 251-8555, Japan.
| | - Noriko Tsubooka-Yamazoe
- iPSC-Derived Pancreatic Islet Cell (iPIC) Therapy Department, Orizuru Therapeutics Inc., Fujisawa, Kanagawa 251-8555 Japan ,Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Kanagawa 251-8555 Japan
| | - Kiyohiro Hashimoto
- grid.419841.10000 0001 0673 6017Drug Safety Research and Evaluation Group, Takeda Pharmaceutical Company Limited, Kanagawa, 251-8555 Japan
| | - Nozomu Sakai
- grid.419841.10000 0001 0673 6017Drug Discovery Sciences, Takeda Pharmaceutical Company Limited, Kanagawa, 251-8555 Japan
| | - Shinya Asano
- Integrated & Translational Science, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa 251-8555 Japan
| | - Saori Watanabe-Matsumoto
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Kanagawa 251-8555 Japan ,grid.258799.80000 0004 0372 2033Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan
| | - Takeshi Watanabe
- grid.419841.10000 0001 0673 6017Drug Safety Research and Evaluation Group, Takeda Pharmaceutical Company Limited, Kanagawa, 251-8555 Japan
| | - Bunnai Saito
- grid.419841.10000 0001 0673 6017Drug Discovery Sciences, Takeda Pharmaceutical Company Limited, Kanagawa, 251-8555 Japan
| | - Hirokazu Matsumoto
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Kanagawa 251-8555 Japan ,grid.419841.10000 0001 0673 6017T-CiRA Discovery and Innovation, Takeda Pharmaceutical Company Limited, Kanagawa, 251-8555 Japan
| | - Hikaru Ueno
- iPSC-Derived Pancreatic Islet Cell (iPIC) Therapy Department, Orizuru Therapeutics Inc., Fujisawa, Kanagawa 251-8555 Japan ,Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Kanagawa 251-8555 Japan
| | - Ryo Ito
- iPSC-Derived Pancreatic Islet Cell (iPIC) Therapy Department, Orizuru Therapeutics Inc., Fujisawa, Kanagawa 251-8555 Japan ,Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Kanagawa 251-8555 Japan
| | - Taro Toyoda
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Kanagawa, 251-8555, Japan. .,Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.
| |
Collapse
|
104
|
Hermann FM, Kjærgaard MF, Tian C, Tiemann U, Jackson A, Olsen LR, Kraft M, Carlsson PO, Elfving IM, Kettunen JLT, Tuomi T, Novak I, Semb H. An insulin hypersecretion phenotype precedes pancreatic β cell failure in MODY3 patient-specific cells. Cell Stem Cell 2023; 30:38-51.e8. [PMID: 36563694 DOI: 10.1016/j.stem.2022.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 10/04/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022]
Abstract
MODY3 is a monogenic hereditary form of diabetes caused by mutations in the transcription factor HNF1A. The patients progressively develop hyperglycemia due to perturbed insulin secretion, but the pathogenesis is unknown. Using patient-specific hiPSCs, we recapitulate the insulin secretion sensitivity to the membrane depolarizing agent sulfonylurea commonly observed in MODY3 patients. Unexpectedly, MODY3 patient-specific HNF1A+/R272C β cells hypersecrete insulin both in vitro and in vivo after transplantation into mice. Consistently, we identified a trend of increased birth weight in human HNF1A mutation carriers compared with healthy siblings. Reduced expression of potassium channels, specifically the KATP channel, in MODY3 β cells, increased calcium signaling, and rescue of the insulin hypersecretion phenotype by pharmacological targeting ATP-sensitive potassium channels or low-voltage-activated calcium channels suggest that more efficient membrane depolarization underlies the hypersecretion of insulin in MODY3 β cells. Our findings identify a pathogenic mechanism leading to β cell failure in MODY3.
Collapse
Affiliation(s)
- Florian M Hermann
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Maya Friis Kjærgaard
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Chenglei Tian
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, München, Germany
| | - Ulf Tiemann
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Abigail Jackson
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Maria Kraft
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Jarno L T Kettunen
- Folkhalsan Research Center, Helsinki, Finland; Institute for Molecular Medicine Finland, University of Finland, Helsinki, Finland; Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Tiinamaija Tuomi
- Folkhalsan Research Center, Helsinki, Finland; Institute for Molecular Medicine Finland, University of Finland, Helsinki, Finland; Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Ivana Novak
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Semb
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, München, Germany.
| |
Collapse
|
105
|
Olaniru OE, Kadolsky U, Kannambath S, Vaikkinen H, Fung K, Dhami P, Persaud SJ. Single-cell transcriptomic and spatial landscapes of the developing human pancreas. Cell Metab 2023; 35:184-199.e5. [PMID: 36513063 DOI: 10.1016/j.cmet.2022.11.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/27/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022]
Abstract
Current differentiation protocols have not been successful in reproducibly generating fully functional human beta cells in vitro, partly due to incomplete understanding of human pancreas development. Here, we present detailed transcriptomic analysis of the various cell types of the developing human pancreas, including their spatial gene patterns. We integrated single-cell RNA sequencing with spatial transcriptomics at multiple developmental time points and revealed distinct temporal-spatial gene cascades. Cell trajectory inference identified endocrine progenitor populations and branch-specific genes as the progenitors differentiate toward alpha or beta cells. Spatial differentiation trajectories indicated that Schwann cells are spatially co-located with endocrine progenitors, and cell-cell connectivity analysis predicted that they may interact via L1CAM-EPHB2 signaling. Our integrated approach enabled us to identify heterogeneity and multiple lineage dynamics within the mesenchyme, showing that it contributed to the exocrine acinar cell state. Finally, we have generated an interactive web resource for investigating human pancreas development for the research community.
Collapse
Affiliation(s)
- Oladapo Edward Olaniru
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London SE1 1UL, UK.
| | - Ulrich Kadolsky
- Genomics Research Platform and Single Cell Laboratory, Biomedical Research Centre, Guy's and St. Thomas' NHS Trust, London, UK; Genomics WA, University of Western Australia, Harry Perkins Institute of Medical Research and Telethon Kids Institute QEII Campus, Nedlands, Perth, WA 6009, Australia
| | - Shichina Kannambath
- Genomics Research Platform and Single Cell Laboratory, Biomedical Research Centre, Guy's and St. Thomas' NHS Trust, London, UK
| | - Heli Vaikkinen
- Genomics Research Platform and Single Cell Laboratory, Biomedical Research Centre, Guy's and St. Thomas' NHS Trust, London, UK
| | - Kathy Fung
- Genomics Research Platform and Single Cell Laboratory, Biomedical Research Centre, Guy's and St. Thomas' NHS Trust, London, UK
| | - Pawan Dhami
- Genomics Research Platform and Single Cell Laboratory, Biomedical Research Centre, Guy's and St. Thomas' NHS Trust, London, UK
| | - Shanta J Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
106
|
Merz S, Breunig M, Melzer MK, Heller S, Wiedenmann S, Seufferlein T, Meier M, Krüger J, Mulaw MA, Hohwieler M, Kleger A. Single-cell profiling of GP2-enriched pancreatic progenitors to simultaneously create acinar, ductal, and endocrine organoids. Theranostics 2023; 13:1949-1973. [PMID: 37064874 PMCID: PMC10091881 DOI: 10.7150/thno.78323] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 03/12/2023] [Indexed: 04/18/2023] Open
Abstract
Rationale: Pancreatic lineage specification follows the formation of tripotent pancreatic progenitors (PPs). Current protocols rebuilding PPs in vitro have an endocrine lineage bias and are mostly based on PDX1/NKX6-1 coexpression neglecting other markers decisive for PP heterogeneity and lineage potential. However, true tripotent PPs are of utmost interest to study also exocrine disorders such as pancreatic cancer and to simultaneously generate all three pancreatic lineages from the same ancestor. Methods: Here, we performed a comprehensive compound testing to advance the generation of multipotent progenitors, which were further characterized for their trilineage potential in vitro and in vivo. The heterogeneity and cell-cell communication across the PP subpopulations were analyzed via single-cell transcriptomics. Results: We introduce a novel PP differentiation platform based on a comprehensive compound screening with an advanced design of experiments computing tool to reduce impurities and to increase Glycoprotein-2 expression and subsequent trilineage potential. Superior PP tripotency was proven in vitro by the generation of acinar, endocrine, and ductal cells as well as in vivo upon orthotopic transplantation revealing all three lineages at fetal maturation level. GP2 expression levels at PP stage ascribed varying pancreatic lineage potential. Intermediate and high GP2 levels were superior in generating endocrine and duct-like organoids (PDLO). FACS-based purification of the GP2high PPs allowed the generation of pancreatic acinar-like organoids (PALO) with proper morphology and expression of digestive enzymes. scRNA-seq confirmed multipotent identity, positioned the GP2/PDX1/NKX6-1high population next to human fetal tip and trunk progenitors and identified novel ligand-receptor (LR) interactions in distinct PP subpopulations. LR validation experiments licensed midkine and VEGF signaling to increase markers labelling the single cell clusters with high GP2 expression. Conclusion: In this study, we guide human pluripotent stem cells into multipotent pancreatic progenitors. This common precursor population, which has the ability to mature into acinar, ductal and functional β-cells, serves as a basis for studying developmental processes and deciphering early cancer formation in a cell type-specific context. Using single-cell RNA sequencing and subsequent validation studies, we were able to dissect PP heterogeneity and specific cell-cell communication signals.
Collapse
Affiliation(s)
- Sarah Merz
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Markus Breunig
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Michael Karl Melzer
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | - Sandra Heller
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Sandra Wiedenmann
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Matthias Meier
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jana Krüger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Medhanie A Mulaw
- Central Unit Single Cell Sequencing, Medical Faculty, Ulm University, Ulm, Germany
| | - Meike Hohwieler
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- ✉ Corresponding author: Prof. Dr. Alexander Kleger, Director, Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany. Phone: +49-731-500-44728; Fax: +49-731-500-44612;
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
- ✉ Corresponding author: Prof. Dr. Alexander Kleger, Director, Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany. Phone: +49-731-500-44728; Fax: +49-731-500-44612;
| |
Collapse
|
107
|
Heaton ES, Hu M, Liu T, Hui H, Tan Y, Ye K, Jin S. Extracellular matrix-derived peptide stimulates the generation of endocrine progenitors and islet organoids from iPSCs. J Tissue Eng 2023; 14:20417314231185858. [PMID: 37435573 PMCID: PMC10331343 DOI: 10.1177/20417314231185858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/17/2023] [Indexed: 07/13/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have enormous potential in producing human tissues endlessly. We previously reported that type V collagen (COL5), a pancreatic extracellular matrix protein, promotes islet development and maturation from iPSCs. In this study, we identified a bioactive peptide domain of COL5, WWASKS, through bioinformatic analysis of decellularized pancreatic ECM (dpECM)-derived collagens. RNA-sequencing suggests that WWASKS induces the formation of pancreatic endocrine progenitors while suppressing the development of other types of organs. The expressions of hypoxic genes were significantly downregulated in the endocrine progenitors formed under peptide stimulation. Furthermore, we unveiled an enhancement of iPSC-derived islets' (i-islets) glucose sensitivity under peptide stimulation. These i-islets secrete insulin in a glucose responsive manner. They were comprised of α, β, δ, and γ cells and were assembled into a tissue architecture similar to that of human islets. Mechanistically, the peptide is able to activate the canonical Wnt signaling pathway, permitting the translocation of β-catenin from the cytoplasm to the nucleus for pancreatic progenitor development. Collectively, for the first time, we demonstrated that an ECM-derived peptide dictates iPSC fate toward the generation of endocrine progenitors and subsequent islet organoids.
Collapse
Affiliation(s)
- Emma S Heaton
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY, USA
| | - Ming Hu
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY, USA
| | - Tianzheng Liu
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY, USA
| | - Huang Hui
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY, USA
| | - Yinfei Tan
- Genomics Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Kaiming Ye
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY, USA
- Center of Biomanufacturing for Regenerative Medicine, State University of New York at Binghamton, Binghamton, NY, USA
| | - Sha Jin
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY, USA
- Center of Biomanufacturing for Regenerative Medicine, State University of New York at Binghamton, Binghamton, NY, USA
| |
Collapse
|
108
|
Sackett SD, Kaplan SJ, Mitchell SA, Brown ME, Burrack AL, Grey S, Huangfu D, Odorico J. Genetic Engineering of Immune Evasive Stem Cell-Derived Islets. Transpl Int 2022; 35:10817. [PMID: 36545154 PMCID: PMC9762357 DOI: 10.3389/ti.2022.10817] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Genome editing has the potential to revolutionize many investigative and therapeutic strategies in biology and medicine. In the field of regenerative medicine, one of the leading applications of genome engineering technology is the generation of immune evasive pluripotent stem cell-derived somatic cells for transplantation. In particular, as more functional and therapeutically relevant human pluripotent stem cell-derived islets (SCDI) are produced in many labs and studied in clinical trials, there is keen interest in studying the immunogenicity of these cells and modulating allogeneic and autoimmune immune responses for therapeutic benefit. Significant experimental work has already suggested that elimination of Human Leukocytes Antigen (HLA) expression and overexpression of immunomodulatory genes can impact survival of a variety of pluripotent stem cell-derived somatic cell types. Limited work published to date focuses on stem cell-derived islets and work in a number of labs is ongoing. Rapid progress is occurring in the genome editing of human pluripotent stem cells and their progeny focused on evading destruction by the immune system in transplantation models, and while much research is still needed, there is no doubt the combined technologies of genome editing and stem cell therapy will profoundly impact transplantation medicine in the future.
Collapse
Affiliation(s)
- Sara D. Sackett
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States,*Correspondence: Sara D. Sackett,
| | - Samuel J. Kaplan
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States,Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Samantha A. Mitchell
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Matthew E. Brown
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Adam L. Burrack
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN,Center for Immunology, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Shane Grey
- Immunology Division, Garvan Institute of Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Danwei Huangfu
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jon Odorico
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
109
|
Berney T, Wassmer CH, Lebreton F, Bellofatto K, Fonseca LM, Bignard J, Hanna R, Peloso A, Berishvili E. From islet of Langerhans transplantation to the bioartificial pancreas. Presse Med 2022; 51:104139. [PMID: 36202182 DOI: 10.1016/j.lpm.2022.104139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
Type 1 diabetes is a disease resulting from autoimmune destruction of the insulin-producing beta cells in the pancreas. When type 1 diabetes develops into severe secondary complications, in particular end-stage nephropathy, or life-threatening severe hypoglycemia, the best therapeutic approach is pancreas transplantation, or more recently transplantation of the pancreatic islets of Langerhans. Islet transplantation is a cell therapy procedure, that is minimally invasive and has a low morbidity, but does not display the same rate of functional success as the more invasive pancreas transplantation because of suboptimal engraftment and survival. Another issue is that pancreas or islet transplantation (collectively known as beta cell replacement therapy) is limited by the shortage of organ donors and by the need for lifelong immunosuppression to prevent immune rejection and recurrence of autoimmunity. A bioartificial pancreas is a construct made of functional, insulin-producing tissue, embedded in an anti-inflammatory, immunomodulatory microenvironment and encapsulated in a perm-selective membrane allowing glucose sensing and insulin release, but isolating from attacks by cells of the immune system. A successful bioartificial pancreas would address the issues of engraftment, survival and rejection. Inclusion of unlimited sources of insulin-producing cells, such as xenogeneic porcine islets or stem cell-derived beta cells would further solve the problem of organ shortage. This article reviews the current status of clinical islet transplantation, the strategies aiming at developing a bioartificial pancreas, the clinical trials conducted in the field and the perspectives for further progress.
Collapse
Affiliation(s)
- Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland; Faculty Diabetes Center, University of Geneva School of Medicine, Geneva, Switzerland; Department of Surgery, School of Medicine and Natural Sciences, Ilia State University, Tbilisi, Georgia
| | - Charles H Wassmer
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland
| | - Fanny Lebreton
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Kevin Bellofatto
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Laura Mar Fonseca
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland
| | - Juliette Bignard
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Reine Hanna
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Andrea Peloso
- Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Faculty Diabetes Center, University of Geneva School of Medicine, Geneva, Switzerland; Institute of Medical and Public Health Research, Ilia State University, Tbilisi, Georgia.
| |
Collapse
|
110
|
Olaniru OE, Hook P, Persaud SJ. Using single-cell multi-omics screening of human fetal pancreas to identify novel players in human beta cell development. Diabet Med 2022; 39:e14992. [PMID: 36302085 PMCID: PMC9828353 DOI: 10.1111/dme.14992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/20/2022] [Indexed: 01/18/2023]
Abstract
Islet transplantation from organ donors can considerably improve glucose homeostasis and well-being in individuals with type 1 diabetes, where the beta cells are destroyed by the autoimmune attack, but there are insufficient donor islets to make this a widespread therapy. Strategies are therefore being developed to generate unlimited amounts of insulin-producing beta cells from pluripotent stem cells, with the aim that they will be transplanted to treat diabetes. Whilst much progress has been made in recent years in the directed differentiation of pluripotent stem cells to beta-like cells, essential gaps still exist in generating stem cell-derived beta cells that are fully functional in vitro. This short review provides details of recent multi-'omics' studies of the human fetal pancreas, which are revealing granular information on the various cell types in the developing pancreas. It is anticipated that this fine mapping of the pancreatic cells at single-cell resolution will provide additional insights that can be utilised to reproducibly produce human beta cells in vitro that have the functional characteristics of beta cells within native human islets.
Collapse
Affiliation(s)
- Oladapo E. Olaniru
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Philippa Hook
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Shanta J. Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| |
Collapse
|
111
|
Verhoeff K, Cuesta-Gomez N, Jasra I, Marfil-Garza B, Dadheech N, Shapiro AMJ. Optimizing Generation of Stem Cell-Derived Islet Cells. Stem Cell Rev Rep 2022; 18:2683-2698. [PMID: 35639237 DOI: 10.1007/s12015-022-10391-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2022] [Indexed: 02/06/2023]
Abstract
Islet transplantation is a highly effective treatment for select patients with type 1 diabetes. Unfortunately, current use is limited to those with brittle disease due to donor limitations and immunosuppression requirements. Discovery of factors for induction of pluripotent stem cells from adult somatic cells into a malleable state has reinvigorated the possibility of autologous-based regenerative cell therapies. Similarly, recent progress in allogeneic human embryonic stem cell islet products is showing early success in clinical trials. Describing safe and standardized differentiation protocols with clear pathways to optimize yield and minimize off-target growth is needed to efficiently move the field forward. This review discusses current islet differentiation protocols with a detailed break-down of differentiation stages to guide step-wise controlled generation of functional islet products.
Collapse
Affiliation(s)
- Kevin Verhoeff
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Nerea Cuesta-Gomez
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Ila Jasra
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Braulio Marfil-Garza
- National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, and CHRISTUS-LatAm Hub - Excellence and Innovation Center, Monterrey, Mexico
| | - Nidheesh Dadheech
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.
- 1-002 Li Ka Shing Centre for Health Research Innovation, 112 St. NW & 87 Ave NW, Edmonton, Alberta, T6G 2E1, Canada.
| |
Collapse
|
112
|
Leavens KF, Alvarez-Dominguez JR, Vo LT, Russ HA, Parent AV. Stem cell-based multi-tissue platforms to model human autoimmune diabetes. Mol Metab 2022; 66:101610. [PMID: 36209784 PMCID: PMC9587366 DOI: 10.1016/j.molmet.2022.101610] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/20/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic insulin-producing β cells are specifically destroyed by the immune system. Understanding the initiation and progression of human T1D has been hampered by the lack of appropriate models that can reproduce the complexity and heterogeneity of the disease. The development of platforms combining multiple human pluripotent stem cell (hPSC) derived tissues to model distinct aspects of T1D has the potential to provide critical novel insights into the etiology and pathogenesis of the human disease. SCOPE OF REVIEW In this review, we summarize the state of hPSC differentiation approaches to generate cell types and tissues relevant to T1D, with a particular focus on pancreatic islet cells, T cells, and thymic epithelium. We present current applications as well as limitations of using these hPSC-derived cells for disease modeling and discuss efforts to optimize platforms combining multiple cell types to model human T1D. Finally, we outline remaining challenges and emphasize future improvements needed to accelerate progress in this emerging field of research. MAJOR CONCLUSIONS Recent advances in reprogramming approaches to create patient-specific induced pluripotent stem cell lines (iPSCs), genome engineering technologies to efficiently modify DNA of hPSCs, and protocols to direct their differentiation into mature cell types have empowered the use of stem cell derivatives to accurately model human disease. While challenges remain before complex interactions occurring in human T1D can be modeled with these derivatives, experiments combining hPSC-derived β cells and immune cells are already providing exciting insight into how these cells interact in the context of T1D, supporting the viability of this approach.
Collapse
Affiliation(s)
- Karla F Leavens
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania and Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Juan R Alvarez-Dominguez
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Linda T Vo
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Holger A Russ
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Audrey V Parent
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
113
|
Stem-Cell-Derived β-Like Cells with a Functional PTPN2 Knockout Display Increased Immunogenicity. Cells 2022; 11:cells11233845. [PMID: 36497105 PMCID: PMC9737324 DOI: 10.3390/cells11233845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
Type 1 diabetes is a polygenic disease that results in an autoimmune response directed against insulin-producing beta cells. PTPN2 is a known high-risk type 1 diabetes associated gene expressed in both immune- and pancreatic beta cells, but how genes affect the development of autoimmune diabetes is largely unknown. We employed CRISPR/Cas9 technology to generate a functional knockout of PTPN2 in human pluripotent stem cells (hPSC) followed by differentiating stem-cell-derived beta-like cells (sBC) and detailed phenotypical analyses. The differentiation efficiency of PTPN2 knockout (PTPN2 KO) sBC is comparable to wild-type (WT) control sBC. Global transcriptomics and protein assays revealed the increased expression of HLA Class I molecules in PTPN2 KO sBC at a steady state and upon exposure to proinflammatory culture conditions, indicating a potential for the increased immune recognition of human beta cells upon differential PTPN2 expression. sBC co-culture with autoreactive preproinsulin-reactive T cell transductants confirmed increased immune stimulations by PTPN2 KO sBC compared to WT sBC. Taken together, our results suggest that the dysregulation of PTPN2 expression in human beta cell may prime autoimmune T cell reactivity and thereby contribute to the development of type 1 diabetes.
Collapse
|
114
|
Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022; 164:6836713. [PMID: 36412119 PMCID: PMC9923807 DOI: 10.1210/endocr/bqac193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.
Collapse
Affiliation(s)
- Roy A Goode
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Julia M Hum
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Michael A Kalwat
- Correspondence: Michael A. Kalwat, PhD, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, 1210 Waterway Blvd, Suite 2000, Indianapolis, IN 46202, USA. or
| |
Collapse
|
115
|
Goswami I, de Klerk E, Carnese P, Hebrok M, Healy KE. Multiplexed microfluidic platform for stem-cell derived pancreatic islet β cells. LAB ON A CHIP 2022; 22:4430-4442. [PMID: 36305868 PMCID: PMC9642094 DOI: 10.1039/d2lc00468b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
Stem cell-derived β cells offer an alternative to primary islets for biomedical discoveries as well as a potential surrogate for islet transplantation. The expense and challenge of obtaining and maintaining functional stem cell-derived β cells calls for a need to develop better high-content and high-throughput culture systems. Microphysiological systems (MPS) are promising high-content in vitro platforms, but scaling for high-throughput screening and discoveries remain a challenge. Traditionally, simultaneous multiplexing of liquid handling and cell loading poses a challenge in the design of high-throughput MPS. Furthermore, although MPS for islet β culture/testing have been developed, studies on multi-day culture of stem-cell derived β cells in MPS have been limited. We present a scalable, multiplexed islet β MPS device that incorporates microfluidic gradient generators to parallelize fluid handling for culture and test conditions. We demonstrated the viability and functionality of the stem cell-derived enriched β clusters (eBCs) for a week, as assessed by the ∼2 fold insulin release by the clusters to glucose challenge. To show the scalable multiplexing for drug testing, we demonstrated the loss of stimulation index after long-term exposure to logarithmic concentration range of glybenclamide. The MPS cultured eBCs also confirmed a glycolytic bottleneck as inferred by insulin secretion responses to metabolites methyl succinate and glyceric acid. Thus, we present an innovative culture platform for eBCs with a balance of high-content and high-throughput characteristics.
Collapse
Affiliation(s)
- Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California Berkeley, Berkeley, CA 94720, USA.
- Department of Materials Science and Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| | - Eleonora de Klerk
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Phichitpol Carnese
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Matthias Hebrok
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California Berkeley, Berkeley, CA 94720, USA.
- Department of Materials Science and Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
116
|
Grapin-Botton A, Kim YH. Pancreas organoid models of development and regeneration. Development 2022; 149:278610. [DOI: 10.1242/dev.201004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
ABSTRACT
Organoids have become one of the fastest progressing and applied models in biological and medical research, and various organoids have now been developed for most of the organs of the body. Here, we review the methods developed to generate pancreas organoids in vitro from embryonic, fetal and adult cells, as well as pluripotent stem cells. We discuss how these systems have been used to learn new aspects of pancreas development, regeneration and disease, as well as their limitations and potential for future discoveries.
Collapse
Affiliation(s)
- Anne Grapin-Botton
- Max Planck Institute of Molecular Cell Biology and Genetics 1 , Dresden D-01307 , Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at The University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden 2 , Dresden D-01307 , Germany
- Cluster of Excellence Physics of Life, TU Dresden 3 , 01062 Dresden , Germany
| | - Yung Hae Kim
- Max Planck Institute of Molecular Cell Biology and Genetics 1 , Dresden D-01307 , Germany
| |
Collapse
|
117
|
Jeyagaran A, Lu CE, Zbinden A, Birkenfeld AL, Brucker SY, Layland SL. Type 1 diabetes and engineering enhanced islet transplantation. Adv Drug Deliv Rev 2022; 189:114481. [PMID: 36002043 PMCID: PMC9531713 DOI: 10.1016/j.addr.2022.114481] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 01/24/2023]
Abstract
The development of new therapeutic approaches to treat type 1 diabetes mellitus (T1D) relies on the precise understanding and deciphering of insulin-secreting β-cell biology, as well as the mechanisms responsible for their autoimmune destruction. β-cell or islet transplantation is viewed as a potential long-term therapy for the millions of patients with diabetes. To advance the field of insulin-secreting cell transplantation, two main research areas are currently investigated by the scientific community: (1) the identification of the developmental pathways that drive the differentiation of stem cells into insulin-producing cells, providing an inexhaustible source of cells; and (2) transplantation strategies and engineered transplants to provide protection and enhance the functionality of transplanted cells. In this review, we discuss the biology of pancreatic β-cells, pathology of T1D and current state of β-cell differentiation. We give a comprehensive view and discuss the different possibilities to engineer enhanced insulin-secreting cell/islet transplantation from a translational perspective.
Collapse
Affiliation(s)
- Abiramy Jeyagaran
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University Tübingen, 72770 Reutlingen, Germany
| | - Chuan-En Lu
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Aline Zbinden
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, German Center for Diabetes Research (DZD e.V.), Munich, Germany
| | - Sara Y Brucker
- Department of Women's Health, Eberhard Karls University, 72076 Tübingen, Germany
| | - Shannon L Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; Department of Women's Health, Eberhard Karls University, 72076 Tübingen, Germany.
| |
Collapse
|
118
|
Brianna, Ling APK, Wong YP. Applying stem cell therapy in intractable diseases: a narrative review of decades of progress and challenges. Stem Cell Investig 2022; 9:4. [PMID: 36238449 PMCID: PMC9552054 DOI: 10.21037/sci-2022-021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/09/2022] [Indexed: 08/10/2023]
Abstract
Background and Objective Stem cell therapy (SCT) is one of the vastly researched branches of regenerative medicine as a therapeutic tool to treat incurable diseases. With the use of human stem cells such as embryonic stem cells (ESCs), adult stem cells (ASCs) and induced pluripotent stem cells (iPSCs), stem cell therapy aims to regenerate or repair damaged tissues and congenital defects. As stem cells are able to undergo infinite self-renewal, differentiate into various types of cells and secrete protective paracrine factors, many researchers have investigated the potential of SCT in regenerative medicine. Therefore, this review aims to provide a comprehensive review on the recent application of SCT in various intractable diseases, namely, haematological diseases, neurological diseases, diabetes mellitus, retinal degenerative disorders and COVID-19 infections along with the challenges faced in the clinical translation of SCT. Methods An extensive search was conducted on Google scholar, PubMed and Clinicaltrials.gov using related keywords. Latest articles on stem cell therapy application in selected diseases along with their challenges in clinical applications were selected. Key content and findings In vitro and in vivo studies involving SCT are shown to be safe and efficacious in treating various diseases covered in this review. There are also a number of small-scale clinical trials that validated the positive therapeutic outcomes of SCT. Nevertheless, the effectiveness of SCT are highly variable as some SCT works best in patients with early-stage diseases while in other diseases, SCT is more likely to work in patients in late stages of illnesses. Among the challenges identified in SCT translation are uncertainty in the underlying stem cell mechanism, ethical issues, genetic instability and immune rejection. Conclusions SCT will be a revolutionary treatment in the future that will provide hope to patients with intractable diseases. Therefore, studies ought to be done to ascertain the long-term effects of SCT while addressing the challenges faced in validating SCT for clinical use. Moreover, as there are many studies investigating the safety and efficacy of SCT, future studies should look into elucidating the regenerative and reparative capabilities of stem cells which largely remains unknown.
Collapse
Affiliation(s)
- Brianna
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Ying Pei Wong
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
119
|
Reprogramming—Evolving Path to Functional Surrogate β-Cells. Cells 2022; 11:cells11182813. [PMID: 36139388 PMCID: PMC9496933 DOI: 10.3390/cells11182813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 12/04/2022] Open
Abstract
Numerous cell sources are being explored to replenish functional β-cell mass since the proof-of -concept for cell therapy of diabetes was laid down by transplantation of islets. Many of these cell sources have been shown to possess a degree of plasticity permitting differentiation along new lineages into insulin-secreting β-cells. In this review, we explore emerging reprograming pathways that aim to generate bone fide insulin producing cells. We focus on small molecules and key transcriptional regulators that orchestrate phenotypic conversion and maintenance of engineered cells.
Collapse
|
120
|
Pignatelli C, Campo F, Neroni A, Piemonti L, Citro A. Bioengineering the Vascularized Endocrine Pancreas: A Fine-Tuned Interplay Between Vascularization, Extracellular-Matrix-Based Scaffold Architecture, and Insulin-Producing Cells. Transpl Int 2022; 35:10555. [PMID: 36090775 PMCID: PMC9452644 DOI: 10.3389/ti.2022.10555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022]
Abstract
Intrahepatic islet transplantation is a promising β-cell replacement strategy for the treatment of type 1 diabetes. Instant blood-mediated inflammatory reactions, acute inflammatory storm, and graft revascularization delay limit islet engraftment in the peri-transplant phase, hampering the success rate of the procedure. Growing evidence has demonstrated that islet engraftment efficiency may take advantage of several bioengineering approaches aimed to recreate both vascular and endocrine compartments either ex vivo or in vivo. To this end, endocrine pancreas bioengineering is an emerging field in β-cell replacement, which might provide endocrine cells with all the building blocks (vascularization, ECM composition, or micro/macro-architecture) useful for their successful engraftment and function in vivo. Studies on reshaping either the endocrine cellular composition or the islet microenvironment have been largely performed, focusing on a single building block element, without, however, grasping that their synergistic effect is indispensable for correct endocrine function. Herein, the review focuses on the minimum building blocks that an ideal vascularized endocrine scaffold should have to resemble the endocrine niche architecture, composition, and function to foster functional connections between the vascular and endocrine compartments. Additionally, this review highlights the possibility of designing bioengineered scaffolds integrating alternative endocrine sources to overcome donor organ shortages and the possibility of combining novel immune-preserving strategies for long-term graft function.
Collapse
Affiliation(s)
- Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Campo
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessia Neroni
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
121
|
Pellegrini S, Zamarian V, Sordi V. Strategies to Improve the Safety of iPSC-Derived β Cells for β Cell Replacement in Diabetes. Transpl Int 2022; 35:10575. [PMID: 36090777 PMCID: PMC9448870 DOI: 10.3389/ti.2022.10575] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022]
Abstract
Allogeneic islet transplantation allows for the re-establishment of glycemic control with the possibility of insulin independence, but is severely limited by the scarcity of organ donors. However, a new source of insulin-producing cells could enable the widespread use of cell therapy for diabetes treatment. Recent breakthroughs in stem cell biology, particularly pluripotent stem cell (PSC) techniques, have highlighted the therapeutic potential of stem cells in regenerative medicine. An understanding of the stages that regulate β cell development has led to the establishment of protocols for PSC differentiation into β cells, and PSC-derived β cells are appearing in the first pioneering clinical trials. However, the safety of the final product prior to implantation remains crucial. Although PSC differentiate into functional β cells in vitro, not all cells complete differentiation, and a fraction remain undifferentiated and at risk of teratoma formation upon transplantation. A single case of stem cell-derived tumors may set the field back years. Thus, this review discusses four approaches to increase the safety of PSC-derived β cells: reprogramming of somatic cells into induced PSC, selection of pure differentiated pancreatic cells, depletion of contaminant PSC in the final cell product, and control or destruction of tumorigenic cells with engineered suicide genes.
Collapse
|
122
|
Cuesta-Gomez N, Verhoeff K, Jasra IT, Pawlick R, Dadheech N, Shapiro AMJ. Characterization of stem-cell-derived islets during differentiation and after implantation. Cell Rep 2022; 40:111238. [PMID: 36001981 DOI: 10.1016/j.celrep.2022.111238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/26/2022] [Accepted: 07/27/2022] [Indexed: 12/11/2022] Open
Abstract
Recapitulation of embryonic pancreatic development has enabled development of methods for in vitro islet cell differentiation using human pluripotent stem cells (hPSCs), which have the potential to cure diabetes. Advanced methods for optimal generation of stem-cell-derived islets (SC-islets) has enabled successful diabetes reversal in rodents and shown promising early clinical trial outcomes. The main impediment for use of SC-islets is concern about safety because of off-target growth resulting from contaminated residual cells. In this review, we summarize the different endocrine and non-endocrine cell populations that have been described to emerge throughout β cell differentiation and after transplantation. We discuss the most recent approaches to enrich endocrine populations and remove off-target cells. Finally, we discuss the critical quality control and release criteria testing that we anticipate will be required prior to transplantation to ensure product safety.
Collapse
Affiliation(s)
- Nerea Cuesta-Gomez
- Alberta Diabetes Institute, Department of Surgery, 1-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, 112 St. NW & 87 Ave. NW, Edmonton, AB T6G 2E1, Canada
| | - Kevin Verhoeff
- Alberta Diabetes Institute, Department of Surgery, 1-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, 112 St. NW & 87 Ave. NW, Edmonton, AB T6G 2E1, Canada
| | - Ila Tewari Jasra
- Alberta Diabetes Institute, Department of Surgery, 1-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, 112 St. NW & 87 Ave. NW, Edmonton, AB T6G 2E1, Canada
| | - Rena Pawlick
- Alberta Diabetes Institute, Department of Surgery, 1-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, 112 St. NW & 87 Ave. NW, Edmonton, AB T6G 2E1, Canada
| | - Nidheesh Dadheech
- Alberta Diabetes Institute, Department of Surgery, 1-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, 112 St. NW & 87 Ave. NW, Edmonton, AB T6G 2E1, Canada.
| | - A M James Shapiro
- Alberta Diabetes Institute, Department of Surgery, 1-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, 112 St. NW & 87 Ave. NW, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
123
|
Fantuzzi F, Toivonen S, Schiavo AA, Chae H, Tariq M, Sawatani T, Pachera N, Cai Y, Vinci C, Virgilio E, Ladriere L, Suleiman M, Marchetti P, Jonas JC, Gilon P, Eizirik DL, Igoillo-Esteve M, Cnop M. In depth functional characterization of human induced pluripotent stem cell-derived beta cells in vitro and in vivo. Front Cell Dev Biol 2022; 10:967765. [PMID: 36060810 PMCID: PMC9428245 DOI: 10.3389/fcell.2022.967765] [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: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 01/05/2023] Open
Abstract
In vitro differentiation of human induced pluripotent stem cells (iPSCs) into beta cells represents an important cell source for diabetes research. Here, we fully characterized iPSC-derived beta cell function in vitro and in vivo in humanized mice. Using a 7-stage protocol, human iPSCs were differentiated into islet-like aggregates with a yield of insulin-positive beta cells comparable to that of human islets. The last three stages of differentiation were conducted with two different 3D culture systems, rotating suspension or static microwells. In the latter, homogeneously small-sized islet-like aggregates were obtained, while in rotating suspension size was heterogeneous and aggregates often clumped. In vitro function was assessed by glucose-stimulated insulin secretion, NAD(P)H and calcium fluctuations. Stage 7 aggregates slightly increased insulin release in response to glucose in vitro. Aggregates were transplanted under the kidney capsule of NOD-SCID mice to allow for further in vivo beta cell maturation. In transplanted mice, grafts showed glucose-responsiveness and maintained normoglycemia after streptozotocin injection. In situ kidney perfusion assays showed modulation of human insulin secretion in response to different secretagogues. In conclusion, iPSCs differentiated with equal efficiency into beta cells in microwells compared to rotating suspension, but the former had a higher experimental success rate. In vitro differentiation generated aggregates lacking fully mature beta cell function. In vivo, beta cells acquired the functional characteristics typical of human islets. With this technology an unlimited supply of islet-like organoids can be generated from human iPSCs that will be instrumental to study beta cell biology and dysfunction in diabetes.
Collapse
Affiliation(s)
- Federica Fantuzzi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium,Endocrinology and Metabolism, Department of Medicine and Surgery, University of Parma, Parma, Italy,*Correspondence: Miriam Cnop, ; Federica Fantuzzi,
| | - Sanna Toivonen
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrea Alex Schiavo
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Heeyoung Chae
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Mohammad Tariq
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Toshiaki Sawatani
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Nathalie Pachera
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Ying Cai
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Chiara Vinci
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Enrico Virgilio
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Ladriere
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Jean-Christophe Jonas
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Patrick Gilon
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Décio L. Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium,*Correspondence: Miriam Cnop, ; Federica Fantuzzi,
| |
Collapse
|
124
|
Panova AV, Klementieva NV, Sycheva AV, Korobko EV, Sosnovtseva AO, Krasnova TS, Karpova MR, Rubtsov PM, Tikhonovich YV, Tiulpakov AN, Kiselev SL. Aberrant Splicing of INS Impairs Beta-Cell Differentiation and Proliferation by ER Stress in the Isogenic iPSC Model of Neonatal Diabetes. Int J Mol Sci 2022; 23:8824. [PMID: 35955956 PMCID: PMC9369396 DOI: 10.3390/ijms23158824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 01/09/2023] Open
Abstract
One of the causes of diabetes in infants is the defect of the insulin gene (INS). Gene mutations can lead to proinsulin misfolding, an increased endoplasmic reticulum (ER) stress and possible beta-cell apoptosis. In humans, the mechanisms underlying beta-cell failure remain unclear. We generated induced pluripotent stem cells (iPSCs) from a patient diagnosed with neonatal diabetes mellitus carrying the INS mutation in the 2nd intron (c.188-31G>A) and engineered isogenic CRISPR/Cas9 mutation-corrected cell lines. Differentiation into beta-like cells demonstrated that mutation led to the emergence of an ectopic splice site within the INS and appearance of the abnormal RNA transcript. Isogenic iPSC lines differentiated into beta-like cells showed a clear difference in formation of organoids at pancreatic progenitor stage of differentiation. Moreover, MIN6 insulinoma cell line expressing mutated cDNA demonstrated significant decrease in proliferation capacity and activation of ER stress and unfolded protein response (UPR)-associated genes. These findings shed light on the mechanism underlying the pathogenesis of monogenic diabetes.
Collapse
Affiliation(s)
- Alexandra V. Panova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- Endocrinology Research Centre, 115478 Moscow, Russia
| | - Natalia V. Klementieva
- Endocrinology Research Centre, 115478 Moscow, Russia
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | | | - Elena V. Korobko
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | - Tatiana S. Krasnova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Maria R. Karpova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Petr M. Rubtsov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | | | - Sergey L. Kiselev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- Endocrinology Research Centre, 115478 Moscow, Russia
| |
Collapse
|
125
|
Jin W, Jiang W. Stepwise differentiation of functional pancreatic β cells from human pluripotent stem cells. CELL REGENERATION 2022; 11:24. [PMID: 35909206 PMCID: PMC9339430 DOI: 10.1186/s13619-022-00125-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/13/2022] [Indexed: 12/15/2022]
Abstract
Pancreatic β cells differentiated from stem cells provide promise for cell replacement therapy of diabetes. Human pluripotent stem cells could be differentiated into definitive endoderm, followed by pancreatic progenitors, and then subjected to endocrinal differentiation and maturation in a stepwise fashion. Many achievements have been made in making pancreatic β cells from human pluripotent stem cells in last two decades, and a couple of phase I/II clinical trials have just been initiated. Here, we overview the major progresses in differentiating pancreatic β cells from human pluripotent stem cells with the focus on recent technical advances in each differentiation stage, and briefly discuss the current limitations as well.
Collapse
|
126
|
Emerging molecular technologies for light-mediated modulation of pancreatic beta-cell function. Mol Metab 2022; 64:101552. [PMID: 35863638 PMCID: PMC9352964 DOI: 10.1016/j.molmet.2022.101552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Background Optogenetic modalities as well as optochemical and photopharmacological strategies, collectively termed optical methods, have revolutionized the control of cellular functions via light with great spatiotemporal precision. In comparison to the major advances in the photomodulation of signaling activities noted in neuroscience, similar applications to endocrine cells of the pancreas, particularly insulin-producing β-cells, have been limited. The availability of tools allowing light-mediated changes in the trafficking of ions such as K+ and Ca2+ and signaling intermediates such as cyclic adenosine monophosphate (cAMP), renders β-cells and their glucose-stimulated insulin secretion (GSIS) amenable to optoengineering for drug-free control of blood sugar. Scope of review The molecular circuit of the GSIS in β-cells is described with emphasis on intermediates which are targetable for optical intervention. Various pharmacological agents modifying the release of insulin are reviewed along with their documented side effects. These are contrasted with optical approaches, which have already been employed for engineering β-cell function or are considered for future such applications. Principal obstacles are also discussed as the implementation of optogenetics is pondered for tissue engineering and biology applications of the pancreas. Major Conclusions Notable advances in optogenetic, optochemical and photopharmacological tools are rendering feasible the smart engineering of pancreatic cells and tissues with light-regulated function paving the way for novel solutions for addressing pancreatic pathologies including diabetes.
Collapse
|
127
|
Sim EZ, Enomoto T, Shiraki N, Furuta N, Kashio S, Kambe T, Tsuyama T, Arakawa A, Ozawa H, Yokoyama M, Miura M, Kume S. Methionine metabolism regulates pluripotent stem cell pluripotency and differentiation through zinc mobilization. Cell Rep 2022; 40:111120. [PMID: 35858556 DOI: 10.1016/j.celrep.2022.111120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 04/19/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Pluripotent stem cells (PSCs) exhibit a unique feature that requires S-adenosylmethionine (SAM) for the maintenance of their pluripotency. Methionine deprivation in the medium causes a reduction in intracellular SAM, thus rendering PSCs in a state potentiated for differentiation. In this study, we find that methionine deprivation triggers a reduction in intracellular protein-bound Zn content and upregulation of Zn exporter SLC30A1 in PSCs. Culturing PSCs in Zn-deprived medium results in decreased intracellular protein-bound Zn content, reduced cell growth, and potentiated differentiation, which partially mimics methionine deprivation. PSCs cultured under Zn deprivation exhibit an altered methionine metabolism-related metabolite profile. We conclude that methionine deprivation potentiates differentiation partly by lowering cellular Zn content. We establish a protocol to generate functional pancreatic β cells by applying methionine and Zn deprivation. Our results reveal a link between Zn signaling and methionine metabolism in the regulation of cell fate in PSCs.
Collapse
Affiliation(s)
- Erinn Zixuan Sim
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Takayuki Enomoto
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Nobuaki Shiraki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| | - Nao Furuta
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Soshiro Kashio
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Taiho Kambe
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Tomonori Tsuyama
- Division of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Akihiro Arakawa
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto, Kawasaki-shi, Kanagawa, Japan
| | - Hiroki Ozawa
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Mizuho Yokoyama
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto, Kawasaki-shi, Kanagawa, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shoen Kume
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| |
Collapse
|
128
|
N 6-methyladenosine modification-mediated mRNA metabolism is essential for human pancreatic lineage specification and islet organogenesis. Nat Commun 2022; 13:4148. [PMID: 35851388 PMCID: PMC9293889 DOI: 10.1038/s41467-022-31698-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic differentiation from human pluripotent stem cells (hPSCs) provides promising avenues for investigating development and treating diseases. N6-methyladenosine (m6A) is the most prevalent internal messenger RNA (mRNA) modification and plays pivotal roles in regulation of mRNA metabolism, while its functions remain elusive. Here, we profile the dynamic landscapes of m6A transcriptome-wide during pancreatic differentiation. Next, we generate knockout hPSC lines of the major m6A demethylase ALKBH5, and find that ALKBH5 plays significant regulatory roles in pancreatic organogenesis. Mechanistic studies reveal that ALKBH5 deficiency reduces the mRNA stability of key pancreatic transcription factors in an m6A and YTHDF2-dependent manner. We further identify that ALKBH5 cofactor α-ketoglutarate can be applied to enhance differentiation. Collectively, our findings identify ALKBH5 as an essential regulator of pancreatic differentiation and highlight that m6A modification-mediated mRNA metabolism presents an important layer of regulation during cell-fate specification and holds great potentials for translational applications. Ma et al. profile the dynamic landscape of m6A during pancreatic differentiation, and identify ALKBH5 as an essential m6A regulator supporting pancreatic differentiation, indicating a role for m6A-mediated mRNA metabolism in cell-fate specification.
Collapse
|
129
|
ZnT8 loss-of-function accelerates functional maturation of hESC-derived β cells and resists metabolic stress in diabetes. Nat Commun 2022; 13:4142. [PMID: 35842441 PMCID: PMC9288460 DOI: 10.1038/s41467-022-31829-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 07/04/2022] [Indexed: 12/21/2022] Open
Abstract
Human embryonic stem cell-derived β cells (SC-β cells) hold great promise for treatment of diabetes, yet how to achieve functional maturation and protect them against metabolic stresses such as glucotoxicity and lipotoxicity remains elusive. Our single-cell RNA-seq analysis reveals that ZnT8 loss of function (LOF) accelerates the functional maturation of SC-β cells. As a result, ZnT8 LOF improves glucose-stimulated insulin secretion (GSIS) by releasing the negative feedback of zinc inhibition on insulin secretion. Furthermore, we demonstrate that ZnT8 LOF mutations endow SC-β cells with resistance to lipotoxicity/glucotoxicity-triggered cell death by alleviating endoplasmic reticulum (ER) stress through modulation of zinc levels. Importantly, transplantation of SC-β cells with ZnT8 LOF into mice with preexisting diabetes significantly improves glycemia restoration and glucose tolerance. These findings highlight the beneficial effect of ZnT8 LOF on the functional maturation and survival of SC-β cells that are useful as a potential source for cell replacement therapies. Immature function and fragility hinder application of hESC-derived β cells (SC-β cell) for diabetes cell therapy. Here, the authors identify ZnT8 as a gene editing target to enhance the insulin secretion and cell survival under metabolic stress by abolishing zinc transport in SC-β cells.
Collapse
|
130
|
Silva IBB, Kimura CH, Colantoni VP, Sogayar MC. Stem cells differentiation into insulin-producing cells (IPCs): recent advances and current challenges. Stem Cell Res Ther 2022; 13:309. [PMID: 35840987 PMCID: PMC9284809 DOI: 10.1186/s13287-022-02977-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 06/19/2022] [Indexed: 11/10/2022] Open
Abstract
Type 1 diabetes mellitus (T1D) is a chronic disease characterized by an autoimmune destruction of insulin-producing β-pancreatic cells. Although many advances have been achieved in T1D treatment, current therapy strategies are often unable to maintain perfect control of glycemic levels. Several studies are searching for new and improved methodologies for expansion of β-cell cultures in vitro to increase the supply of these cells for pancreatic islets replacement therapy. A promising approach consists of differentiation of stem cells into insulin-producing cells (IPCs) in sufficient number and functional status to be transplanted. Differentiation protocols have been designed using consecutive cytokines or signaling modulator treatments, at specific dosages, to activate or inhibit the main signaling pathways that control the differentiation of induced pluripotent stem cells (iPSCs) into pancreatic β-cells. Here, we provide an overview of the current approaches and achievements in obtaining stem cell-derived β-cells and the numerous challenges, which still need to be overcome to achieve this goal. Clinical translation of stem cells-derived β-cells for efficient maintenance of long-term euglycemia remains a major issue. Therefore, research efforts have been directed to the final steps of in vitro differentiation, aiming at production of functional and mature β-cells and integration of interdisciplinary fields to generate efficient cell therapy strategies capable of reversing the clinical outcome of T1D.
Collapse
Affiliation(s)
- Isaura Beatriz Borges Silva
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil.,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Camila Harumi Kimura
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil
| | - Vitor Prado Colantoni
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil.,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Mari Cleide Sogayar
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil. .,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
131
|
Lee J, Yoon KH. β cell replacement therapy for the cure of diabetes. J Diabetes Investig 2022; 13:1798-1802. [PMID: 35818819 PMCID: PMC9623521 DOI: 10.1111/jdi.13884] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 11/27/2022] Open
Abstract
Islet transplantation is an important option in the treatment of type 1 diabetes. However, a donor shortage and immunosuppressant‐related complications are the current major hurdles of islet transplantation. In this review, we discuss recent updates on islet transplantation to overcome these current obstacles and we share our perspectives on future β cell replacement therapy.
Collapse
Affiliation(s)
- Joonyub Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Kun-Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
132
|
Szlachcic WJ, Dabrowska A, Milewska A, Ziojla N, Blaszczyk K, Barreto-Duran E, Sanak M, Surmiak M, Owczarek K, Grzanka D, Durzynska J, Pyrc K, Borowiak M. SARS-CoV-2 infects an in vitro model of the human developing pancreas through endocytosis. iScience 2022; 25:104594. [PMID: 35756892 PMCID: PMC9212970 DOI: 10.1016/j.isci.2022.104594] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 03/21/2022] [Accepted: 06/08/2022] [Indexed: 01/08/2023] Open
Abstract
Recent studies showed that SARS-CoV-2 can infect adult human pancreas and trigger pancreatic damage. Here, using human fetal pancreas samples and 3D differentiation of human pluripotent cells into pancreatic endocrine cells, we determined that SARS-CoV-2 receptors ACE2, TMPRSS2, and NRP1 are expressed in precursors of insulin-producing pancreatic β-cells, rendering them permissive to SARS-CoV-2 infection. We also show that SARS-CoV-2 enters and undergoes efficient replication in human multipotent pancreatic and endocrine progenitors in vitro. Moreover, we investigated mechanisms by which SARS-CoV-2 enters pancreatic cells, and found that ACE2 mediates the entry, while NRP1 and TMPRSS2 do not. Surprisingly, we found that in pancreatic progenitors, SARS-CoV-2 enters cells via cathepsin-dependent endocytosis, which is a different route than in respiratory tract. Therefore, pancreatic spheroids might serve as a model to study candidate drugs for endocytosis-mediated viral entry inhibition and to investigate whether SARS-CoV-2 infection may affect pancreas development, possibly causing lifelong health consequences. SARS-CoV-2 receptors are present in human developing pancreas in vivo and in vitro SARS-CoV-2 infects multipotent and endocrine pancreatic progenitors in vitro SARS-CoV-2 enters the progenitors via alternate cathepsin-mediated endocytosis
Collapse
Affiliation(s)
- Wojciech J Szlachcic
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland
| | - Agnieszka Dabrowska
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.,Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Aleksandra Milewska
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.,Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Natalia Ziojla
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland
| | - Katarzyna Blaszczyk
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland
| | - Emilia Barreto-Duran
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Marek Sanak
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Marcin Surmiak
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Katarzyna Owczarek
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland
| | - Julia Durzynska
- Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Krzysztof Pyrc
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland.,Stem Cell and Regenerative Medicine Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
133
|
Karimova MV, Gvazava IG, Vorotelyak EA. Overcoming the Limitations of Stem Cell-Derived Beta Cells. Biomolecules 2022; 12:biom12060810. [PMID: 35740935 PMCID: PMC9221417 DOI: 10.3390/biom12060810] [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: 05/13/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
Great advances in type 1 diabetes (T1D) and type 2 diabetes (T2D) treatment have been made to this day. However, modern diabetes therapy based on insulin injections and cadaveric islets transplantation has many disadvantages. That is why researchers are developing new methods to regenerate the pancreatic hormone-producing cells in vitro. The most promising approach is the generation of stem cell-derived beta cells that could provide an unlimited source of insulin-secreting cells. Recent studies provide methods to produce beta-like cell clusters that display glucose-stimulated insulin secretion—one of the key characteristics of the beta cell. However, in comparison with native beta cells, stem cell-derived beta cells do not undergo full functional maturation. In this paper we review the development and current state of various protocols, consider advantages, and propose ways to improve them. We examine molecular pathways, epigenetic modifications, intracellular components, and the microenvironment as a possible leverage to promote beta cell functional maturation. A possibility to create islet organoids from stem cell-derived components, as well as their encapsulation and further transplantation, is also examined. We try to combine modern research on beta cells and their crosstalk to create a holistic overview of developing insulin-secreting systems.
Collapse
Affiliation(s)
- Mariana V. Karimova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.K.); (I.G.G.)
| | - Inessa G. Gvazava
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.K.); (I.G.G.)
| | - Ekaterina A. Vorotelyak
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.K.); (I.G.G.)
- Department of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence:
| |
Collapse
|
134
|
Abstract
The ability to maintain normoglycaemia, through glucose-sensitive insulin release, is a key aspect of postnatal beta cell function. However, terminally differentiated beta cell identity does not necessarily imply functional maturity. Beta cell maturation is therefore a continuation of beta cell development, albeit a process that occurs postnatally in mammals. Although many important features have been identified in the study of beta cell maturation, as of yet no unified mechanistic model of beta cell functional maturity exists. Here, we review recent findings about the underlying mechanisms of beta cell functional maturation. These findings include systemic hormonal and nutritional triggers that operate through energy-sensing machinery shifts within beta cells, resulting in primed metabolic states that allow for appropriate glucose trafficking and, ultimately, insulin release. We also draw attention to the expansive synergistic nature of these pathways and emphasise that beta cell maturation is dependent on overlapping regulatory and metabolic networks.
Collapse
Affiliation(s)
- Tom Barsby
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| |
Collapse
|
135
|
Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, Pastori RL, Russ HA, Wagner BK, Dominguez-Bendala J. Emerging diabetes therapies: Bringing back the β-cells. Mol Metab 2022; 60:101477. [PMID: 35331962 PMCID: PMC8987999 DOI: 10.1016/j.molmet.2022.101477] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Stem cell therapies are finally coming of age as a viable alternative to pancreatic islet transplantation for the treatment of insulin-dependent diabetes. Several clinical trials using human embryonic stem cell (hESC)-derived β-like cells are currently underway, with encouraging preliminary results. Remaining challenges notwithstanding, these strategies are widely expected to reduce our reliance on human isolated islets for transplantation procedures, making cell therapies available to millions of diabetic patients. At the same time, advances in our understanding of pancreatic cell plasticity and the molecular mechanisms behind β-cell replication and regeneration have spawned a multitude of translational efforts aimed at inducing β-cell replenishment in situ through pharmacological means, thus circumventing the need for transplantation. SCOPE OF REVIEW We discuss here the current state of the art in hESC transplantation, as well as the parallel quest to discover agents capable of either preserving the residual mass of β-cells or inducing their proliferation, transdifferentiation or differentiation from progenitor cells. MAJOR CONCLUSIONS Stem cell-based replacement therapies in the mold of islet transplantation are already around the corner, but a permanent cure for type 1 diabetes will likely require the endogenous regeneration of β-cells aided by interventions to restore the immune balance. The promise of current research avenues and a strong pipeline of clinical trials designed to tackle these challenges bode well for the realization of this goal.
Collapse
Affiliation(s)
- G Basile
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - M M F Qadir
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - F Mauvais-Jarvis
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - A Vetere
- Broad Institute, Cambridge, MA, USA
| | - V Shoba
- Broad Institute, Cambridge, MA, USA
| | | | - R L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H A Russ
- Barbara Davis Center for Diabetes, Colorado University Anschutz Medical Campus, Aurora, CO, USA.
| | | | - J Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
136
|
Abstract
The pancreatic β-cells are essential for regulating glucose homeostasis through the coordinated release of the insulin hormone. Dysfunction of the highly specialized β-cells results in diabetes mellitus, a growing global health epidemic. In this review, we describe the development and function of β-cells the emerging concept of heterogeneity within insulin-producing cells, and the potential of other cell types to assume β-cell functionality via transdifferentiation. We also discuss emerging routes to design cells with minimal β-cell properties and human stem cell differentiation efforts that carry the promise to restore normoglycemia in patients suffering from diabetes.
Collapse
Affiliation(s)
- Natanya Kerper
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Sudipta Ashe
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
137
|
Yoshihara E. Adapting Physiology in Functional Human Islet Organogenesis. Front Cell Dev Biol 2022; 10:854604. [PMID: 35557947 PMCID: PMC9086403 DOI: 10.3389/fcell.2022.854604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023] Open
Abstract
Generation of three-dimensional (3D)-structured functional human islets is expected to be an alternative cell source for cadaveric human islet transplantation for the treatment of insulin-dependent diabetes. Human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer infinite resources for newly synthesized human islets. Recent advancements in hPSCs technology have enabled direct differentiation to human islet-like clusters, which can sense glucose and secrete insulin, and those islet clusters can ameliorate diabetes when transplanted into rodents or non-human primates (NHPs). However, the generated hPSC-derived human islet-like clusters are functionally immature compared with primary human islets. There remains a challenge to establish a technology to create fully functional human islets in vitro, which are functionally and transcriptionally indistinguishable from cadaveric human islets. Understanding the complex differentiation and maturation pathway is necessary to generate fully functional human islets for a tremendous supply of high-quality human islets with less batch-to-batch difference for millions of patients. In this review, I summarized the current progress in the generation of 3D-structured human islets from pluripotent stem cells and discussed the importance of adapting physiology for in vitro functional human islet organogenesis and possible improvements with environmental cues.
Collapse
Affiliation(s)
- Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
138
|
Abstract
Embryoids and organoids hold great promise for human biology and medicine. Herein, we discuss conceptual and technological frameworks useful for developing high-fidelity embryoids and organoids that display tissue- and organ-level phenotypes and functions, which are critically needed for decoding developmental programs and improving translational applications. Through dissecting the layers of inputs controlling mammalian embryogenesis, we review recent progress in reconstructing multiscale structural orders in embryoids and organoids. Bioengineering tools useful for multiscale, multimodal structural engineering of tissue- and organ-level cellular organization and microenvironment are also discussed to present integrative, bioengineering-directed approaches to achieve next-generation, high-fidelity embryoids and organoids.
Collapse
Affiliation(s)
- Yue Shao
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China; State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
139
|
Stem Cell-Derived Islets for Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23095099. [PMID: 35563490 PMCID: PMC9105352 DOI: 10.3390/ijms23095099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Since the discovery of insulin a century ago, insulin injection has been a primary treatment for both type 1 (T1D) and type 2 diabetes (T2D). T2D is a complicated disea se that is triggered by the dysfunction of insulin-producing β cells and insulin resistance in peripheral tissues. Insulin injection partially compensates for the role of endogenous insulin which promotes glucose uptake, lipid synthesis and organ growth. However, lacking the continuous, rapid, and accurate glucose regulation by endogenous functional β cells, the current insulin injection therapy is unable to treat the root causes of the disease. Thus, new technologies such as human pluripotent stem cell (hPSC)-derived islets are needed for both identifying the key molecular and genetic causes of T2D and for achieving a long-term treatment. This perspective review will provide insight into the efficacy of hPSC-derived human islets for treating and understanding T2D. We discuss the evidence that β cells should be the primary target for T2D treatment, the use of stem cells for the modeling of T2D and the potential use of hPSC-derived islet transplantation for treating T2D.
Collapse
|
140
|
Patel SN, Mathews CE, Chandler R, Stabler CL. The Foundation for Engineering a Pancreatic Islet Niche. Front Endocrinol (Lausanne) 2022; 13:881525. [PMID: 35600597 PMCID: PMC9114707 DOI: 10.3389/fendo.2022.881525] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/30/2022] [Indexed: 12/01/2022] Open
Abstract
Progress in diabetes research is hindered, in part, by deficiencies in current experimental systems to accurately model human pathophysiology and/or predict clinical outcomes. Engineering human-centric platforms that more closely mimic in vivo physiology, however, requires thoughtful and informed design. Summarizing our contemporary understanding of the unique and critical features of the pancreatic islet can inform engineering design criteria. Furthermore, a broad understanding of conventional experimental practices and their current advantages and limitations ensures that new models address key gaps. Improving beyond traditional cell culture, emerging platforms are combining diabetes-relevant cells within three-dimensional niches containing dynamic matrices and controlled fluidic flow. While highly promising, islet-on-a-chip prototypes must evolve their utility, adaptability, and adoptability to ensure broad and reproducible use. Here we propose a roadmap for engineers to craft biorelevant and accessible diabetes models. Concurrently, we seek to inspire biologists to leverage such tools to ask complex and nuanced questions. The progenies of such diabetes models should ultimately enable investigators to translate ambitious research expeditions from benchtop to the clinic.
Collapse
Affiliation(s)
- Smit N. Patel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Rachel Chandler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Cherie L. Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
141
|
Sasaki S, Lee MYY, Wakabayashi Y, Suzuki L, Winata H, Himuro M, Matsuoka TA, Shimomura I, Watada H, Lynn FC, Miyatsuka T. Spatial and transcriptional heterogeneity of pancreatic beta cell neogenesis revealed by a time-resolved reporter system. Diabetologia 2022; 65:811-828. [PMID: 35243521 DOI: 10.1007/s00125-022-05662-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/14/2021] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS While pancreatic beta cells have been shown to originate from endocrine progenitors in ductal regions, it remains unclear precisely where beta cells emerge from and which transcripts define newborn beta cells. We therefore investigated characteristics of newborn beta cells extracted by a time-resolved reporter system. METHODS We established a mouse model, 'Ins1-GFP; Timer', which provides spatial information during beta cell neogenesis with high temporal resolution. Single-cell RNA-sequencing (scRNA-seq) was performed on mouse beta cells sorted by fluorescent reporter to uncover transcriptomic profiles of newborn beta cells. scRNA-seq of human embryonic stem cell (hESC)-derived beta-like cells was also performed to compare newborn beta cell features between mouse and human. RESULTS Fluorescence imaging of Ins1-GFP; Timer mouse pancreas successfully dissected newly generated beta cells as green fluorescence-dominant cells. This reporter system revealed that, as expected, some newborn beta cells arise close to the ducts (βduct); unexpectedly, the others arise away from the ducts and adjacent to blood vessels (βvessel). Single-cell transcriptomic analyses demonstrated five distinct populations among newborn beta cells, confirming spatial heterogeneity of beta cell neogenesis such as high probability of glucagon-positive βduct, musculoaponeurotic fibrosarcoma oncogene family B (MafB)-positive βduct and musculoaponeurotic fibrosarcoma oncogene family A (MafA)-positive βvessel cells. Comparative analysis with scRNA-seq data of mouse newborn beta cells and hESC-derived beta-like cells uncovered transcriptional similarity between mouse and human beta cell neogenesis including microsomal glutathione S-transferase 1 (MGST1)- and synaptotagmin 13 (SYT13)-highly-expressing state. CONCLUSIONS/INTERPRETATION The combination of time-resolved histological imaging with single-cell transcriptional mapping demonstrated novel features of spatial and transcriptional heterogeneity in beta cell neogenesis, which will lead to a better understanding of beta cell differentiation for future cell therapy. DATA AVAILABILITY Raw and processed single-cell RNA-sequencing data for this study has been deposited in the Gene Expression Omnibus under accession number GSE155742.
Collapse
Affiliation(s)
- Shugo Sasaki
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.
- Department of Diabetes Care Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Department of Surgery, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| | - Michelle Y Y Lee
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Yuka Wakabayashi
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Luka Suzuki
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Helena Winata
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Miwa Himuro
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Taka-Aki Matsuoka
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Department of Surgery, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| | - Takeshi Miyatsuka
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Endocrinology, Diabetes and Metabolism, Kitasato University School of Medicine, Kanagawa, Japan.
| |
Collapse
|
142
|
Flisher MF, Shin D, Huising MO. Urocortin3: Local inducer of somatostatin release and bellwether of beta cell maturity. Peptides 2022; 151:170748. [PMID: 35065098 PMCID: PMC10881066 DOI: 10.1016/j.peptides.2022.170748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/03/2022] [Accepted: 01/17/2022] [Indexed: 11/25/2022]
Abstract
Urocortin 3 (UCN3) is a peptide hormone expressed in pancreatic islets of Langerhans of both human alpha and human beta cells and solely in murine beta cells. UCN3 signaling acts locally within the islet to activate its cognate receptor, corticotropin releasing hormone receptor 2 (CRHR2), which is expressed by delta cells, to potentiate somatostatin (SST) negative feedback to reduce islet cell hormone output. The functional importance of UCN3 signaling in the islet is to modulate the amount of SST tone allowing for finely tuned regulation of insulin and glucagon secretion. UCN3 signaling is a hallmark of functional beta cell maturation, increasing the beta cell glucose threshold for insulin secretion. In doing so, UCN3 plays a relevant functional role in accurately maintaining blood glucose homeostasis. Additionally, UCN3 acts as an indicator of beta cell maturation and health, as UCN3 is not expressed in immature beta cells and is downregulated in dedifferentiated and dysfunctional beta cell states. Here, we review the mechanistic underpinnings of UCN3 signaling, its net effect on islet cell hormone output, as well as its value as a marker for beta cell maturation and functional status.
Collapse
Affiliation(s)
- Marcus F Flisher
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, CA, United States
| | - Donghan Shin
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, CA, United States
| | - Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, CA, United States; Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, United States.
| |
Collapse
|
143
|
Carvalho AM, Nunes R, Sarmento B. From pluripotent stem cells to bioengineered islets: A challenging journey to diabetes treatment. Eur J Pharm Sci 2022; 172:106148. [DOI: 10.1016/j.ejps.2022.106148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/27/2022]
|
144
|
Tan LS, Chen JT, Lim LY, Teo AKK. Manufacturing clinical-grade human induced pluripotent stem cell-derived beta cells for diabetes treatment. Cell Prolif 2022; 55:e13232. [PMID: 35474596 PMCID: PMC9357357 DOI: 10.1111/cpr.13232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/26/2022] [Accepted: 03/28/2022] [Indexed: 12/25/2022] Open
Abstract
The unlimited proliferative capacity of human pluripotent stem cells (hPSCs) fortifies it as one of the most attractive sources for cell therapy application in diabetes. In the past two decades, vast research efforts have been invested in developing strategies to differentiate hPSCs into clinically suitable insulin‐producing endocrine cells or functional beta cells (β cells). With the end goal being clinical translation, it is critical for hPSCs and insulin‐producing β cells to be derived, handled, stored, maintained and expanded with clinical compliance. This review focuses on the key processes and guidelines for clinical translation of human induced pluripotent stem cell (hiPSC)‐derived β cells for diabetes cell therapy. Here, we discuss the (1) key considerations of manufacturing clinical‐grade hiPSCs, (2) scale‐up and differentiation of clinical‐grade hiPSCs into β cells in clinically compliant conditions and (3) mandatory quality control and product release criteria necessitated by various regulatory bodies to approve the use of the cell‐based products.
Collapse
Affiliation(s)
- Lay Shuen Tan
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Precision Medicine Translational Research Programme (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juin Ting Chen
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Precision Medicine Translational Research Programme (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lillian Yuxian Lim
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Precision Medicine Translational Research Programme (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
145
|
Generation of Isogenic hiPSCs with Targeted Edits at Multiple Intronic SNPs to Study the Effects of the Type 2 Diabetes Associated KCNQ1 Locus in American Indians. Cells 2022; 11:cells11091446. [PMID: 35563754 PMCID: PMC9102014 DOI: 10.3390/cells11091446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
The top genetic association signal for type 2 diabetes (T2D) in Southwestern American Indians maps to intron 15 of KCNQ1, an imprinted gene. We aim to understand the biology whereby variation at this locus affects T2D specifically in this genomic background. To do so, we obtained human induced pluripotent stem cells (hiPSC) derived from American Indians. Using these iPSCs, we show that imprinting of KCNQ1 and CDKN1C during pancreatic islet-like cell generation from iPSCs is consistent with known imprinting patterns in fetal pancreas and adult islets and therefore is an ideal model system to study this locus. In this report, we detail the use of allele-specific guide RNAs and CRISPR to generate isogenic hiPSCs that differ only at multiple T2D associated intronic SNPs at this locus which can be used to elucidate their functional effects. Characterization of these isogenic hiPSCs identified a few aberrant cell lines; namely cell lines with large hemizygous deletions in the putative functional region of KCNQ1 and cell lines hypomethylated at the KCNQ1OT1 promoter. Comparison of an isogenic cell line with a hemizygous deletion to the parental cell line identified CDKN1C and H19 as differentially expressed during the endocrine progenitor stage of pancreatic-islet development.
Collapse
|
146
|
Parent AV, Ashe S, Nair GG, Li ML, Chavez J, Liu JS, Zhong Y, Streeter PR, Hebrok M. Development of a scalable method to isolate subsets of stem cell-derived pancreatic islet cells. Stem Cell Reports 2022; 17:979-992. [PMID: 35245441 PMCID: PMC9023773 DOI: 10.1016/j.stemcr.2022.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/24/2022] Open
Abstract
Cell replacement therapy using β cells derived from stem cells is a promising alternative to conventional diabetes treatment options. Although current differentiation methods produce glucose-responsive β cells, they can also yield populations of undesired endocrine progenitors and other proliferating cell types that might interfere with long-term islet function and safety of transplanted cells. Here, we describe the generation of an array of monoclonal antibodies against cell surface markers that selectively label stem cell-derived islet cells. A high-throughput screen identified promising candidates, including three clones that mark a high proportion of endocrine cells in differentiated cultures. A scalable magnetic sorting method was developed to enrich for human pluripotent stem cell (hPSC)-derived islet cells using these three antibodies, leading to the formation of islet-like clusters with improved glucose-stimulated insulin secretion and reduced growth upon transplantation. This strategy should facilitate large-scale production of functional islet clusters from stem cells for disease modeling and cell replacement therapy.
Collapse
Affiliation(s)
- Audrey V Parent
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Sudipta Ashe
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gopika G Nair
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mei-Lan Li
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jessica Chavez
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jennifer S Liu
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yongping Zhong
- Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR 97239, USA
| | - Philip R Streeter
- Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR 97239, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
147
|
Chmielowiec J, Szlachcic WJ, Yang D, Scavuzzo MA, Wamble K, Sarrion-Perdigones A, Sabek OM, Venken KJT, Borowiak M. Human pancreatic microenvironment promotes β-cell differentiation via non-canonical WNT5A/JNK and BMP signaling. Nat Commun 2022; 13:1952. [PMID: 35414140 PMCID: PMC9005503 DOI: 10.1038/s41467-022-29646-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 03/21/2022] [Indexed: 12/24/2022] Open
Abstract
In vitro derivation of pancreatic β-cells from human pluripotent stem cells holds promise as diabetes treatment. Despite recent progress, efforts to generate physiologically competent β-cells are still hindered by incomplete understanding of the microenvironment's role in β-cell development and maturation. Here, we analyze the human mesenchymal and endothelial primary cells from weeks 9-20 fetal pancreas and identify a time point-specific microenvironment that permits β-cell differentiation. Further, we uncover unique factors that guide in vitro development of endocrine progenitors, with WNT5A markedly improving human β-cell differentiation. WNT5A initially acts through the non-canonical (JNK/c-JUN) WNT signaling and cooperates with Gremlin1 to inhibit the BMP pathway during β-cell maturation. Interestingly, we also identify the endothelial-derived Endocan as a SST+ cell promoting factor. Overall, our study shows that the pancreatic microenvironment-derived factors can mimic in vivo conditions in an in vitro system to generate bona fide β-cells for translational applications.
Collapse
Affiliation(s)
- Jolanta Chmielowiec
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wojciech J Szlachcic
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, ul. Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Diane Yang
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marissa A Scavuzzo
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Katrina Wamble
- Stem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alejandro Sarrion-Perdigones
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Omaima M Sabek
- Department of Surgery, The Methodist Hospital, Houston, TX, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Koen J T Venken
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,McNair Medical Institute, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Malgorzata Borowiak
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX, 77030, USA. .,Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, ul. Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland. .,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Stem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,McNair Medical Institute, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
148
|
Carrasco M, Wang C, Søviknes AM, Bjørlykke Y, Abadpour S, Paulo JA, Tjora E, Njølstad P, Ghabayen J, Nermoen I, Lyssenko V, Chera S, Ghila LM, Vaudel M, Scholz H, Ræder H. Spatial Environment Affects HNF4A Mutation-Specific Proteome Signatures and Cellular Morphology in hiPSC-Derived β-Like Cells. Diabetes 2022; 71:862-869. [PMID: 35043148 PMCID: PMC8965667 DOI: 10.2337/db20-1279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/03/2022] [Indexed: 11/13/2022]
Abstract
Studies of monogenic diabetes are particularly useful because we can gain insight into the molecular events of pancreatic β-cell failure. Maturity-onset diabetes of the young 1 (MODY1) is a form of monogenic diabetes caused by a mutation in the HNF4A gene. Human-induced pluripotent stem cells (hiPSCs) provide an excellent tool for disease modeling by subsequently directing differentiation toward desired pancreatic islet cells, but cellular phenotypes in terminally differentiated cells are notoriously difficult to detect. Re-creating a spatial (three-dimensional [3D]) environment may facilitate phenotype detection. We studied MODY1 by using hiPSC-derived pancreatic β-like patient and isogenic control cell lines in two different 3D contexts. Using size-adjusted cell aggregates and alginate capsules, we show that the 3D context is critical to facilitating the detection of mutation-specific phenotypes. In 3D cell aggregates, we identified irregular cell clusters and lower levels of structural proteins by proteome analysis, whereas in 3D alginate capsules, we identified altered levels of glycolytic proteins in the glucose sensing apparatus by proteome analysis. Our study provides novel knowledge on normal and abnormal function of HNF4A, paving the way for translational studies of new drug targets that can be used in precision diabetes medicine in MODY.
Collapse
Affiliation(s)
- Manuel Carrasco
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Cancer Biomarkers, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Chencheng Wang
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub–Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Anne M. Søviknes
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Yngvild Bjørlykke
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Shadab Abadpour
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub–Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Erling Tjora
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Pål Njølstad
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Jonas Ghabayen
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ingrid Nermoen
- Department of Endocrinology, Akershus University Hospital, Lorenskog, Norway
| | - Valeriya Lyssenko
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simona Chera
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Luiza M. Ghila
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marc Vaudel
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hanne Scholz
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub–Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Helge Ræder
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
- Corresponding author: Helge Ræder,
| |
Collapse
|
149
|
Mitofusin2 Promotes β Cell Maturation from Mouse Embryonic Stem Cells via Sirt3/Idh2 Activation. Stem Cells Int 2022; 2022:1172795. [PMID: 35386849 PMCID: PMC8977338 DOI: 10.1155/2022/1172795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 03/02/2022] [Indexed: 12/01/2022] Open
Abstract
β cell dysfunction is the leading cause of diabetes. Adult β cells have matured glucose-stimulated insulin secretion (GSIS), whereas fetal and neonatal β cells are insensitive to glucose and are functionally immature. However, how β cells mature and acquire robust GSIS is not fully understood. Here, we explored the potential regulatory proteins of β cell maturation process and the capacity for GSIS. Combined with the data from public databases, we found that the gene expression of Mitofusin2 (Mfn2) showed an increasing trend from mouse neonatal β cells to mature β cells. Moreover, its protein expression increased during mouse embryonic pancreas development and β cell differentiation from mouse embryonic stem cells. Knocking down Mfn2 reduced Urocortin3 (Ucn3) expression, GSIS, and ATP production in induced β cells, while overexpressing it had the opposite effect. However, neither Mfn2 knockdown nor overexpression affected the differentiation rate of insulin-positive cells. In immature and mature β cells, Mfn2 and its correlated genes were enriched in tricarboxylic acid (TCA) cycle-related pathways. The expressions of Sirtuin 3 (Sirt3) and isocitrate dehydrogenase 2 (NADP+) and mitochondrial (Idh2) were Mfn2-regulated during β cell differentiation. Inhibiting Idh2 or Sirt3 reduced cellular ATP content and insulin secretion levels that increased by Mfn2 overexpression. Thus, Mfn2 modulated the induced β cell GSIS by influencing the TCA cycle through Sirt3/Idh2 activation. We demonstrated that Mfn2 promoted embryonic stem cell-derived β cell maturation via the Sirt3/Idh2 pathway, providing new insights into β cell development. Our data contribute to understanding diabetes pathogenesis and offer potential new targets for β cell regeneration therapies.
Collapse
|
150
|
Zambom AZ, Collazos JA, Dias R. Selection of the number of clusters in functional data analysis. J STAT COMPUT SIM 2022. [DOI: 10.1080/00949655.2022.2053855] [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]
Affiliation(s)
- Adriano Zanin Zambom
- Dept. of Mathematics, California State University Northridge, Northridge, CA, USA
| | | | - Ronaldo Dias
- Dept. of Stat., State University of Campinas (UNICAMP), São Paulo, Brasil
| |
Collapse
|