1
|
Li Y, He C, Ahamed Younis D, Ni C, Liu R, Sun Z, Lin H, Wang Y, Zhu P, Xiao Z, Sun B. Engineered promoter-free insulin-secreting cells provide closed-loop glycemic control. Life Sci 2025; 371:123587. [PMID: 40147530 DOI: 10.1016/j.lfs.2025.123587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Diabetes mellitus is currently a priority health issue worldwide, but existing therapies suffer from insufficient donors, inability to provide glucose-dependent endogenous insulin secretion, transplantation risks, and immune rejection. Especially, reported engineered cells are mostly promoter-induced glucose-independent insulin producing cells. Here we constructed a closed-loop of insulin secretion with glucose-dependent IRES to achieve glucose-sensitive endogenous insulin secretion. Those cells successfully reversed hyperglycemia in diabetic mice for at least 60 days after transplantation without any significant immune rejection, demonstrating that our constructed engineered cellular grafts have good biocompatibility. Our findings hold great promise in the field of diabetes treatment and provide a new, glucose-dependent genetic engineering approach to insulin production, which is expected to solve many of the current problems faced in the clinical treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Yumin Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Cong He
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Key Laboratory of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China.
| | - Doulathunnisa Ahamed Younis
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Chengming Ni
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, Jiangsu 210008, China
| | - Rui Liu
- Department of Genetic Engineering, College of Natural Science, University of Suwon, Kyunggi-Do 445-743, Republic of Korea.
| | - Zilin Sun
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, Jiangsu 210008, China
| | - Hao Lin
- Department of Clinical Science and Research, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Yuxin Wang
- Key Laboratory of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China
| | - Pengyu Zhu
- Key Laboratory of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China
| | - Zhongdang Xiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Bo Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| |
Collapse
|
2
|
Gilglioni EH, Bansal M, St-Pierre-Wijckmans W, Talamantes S, Kasarinaite A, Hay DC, Gurzov EN. Therapeutic potential of stem cell-derived somatic cells to treat metabolic dysfunction-associated steatotic liver disease and diabetes. Obes Rev 2025; 26:e13899. [PMID: 39861937 DOI: 10.1111/obr.13899] [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: 03/25/2024] [Revised: 10/22/2024] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Abstract
Developments in basic stem cell biology have paved the way for technology translation in human medicine. An exciting prospective use of stem cells is the ex vivo generation of hepatic and pancreatic endocrine cells for biomedical applications. This includes creating novel models 'in a dish' and developing therapeutic strategies for complex diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD) and diabetes. In this review, we explore recent advances in the generation of stem cell-derived hepatocyte-like cells and insulin-producing β-like cells. We cover the different differentiation strategies, new discoveries, and the caveats that still exist regarding their routine use. Finally, we discuss the challenges and limitations of stem cell-derived therapies as a clinical strategy to manage metabolic diseases in humans.
Collapse
Affiliation(s)
- Eduardo H Gilglioni
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Mayank Bansal
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | | | - Stephanie Talamantes
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Alvile Kasarinaite
- Institute for Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - David C Hay
- Institute for Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
3
|
Wang Y, Chen Y, McGarrigle J, Cook J, Rios PD, La Monica G, Wei W, Oberholzer J. Cell Therapy for T1D Beyond BLA: Gearing Up Toward Clinical Practice. Diabetes Ther 2025; 16:1125-1138. [PMID: 40214896 PMCID: PMC12085407 DOI: 10.1007/s13300-025-01732-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/18/2025] [Indexed: 05/18/2025] Open
Abstract
Type 1 diabetes (T1D) remains a significant global health challenge and patients with T1D need lifelong insulin therapy. Islet transplantation holds transformative potential by replacing autoimmune-mediated destruction of insulin-producing beta cells. This review examines the trajectory of islet transplantation for T1D, focusing on the process and benefits of obtaining biologics license application (BLA) approval for cell-based therapies. Following US Food and Drug Administration (FDA) approval, the authors identify key steps urgently needed to foster islet transplantation as a viable treatment for a broader population of patients with T1D. Furthermore, the authors highlight recent advances in encapsulation technologies, stem cell-derived islets, xenogeneic islets, and gene editing as strategies to overcome challenges such as immune rejection and limited islet sources. These innovations are pivotal in enhancing the safety and efficacy of islet transplantation. Ultimately, this review emphasizes that while BLA approval represents a critical milestone, realizing the full potential of cell therapy for T1D requires addressing the scientific, clinical, and logistical challenges of its real-world implementation. By fostering innovation, collaboration, and strategic partnerships, the field can transform T1D care, offering patients a durable, life-changing alternative to traditional insulin therapy.
Collapse
Affiliation(s)
- Yong Wang
- CellTrans, Inc., 2201 W. Campbell Park Dr, Chicago, IL, 60612, USA.
- University of Zürich Hospital, Ramistrasse 100, 8991, Zürich, Switzerland.
- Visceral and Transplant Department, University of Zürich Hospital, Ramistrasse 100, 8991, Zürich, Switzerland.
| | - YingYing Chen
- University of Zürich Hospital, Ramistrasse 100, 8991, Zürich, Switzerland
| | - James McGarrigle
- CellTrans, Inc., 2201 W. Campbell Park Dr, Chicago, IL, 60612, USA
| | - Jenny Cook
- CellTrans, Inc., 2201 W. Campbell Park Dr, Chicago, IL, 60612, USA
| | - Peter D Rios
- CellTrans, Inc., 2201 W. Campbell Park Dr, Chicago, IL, 60612, USA
| | | | - Wei Wei
- University of Zürich Hospital, Ramistrasse 100, 8991, Zürich, Switzerland
| | - Jose Oberholzer
- CellTrans, Inc., 2201 W. Campbell Park Dr, Chicago, IL, 60612, USA.
- University of Zürich Hospital, Ramistrasse 100, 8991, Zürich, Switzerland.
- Visceral and Transplant Department, University of Zürich Hospital, Ramistrasse 100, 8991, Zürich, Switzerland.
| |
Collapse
|
4
|
Dadheech N, Bermúdez de León M, Czarnecka Z, Cuesta-Gomez N, Jasra IT, Pawlick R, Marfil-Garza B, Sapkota S, Verhoeff K, Razavy H, Anwar P, Singh A, Ray N, O' Gorman D, Jickling G, Lyon J, MacDonald P, Shapiro AMJ. Scale up manufacturing approach for production of human induced pluripotent stem cell-derived islets using Vertical Wheel® bioreactors. NPJ Regen Med 2025; 10:24. [PMID: 40442082 PMCID: PMC12122946 DOI: 10.1038/s41536-025-00409-y] [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: 08/22/2024] [Accepted: 04/11/2025] [Indexed: 06/02/2025] Open
Abstract
Advanced protocols show potential for human stem cells (SC)-derived islets generation under planar (2D) alone or three-dimensional (3D) cultures, but show challenges in scalability, cell loss, and batch-to-batch consistency. This study explores Vertical Wheel (VW)® bioreactor suspension technology to differentiate islets from human induced pluripotent stem cells, achieving uniform, transcriptionally mature, and functional SC-islets. A 5x increase in scale from 0.1 L to 0.5 L reactors resulted in a 12-fold (15,005-183,002) increase in islet equivalent count (IEQ) without compromising islet structure. SC-islets show enriched β-cell composition (~63% CPPT+NKX6.1+ISL1+), glucose responsive insulin release (3.9-6.1-fold increase), and reversed diabetes in STZ-treated mice. Single cell RNA sequencing and flowcytometry analysis confirmed transcriptional maturity and functional identity, similar to adult islets. Lastly, harvested SC-islet grafts demonstrate improved islet functionality and mature transcriptomic signatures. Overall, scale-up in VW® bioreactor technology enhances IEQ yield with minimal variability and reduced cell loss, offering a pathway for clinical-grade SC-islet production.
Collapse
Affiliation(s)
- Nidheesh Dadheech
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada.
| | - Mario Bermúdez de León
- Centro de Investigación Biomédica del Noreste, Departamento de Biología Molecular, Instituto Mexicano del Seguro Social, Monterrey, Nuevo León, Mexico
| | - Zofia Czarnecka
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nerea Cuesta-Gomez
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Ila Tewari Jasra
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Rena Pawlick
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Braulio Marfil-Garza
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- CHRISTUS-LatAm Hub-Excellence and Innovation Center, Monterrey, Mexico
| | - Sandhya Sapkota
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Kevin Verhoeff
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Haide Razavy
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Perveen Anwar
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Abhineet Singh
- Department of Computing Science, University of Alberta, Edmonton, AB, Canada
| | - Nilanjan Ray
- Department of Computing Science, University of Alberta, Edmonton, AB, Canada
| | - Doug O' Gorman
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Glen Jickling
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - James Lyon
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Patrick MacDonald
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - A M James Shapiro
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
5
|
Wang X, Brielle S, Kenty-Ryu J, Korover N, Bavli D, Pop R, Melton DA. Improving cellular fitness of human stem cell-derived islets under hypoxia. Nat Commun 2025; 16:4787. [PMID: 40404627 PMCID: PMC12098657 DOI: 10.1038/s41467-025-59924-7] [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: 06/04/2024] [Accepted: 05/02/2025] [Indexed: 05/24/2025] Open
Abstract
Stem cell-derived islet cell therapy can effectively treat type 1 diabetes, but its efficacy is hindered by low oxygen supply post-transplantation, particularly in subcutaneous spaces and encapsulation devices, leading to cell dysfunction. The response to hypoxia and effective strategies to alleviate its detrimental effects remain poorly understood. Here, we show that β cells within stem cell-derived islets gradually undergo a decline in cell identity and metabolic function in hypoxia. This is linked to reduced expression of immediate early genes (EGR1, FOS, and JUN), which downregulates key β cell transcription factors. We further identified genes important for maintaining β cell fitness in hypoxia, with EDN3 as a potent player. Elevated EDN3 expression preserves β cell identity and function in hypoxia by modulating genes involved in β cell maturation, glucose sensing and regulation. These insights improve the understanding of hypoxia's impact on stem cell-derived islets, offering a potential intervention for clinical applications.
Collapse
Affiliation(s)
- Xi Wang
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Shlomi Brielle
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Jennifer Kenty-Ryu
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Vertex Pharmaceuticals, Boston, USA
| | - Nataly Korover
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Danny Bavli
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Vertex Pharmaceuticals, Boston, USA
| | - Ramona Pop
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Vertex Pharmaceuticals, Boston, USA.
| |
Collapse
|
6
|
Marei HE. Stem cell therapy: a revolutionary cure or a pandora's box. Stem Cell Res Ther 2025; 16:255. [PMID: 40405306 PMCID: PMC12096755 DOI: 10.1186/s13287-025-04334-1] [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: 02/18/2025] [Accepted: 04/10/2025] [Indexed: 05/24/2025] Open
Abstract
This review article examines how stem cell therapies can cure various diseases and injuries while also discussing the difficulties and moral conundrums that come with their application. The article focuses on the revolutionary developments in stem cell research, especially the introduction of gene editing tools like CRISPR-Cas9, which can potentially improve the safety and effectiveness of stem cell-based treatments. To guarantee the responsible use of stem cells in clinical applications, it is also argued that standardizing clinical procedures and fortifying ethical and regulatory frameworks are essential first steps. The assessment also highlights the substantial obstacles that still need to be addressed, such as the moral dilemmas raised by the use of embryonic stem cells, the dangers of unlicensed stem cell clinics, and the difficulties in obtaining and paying for care for patients. The study emphasizes how critical it is to address these problems to stop exploitation, guarantee patient safety, and increase the accessibility of stem cell therapy. The review also addresses the significance of thorough clinical trials, public education, and policy development to guarantee that stem cell research may fulfill its full potential. The review concludes by describing stem cell research as a promising but complicated topic that necessitates a thorough evaluation of both the hazards and the benefits. To overcome the ethical, legal, and accessibility obstacles and eventually guarantee that stem cell treatments may be safely and fairly included in conventional healthcare, it urges cooperation between the scientific community, legislators, and the general public.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| |
Collapse
|
7
|
Patel A, Rajgopal B, Jaiswal M. Various strategies to induce beta cell neogenesis: a comprehensive review for unravelling the potential future therapy for curing diabetes. Growth Factors 2025:1-28. [PMID: 40400239 DOI: 10.1080/08977194.2025.2508723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Pancreatic endocrine cells are categorized in to 5 types (alpha, beta, delta, pancreatic polypeptide cells and epsilon), which expresses glucagon, insulin, somatostatin, pancreatic polypeptide, and ghrelin, respectively. Several studies including lineage tracing in Ins2Akita diabetic mice have been done to investigate the identities of pancreatic endocrine cells which concludes, alpha cells have enormous plasticity, which enables them to be reprogrammed by specific transcription factors into insulin secreting beta like cells. Gene therapy has provided the beneficial outcome. Pdx1, MaFA and PAX4 (the transcription factors) in alpha cells can be over expressed which results in reprogramming the targeted alpha cells into beta cells. This trans-differentiation may be induced by infusing an adeno-associated virus (AAV) loaded with distinct transcription factors in the duct of pancreas. Several researches have demonstrated the successful restoration of enhanced insulin secretion in diabetes induced mice. Additionally ductal neurogenin3 (Ngn3), Sglt2 inhibitors, Igfbp1, GLP1 and several clinical and non-clinical agents has been postulated as a basis of beta cell neogenesis. Alpha cell owing to its high plasticity, on prolonged exposure to GABA reprogrammed into beta-like cell due to downregulation of Arx expression by GABA. The various approaches for beta cell neogenesis open a new window towards the establishment of novel gene therapy accession to treat diabetes. However, broad studies are still needed to improve and optimize this treatment methodology. The potentiality of endogenous pancreatic alpha cell to beta cell conversion methods and its outcomes are invigorating. This accomplishment is presently being under trial in non-human primates.
Collapse
Affiliation(s)
- Anjali Patel
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, India
| | - B Rajgopal
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, India
| | - Manisha Jaiswal
- Rungta Institute of Pharmaceutical Education and Research, Bhilai, India
| |
Collapse
|
8
|
Tian C, Tiemann U, Hermann F, Semb H. Protocol for in vitro modeling of specification and morphogenesis of early pancreas development using human pluripotent stem cell-based organoid differentiation. STAR Protoc 2025; 6:103834. [PMID: 40402748 DOI: 10.1016/j.xpro.2025.103834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/11/2025] [Accepted: 05/01/2025] [Indexed: 05/24/2025] Open
Abstract
Here, we present a protocol to generate key pancreatic cell types in vitro using human pluripotent stem cell (hPSC)-based Matrigel-overlay organoid differentiation. These include multipotent and bipotent progenitors, endocrine progenitors, and hormone-producing endocrine cells. We describe steps for culturing hPSCs as a 2D monolayer, applying a Matrigel overlay to create a 3D epithelial niche, and guiding stepwise differentiation. This system supports live-cell imaging and real-time tracking of morphogenesis and fate decisions, providing a platform for studying organ development and disease. For complete details on the use and execution of this protocol, please refer to Ulf et al.1.
Collapse
Affiliation(s)
- Chenglei Tian
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum Munchen, 85764 Munich, Germany; Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ulf Tiemann
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Florian Hermann
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Henrik Semb
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum Munchen, 85764 Munich, Germany; Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
9
|
Kelley AB, Shunkarova M, Maestas MM, Gale SE, Mukherjee N, Hogrebe NJ, Millman JR. Controlling Human Stem Cell-Derived Islet Composition Using Magnetic Sorting. Biotechnol Bioeng 2025. [PMID: 40400171 DOI: 10.1002/bit.29030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/18/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Stem cell-derived islets (SC-islets) consist of multiple hormone-producing cell types and offer a promising therapeutic avenue for treating type 1 diabetes (T1D). Currently, the composition of cell types generated within these SC-islets cannot be controlled via soluble factors during this differentiation process and consist of off-target cell types. In this study, we devised a magnetic-activated cell sorting protocol to enrich SC-islets for CD49a, a marker associated with functional insulin-producing β cells. SC-islets were generated from human pluripotent stem cells using an adherent differentiation protocol and then sorted and aggregated into islet-like clusters to produce CD49a-enriched, CD49a-depleted, and unsorted SC-islets. Single-cell RNA sequencing (scRNA-seq) and immunostaining revealed that CD49a-enriched SC-islets had higher proportions of β cells and improved transcriptional identity compared to other cell types. Functional assays demonstrated that CD49a-enriched SC-islets exhibited enhanced glucose-stimulated insulin secretion both In vitro and In Vivo following transplantation into diabetic mice. These findings suggest that CD49a-based sorting significantly improves β cell identity and the overall function of SC-islets, improving their effectiveness for T1D cell replacement therapies.
Collapse
Affiliation(s)
- Allison B Kelley
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mira Shunkarova
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marlie M Maestas
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sarah E Gale
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Noyonika Mukherjee
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Silvano S, Napolitano T, Plaisant M, Sousa-De-Veiga A, Fofo H, Ayachi C, Allegrini B, Rekima S, Pichery E, Becam J, Lepage V, Treins C, Etasse L, Tran L, Thévenet J, Pasquetti G, Kerr-Conte J, Pattou F, Botti P, Arduini A, Mizrahi J, Charles B, Collombat P. RSPO1, a potent inducer of pancreatic β cell neogenesis. Cell Rep Med 2025; 6:102126. [PMID: 40339569 DOI: 10.1016/j.xcrm.2025.102126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 08/02/2024] [Accepted: 04/11/2025] [Indexed: 05/10/2025]
Abstract
Inducing the neogenesis of pancreatic insulin-producing β cells holds great promise for diabetes research. However, non-toxic compounds with such activities remain to be discovered. Herein, we report the identification of RSPO1, a key agonist of the Wnt/β-catenin pathway, as an inducer of β cell replication. Specifically, we provide evidence that RSPO1 promotes a significant increase in β cell neogenesis in vitro, ex vivo, and in vivo. Importantly, RSPO1 administration is sufficient to activate Wnt/β-catenin signaling in β cells and counter chemically induced or autoimmune-mediated diabetes. Similarly, an optimized analog of RSPO1, allowing for weekly administration, also prevents diabetes in vivo. Lastly, the treatment of transplanted human islets with RSPO1 induces a significant 2.78-fold increase in human β cell numbers in only 60 days, these cells being functional. Such activities of RSPO1 to promote β cell neogenesis could therefore represent an unprecedented hope in the continued search for diabetes alternative therapies.
Collapse
Affiliation(s)
| | | | | | - Anette Sousa-De-Veiga
- University Nice Cote D'Azur, Inserm, CNRS, iBV, 06100 Nice, France; iBV, Institut de Biologie Valrose, University Nice Sophia Antipolis, Centre de Biochimie, Parc Valrose, 28, Avenue Valrose, 06108 Nice Cedex 2, France
| | - Hugo Fofo
- University Nice Cote D'Azur, Inserm, CNRS, iBV, 06100 Nice, France; iBV, Institut de Biologie Valrose, University Nice Sophia Antipolis, Centre de Biochimie, Parc Valrose, 28, Avenue Valrose, 06108 Nice Cedex 2, France
| | - Chaïma Ayachi
- University Nice Cote D'Azur, Inserm, CNRS, iBV, 06100 Nice, France; iBV, Institut de Biologie Valrose, University Nice Sophia Antipolis, Centre de Biochimie, Parc Valrose, 28, Avenue Valrose, 06108 Nice Cedex 2, France
| | - Benoit Allegrini
- University Nice Cote D'Azur, Inserm, CNRS, iBV, 06100 Nice, France; iBV, Institut de Biologie Valrose, University Nice Sophia Antipolis, Centre de Biochimie, Parc Valrose, 28, Avenue Valrose, 06108 Nice Cedex 2, France
| | - Samah Rekima
- University Nice Cote D'Azur, Inserm, CNRS, iBV, 06100 Nice, France; iBV, Institut de Biologie Valrose, University Nice Sophia Antipolis, Centre de Biochimie, Parc Valrose, 28, Avenue Valrose, 06108 Nice Cedex 2, France
| | | | - Jérôme Becam
- Aix-Marseille Université, CNRS, Laboratoire de Chimie Bactérienne, UMR 7283, Institut de Microbiologie de la Méditerranée, 31 Chemin Joseph Aiguier, 13009 Marseille, France
| | - Valentin Lepage
- University Nice Cote D'Azur, Inserm, CNRS, iBV, 06100 Nice, France; iBV, Institut de Biologie Valrose, University Nice Sophia Antipolis, Centre de Biochimie, Parc Valrose, 28, Avenue Valrose, 06108 Nice Cedex 2, France
| | | | - Laura Etasse
- DiogenX, 180 Avenue du Prado, 13008 Marseille, France
| | - Loan Tran
- DiogenX, 180 Avenue du Prado, 13008 Marseille, France
| | - Julien Thévenet
- University Lille, Inserm, CHU Lille, U1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, EGID, 59000 Lille, France
| | - Gianni Pasquetti
- University Lille, Inserm, CHU Lille, U1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, EGID, 59000 Lille, France
| | - Julie Kerr-Conte
- University Lille, Inserm, CHU Lille, U1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, EGID, 59000 Lille, France
| | - François Pattou
- University Lille, Inserm, CHU Lille, U1190 Translational Research for Diabetes, European Genomic Institute for Diabetes, EGID, 59000 Lille, France
| | - Paolo Botti
- DiogenX, 180 Avenue du Prado, 13008 Marseille, France
| | | | | | | | - Patrick Collombat
- DiogenX, 180 Avenue du Prado, 13008 Marseille, France; University Nice Cote D'Azur, Inserm, CNRS, iBV, 06100 Nice, France; iBV, Institut de Biologie Valrose, University Nice Sophia Antipolis, Centre de Biochimie, Parc Valrose, 28, Avenue Valrose, 06108 Nice Cedex 2, France.
| |
Collapse
|
11
|
Jun Y, Nguyen-Ngoc KV, Sai S, Bender RHF, Gong W, Kravets V, Zhu H, Hatch CJ, Schlichting M, Gaetani R, Mallick M, Hachey SJ, Christman KL, George SC, Hughes CCW, Sander M. Engineered vasculature induces functional maturation of pluripotent stem cell-derived islet organoids. Dev Cell 2025:S1534-5807(25)00262-X. [PMID: 40412386 DOI: 10.1016/j.devcel.2025.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/05/2024] [Accepted: 04/28/2025] [Indexed: 05/27/2025]
Abstract
Blood vessels play a critical role in pancreatic islet function, yet current methods for deriving islet organoids from human pluripotent stem cells (SC-islets) lack vasculature. We engineered three-dimensional (3D) vascularized SC-islet organoids by assembling SC-islet cells, human primary endothelial cells (ECs), and fibroblasts in a non-perfused model and a microfluidic device with perfused vessels. Vasculature improved stimulus-dependent Ca2+ influx into SC-β cells, a hallmark of β cell function that is blunted in non-vascularized SC-islets. Moreover, vascularization accelerated diabetes reversal post engraftment of a subtherapeutic SC-islet dose into mice. We show that vasculature leads to the formation of an islet-like basement membrane that contributes to the functional improvement of SC-β cells. Furthermore, single-cell RNA sequencing (scRNA-seq) data predicted BMP2/4-BMPR2 signaling from ECs to SC-β cells, and correspondingly, BMP4 enhanced the SC-β cell Ca2+ response and insulin secretion. Vascularized SC-islets will enable further studies of crosstalk between β cells and ECs and will serve as an in vitro platform for disease modeling and therapeutic testing.
Collapse
Affiliation(s)
- Yesl Jun
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA; Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Kim-Vy Nguyen-Ngoc
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Somesh Sai
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany; Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin 14195, Germany
| | - R Hugh F Bender
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Winnie Gong
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vira Kravets
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA; Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Han Zhu
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christopher J Hatch
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Michael Schlichting
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA; Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Roberto Gaetani
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Medhavi Mallick
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephanie J Hachey
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Karen L Christman
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Stem Cell Institute, University of California, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Maike Sander
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA; Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Cellular & Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Charité-Universitätsmedizin, Berlin 10117, Germany.
| |
Collapse
|
12
|
Miller J, Perrier Q, Rengaraj A, Bowlby J, Byers L, Peveri E, Jeong W, Ritchey T, Gambelli AM, Rossi A, Calafiore R, Tomei A, Orlando G, Asthana A. State of the Art of Bioengineering Approaches in Beta-Cell Replacement. CURRENT TRANSPLANTATION REPORTS 2025; 12:17. [PMID: 40342868 PMCID: PMC12055624 DOI: 10.1007/s40472-025-00470-y] [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] [Accepted: 04/09/2025] [Indexed: 05/11/2025]
Abstract
Purpose of the Review Despite recent advancements in technology for the treatment of type 1 diabetes (T1D), exogenous insulin delivery through automated devices remains the gold standard for treatment. This review will explore progress made in pancreatic islet bioengineering within the field of beta-cell replacement for T1D treatment. Recent Findings First, we will focus on the use of decellularized extracellular matrices (dECM) as a platform for pancreatic organoid development. These matrices preserve microarchitecture and essential biochemical signals for cell differentiation, offering a promising alternative to synthetic matrices. Second, advancements in 3D bioprinting for creating complex organ structures like pancreatic islets will be discussed. This technology allows for increased precision and customization of cellular models, crucial for replicating native pancreatic islet functionality. Finally, this review will explore the use of stem cell-derived organoids to generate insulin-producing islet-like cells. While these organoids face challenges such as functional immaturity and poor vascularization, they represent a significant advancement for disease modeling, drug screening, and autologous islet transplantation. Summary These innovative approaches promise to revolutionize T1D treatment by overcoming the limitations of traditional therapies based on human pancreatic islets.
Collapse
Affiliation(s)
- Jake Miller
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Quentin Perrier
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
- Univ. Grenoble Alpes, Department of Pharmacy, Grenoble Alpes University Hospital, Grenoble, France
| | - Arunkumar Rengaraj
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Joshua Bowlby
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Lori Byers
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Emma Peveri
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Wonwoo Jeong
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Thomas Ritchey
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | | | - Arianna Rossi
- Department of Engineering, University of Perugia, Perugia, Italy
| | | | - Alice Tomei
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Giuseppe Orlando
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Amish Asthana
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| |
Collapse
|
13
|
Kieffer TJ, Hoesli CA, Shapiro AMJ. Advances in Islet Transplantation and the Future of Stem Cell-Derived Islets to Treat Diabetes. Cold Spring Harb Perspect Med 2025; 15:a041624. [PMID: 39074874 PMCID: PMC12047745 DOI: 10.1101/cshperspect.a041624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
β-Cell replacement for type 1 diabetes (T1D) can restore normal glucose homeostasis, thereby eliminating the need for exogenous insulin and halting the progression of diabetes complications. Success in achieving insulin independence following transplantation of cadaveric islets fueled academic and industry efforts to develop techniques to mass produce β cells from human pluripotent stem cells, and these have now been clinically validated as an alternative source of regulated insulin production. Various encapsulation strategies are being pursued to contain implanted cells in a retrievable format, and different implant sites are being explored with some strategies reaching clinical studies. Stem cell lines, whether derived from embryonic sources or reprogrammed somatic cells, are being genetically modified for designer features, including immune evasiveness to enable implant without the use of chronic immunosuppression. Although hurdles remain in optimizing large-scale manufacturing, demonstrating efficacy, durability, and safety, products containing stem cell-derived β cells promise to provide a potent treatment for insulin-dependent diabetes.
Collapse
Affiliation(s)
- Timothy J Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, School of Biomedical Engineering
- Department of Surgery, The University of British Columbia, Vancouver V6T1Z3, British Columbia, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, Department of Biomedical Engineering, McGill University, Montreal H3A 0C5, Québec, Canada
- Associate Member, Department of Biomedical Engineering, McGill University, Montreal H3A 0C5, Québec, Canada
| | - A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, Edmonton T6G2E1, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton T6G2E1, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton T6G2E1, Alberta, Canada
| |
Collapse
|
14
|
Britt M, Abdilmasih N, Rezanejad H. Pancreatic Ductal Cell Heterogeneity: Insights into the Potential for β-Cell Regeneration in Diabetes. Stem Cell Rev Rep 2025; 21:953-963. [PMID: 40063303 DOI: 10.1007/s12015-025-10859-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2025] [Indexed: 05/24/2025]
Abstract
Diabetes mellitus is a significant and fast-growing health problem worldwide. Cost, donor shortages, and immune rejection limit current treatment strategies. While considerable progress has been made in creating β-cells in vitro with remarkable morphological and functional resemblance to those in primary pancreatic islets, exploring alternative sources for β-cell replacement is crucial. With adult pancreatic stem cells still not conclusively identified, researchers focus their attention on heterogeneity within pancreatic ductal epithelial cells, exploring these cells as a potential source of progenitor cells for pancreatic regeneration and β-cell formation. Recent studies using techniques such as fluorescence-activated cell sorting, immunostaining and single cell RNA-sequencing have identified ductal cell heterogeneity with several subpopulations of ductal cells with progenitor-like properties and their capacity for differentiation into insulin producing cells. Here, we have reviewed the most recent studies on pancreatic ductal cell subpopulations that offer insights into potential stem-cell populations to form β-cells in diabetes treatment.
Collapse
Affiliation(s)
- Madelaine Britt
- Biological Sciences Department, Macewan University, Edmonton, Canada
| | | | - Habib Rezanejad
- Biological Sciences Department, Macewan University, Edmonton, Canada.
| |
Collapse
|
15
|
Jiang Y, Xu Z, Wu Y, Li X, Ling J, Chen Y, Zhu Z, Yang P, Liu X, Zhang D, Liu J, Yin X, Zhang J, Yu P. Exploring the progress and trends of immunotherapy for type 1 diabetes: A comprehensive bibliometric analysis spanning nearly two decades. Obes Rev 2025; 26:e13888. [PMID: 39871677 DOI: 10.1111/obr.13888] [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: 07/19/2024] [Revised: 09/19/2024] [Accepted: 10/21/2024] [Indexed: 01/29/2025]
Abstract
INTRODUCTION Immunotherapy is a crucial treatment for type 1 diabetes (T1D), yet analyses focusing on research priorities and trends in this field are limited. Therefore, this study employs bibliometric methods to systematically explore the current research status of immunotherapy for T1D. METHODS Based on the Web of Science Core Collection Database, 1573 articles and review articles related to immunotherapy for T1D published from 2004 to 2023 were screened for bibliometric analysis. VOSviewer, CiteSpace, and R software were applied to comprehensively analyze the number of publications, journals, countries, authors, institutions, keywords, and references. RESULTS In the past two decades, the global annual publication rate has seen a significant increase of 238.24%. Almost 40% of all publications have appeared in the last 5 years, accounting for over 50% of total citations. Journals such as Diabetes, Journal of Autoimmunity and Frontiers in Immunology have exerted substantial influence. Collaboration across nations has been notably strong, with the United States leading the way. The University of Florida is the most productive institution. Terms like "nivolumab," "ipilimumab," "pembrolizumab," and "immune checkpoint inhibitor(s)" gain considerable traction. The majority of research has clustered around themes such as immunomodulation, autoimmune diseases, immune checkpoint inhibitors, mesenchymal stem cells, and cell therapy. Precision medicine, immune checkpoint inhibitors, and nanotechnology are trending focal points in contemporary research. CONCLUSION The outcomes of the study are instrumental in enabling scholars to comprehend the evolving trajectory of immunotherapeutic approaches for T1D and facilitate the swift recognition of emerging research pathways.
Collapse
Affiliation(s)
- Yixin Jiang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, The Queen Mary College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhou Xu
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xinglei Li
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Zicheng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
16
|
Shang KM, Suzuki T, Kato H, Toyoda T, Tai YC, Komatsu H. Oxygen dynamics and delivery strategies to enhance beta cell replacement therapy. Am J Physiol Cell Physiol 2025; 328:C1667-C1684. [PMID: 40204281 DOI: 10.1152/ajpcell.00984.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/06/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Beta cell replacement therapy via pancreatic islet transplantation offers a promising treatment for type 1 diabetes as an alternative to insulin injections. However, posttransplantation oxygenation remains a critical challenge; isolated islets from donors lose vascularity and rely on slow oxygen diffusion for survival until revascularization occurs in the host tissue. This often results in significant hypoxia-induced acute graft loss. Overcoming the oxygenation barrier is crucial for advancing islet transplantation. This review is structured in three sections: the first examines oxygen dynamics in islet transplantation, focusing on factors affecting oxygen supply, including vascularity. It highlights oxygen dynamics specific to both transplant sites and islet grafts, with particular attention to extrahepatic sites such as subcutaneous tissue. The second section explores current oxygen delivery strategies, categorized into two main approaches: augmenting oxygen supply and enhancing effective oxygen solubility. The final section addresses key challenges, such as the lack of a clearly defined oxygen threshold for islet survival and the limited precision in measuring oxygen levels within small islet constructs. Recent advancements addressing these challenges are introduced. By deepening the understanding of oxygen dynamics and identifying current obstacles, this review aims to guide the development of innovative strategies for future research and clinical applications. These advancements are anticipated to enhance transplantation outcomes and bring us closer to a cure for type 1 diabetes.
Collapse
Affiliation(s)
- Kuang-Ming Shang
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, United States
| | - Tomoharu Suzuki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hiroyuki Kato
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Taro Toyoda
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yu-Chong Tai
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, United States
| | - Hirotake Komatsu
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| |
Collapse
|
17
|
Dor Y, Spitzer NC. Hormone-switching islet cells: parallels to transmitter-switching neurons. Front Cell Dev Biol 2025; 13:1587893. [PMID: 40356600 PMCID: PMC12066745 DOI: 10.3389/fcell.2025.1587893] [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: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Although originating from different germ layers, pancreatic islet cells and neurons share extensive similarities, both physiological (e.g., voltage-dependent release of a bioactive molecule) and molecular (e.g., highly similar composition of transcription factors and structural genes). Here we propose that two seemingly unrelated phenomena recognized in these cell types-neurotransmitter switching in neurons and the expression of two or more hormones in individual islet cells-share a deep resemblance, potentially reflecting an ancient molecular circuit of cell plasticity. Comparing and contrasting dynamic hormone expression in islet cells and transmitter switching in neurons may provide insights into the functions and underlying mechanisms of these phenomena.
Collapse
Affiliation(s)
- Yuval Dor
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Nicholas C. Spitzer
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
18
|
Geng A, Yuan S, Yu QC, Zeng YA. The role of endothelial cells in pancreatic islet development, transplantation and culture. Front Cell Dev Biol 2025; 13:1558137. [PMID: 40330424 PMCID: PMC12052768 DOI: 10.3389/fcell.2025.1558137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/03/2025] [Indexed: 05/08/2025] Open
Abstract
Endothelial cells (ECs) play pivotal roles in the development and maintenance of tissue homeostasis. During development, vasculature actively involves in organ morphogenesis and functional maturation, through the secretion of angiocrine factors and extracellular matrix components. Islets of Langerhans, essential functional units of glucose homeostasis, are embedded in a dense endothelial capillary network. Islet vasculature not only supplies nutrients and oxygen to endocrine cells but also facilitate the rapid delivery of pancreatic hormones to target tissues, thereby ensuring precise glucose regulation. Diabetes mellitus is a major disease burden and is caused by islet dysfunction or depletion, often accompanied by vessel loss and dysregulation. Therefore, elucidating the regulatory mechanisms of ECs within islets hold profound implications for diabetes therapy. This review provides an overview of recent research advancements on the functional roles of ECs in islet biology, transplantation, and in vitro islet organoid culture.
Collapse
Affiliation(s)
- Ajun Geng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shubo Yuan
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing Cissy Yu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Arial Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
19
|
Wei H, Kappler C, Green E, Jiang H, Yeung T, Wang H. GRP94 is indispensable for definitive endoderm specification of human induced pluripotent stem cells. Mol Ther 2025:S1525-0016(25)00299-0. [PMID: 40254879 DOI: 10.1016/j.ymthe.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/24/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived insulin-producing β cell therapy shows promise in treating type 1 diabetes and potentially type 2 diabetes. Understanding the genetic factors controlling hiPSC differentiation could optimize this therapy. In this study, we investigated the role of glucose-regulated protein 94 (GRP94) in human β cell development by generating HSP90B1/GRP94 knockout (KO) hiPSCs, re-expressing GRP94 in the mutants and inducing their β cell differentiation. Our results revealed that GRP94 depletion hindered β cell generation by promoting cell death induced by endoplasmic reticulum (ER) stress and other stressors during definitive endoderm (DE) differentiation. Moreover, GRP94 deletion resulted in decreased activation of WNT/β-catenin signaling, which is critical for DE specification. Re-expression of GRP94 in GRP94 KO iPSCs partially reversed DE differentiation deficiency and alleviated cell death. These findings highlight the previously unrecognized indispensable role of GRP94 in human DE formation and consequent β cell development from hiPSCs. GRP94 mitigates ER stress-induced cell death and regulates the WNT/β-catenin signaling pathway, which is both crucial for successful β cell differentiation. These results provide new insights into the molecular mechanisms underlying β cell differentiation from hiPSCs and suggest that targeting GRP94 pathways could enhance the efficiency of hiPSC-derived insulin-producing cell therapies for diabetes treatment.
Collapse
Affiliation(s)
- Hua Wei
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Christiana Kappler
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Erica Green
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Hanna Jiang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Tiffany Yeung
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA; Ralph H Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
20
|
Chen J, Guo Y, Zheng Y, Chen Z, Xu H, Pan S, Liang X, Zhai L, Guan YQ. Oral glucose-responsive nanoparticles loaded with artemisinin induce pancreatic β-cell regeneration for the treatment of type 2 diabetes. J Colloid Interface Sci 2025; 684:769-782. [PMID: 39823951 DOI: 10.1016/j.jcis.2025.01.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
Type 2 diabetes (T2D) is a chronic disease characterized by long-term insulin resistance (IR) and pancreatic β-cell dysfunction. Conventional T2D medication ignores pancreatic β-cell damage. In this study, we designed an oral glucose-responsive nanoparticle for pancreatic β-cell regeneration and treatment of T2D. It was formed by carboxymethyl chitosan (CMC) grafted with 3-aminophenylboronic acid (APBA) as the shell and small-molecule citrus pectin (MCP) spheres encapsulating artemisinin (Art) connected by borate ester bonds. The prepared CMC-APBA wrapped Art-loaded MCP nanoparticles (CAM@Art) had therapeutic effects for the treatment of IR, antioxidant and promotion of pancreatic α-cell differentiation in vitro experiments. In addition, in vivo experiments showed that CAM@Art could reduce blood glucose, oxidative stress and inflammation levels and reverse IR in diabetic rats. Importantly, pancreatic β-cell regeneration was found in islets in vivo. Mechanistically, CAM@Art promotes pancreatic α-cell differentiation by promoting overexpression of the transcription factor Pax4 and ectopic expression of Arx. The results suggest that the present study provides a promising therapeutic strategy for the treatment of diabetic pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Jiapeng Chen
- School of Life Science, South China Normal University, Guangzhou 510631 China
| | - Yiyan Guo
- School of Life Science, South China Normal University, Guangzhou 510631 China
| | - Yuxin Zheng
- School of Life Science, South China Normal University, Guangzhou 510631 China
| | - Zhendong Chen
- School of Life Science, South China Normal University, Guangzhou 510631 China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou 511400 China
| | - Haoming Xu
- School of Life Science, South China Normal University, Guangzhou 510631 China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou 511400 China
| | - Shengjun Pan
- School of Life Science, South China Normal University, Guangzhou 510631 China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou 511400 China
| | - Xuanxi Liang
- School of Life Science, South China Normal University, Guangzhou 510631 China
| | - Limin Zhai
- School of Life Science, South China Normal University, Guangzhou 510631 China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou 510631 China; Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631 China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou 511400 China.
| |
Collapse
|
21
|
Tian C, Rump A, Ebeid C, Mamidi A, Semb H. Salt-inducible kinases transduce mechanical forces into the specification of the pancreatic endocrine lineage. Stem Cell Reports 2025; 20:102444. [PMID: 40054471 PMCID: PMC12069894 DOI: 10.1016/j.stemcr.2025.102444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 04/11/2025] Open
Abstract
The extracellular matrix-F-actin-Yes-associated protein 1 (YAP1)-Notch mechanosignaling axis is a gatekeeper in the fate decisions of bipotent pancreatic progenitors (bi-PPs). However, the link between F-actin dynamics and YAP1 activity remains poorly understood. Here, we identify salt-inducible kinases (SIKs) as mediators of F-actin-triggered changes in YAP1 activity. Interestingly, sodium chloride treatment promotes the differentiation of bi-PPs into NEUROG3+ endocrine progenitors (EPs) through enhanced SIK expression. Consistently, the pan-SIK inhibitor HG-9-09-01 (HG) inhibits latrunculin B (LatB)-induced EP differentiation via nuclear YAP1 accumulation. Unexpectedly, withdrawal of HG after a 12-h treatment increased SIK expression by a negative feedback mechanism, leading to significantly enhanced endocrinogenesis. Therefore, the combined treatment of bi-PPs with LatB and HG for 12 h boosted endocrinogenesis, ultimately leading to an increased number of beta cells. In summary, we identify SIKs as new transducers of mechanotransduction-triggered induction of pancreatic endocrine cell fates.
Collapse
Affiliation(s)
- Chenglei Tian
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum Munchen, Munich, Germany; Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Adam Rump
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum Munchen, Munich, Germany; Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Christine Ebeid
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Anant Mamidi
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Henrik Semb
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum Munchen, Munich, Germany; Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
22
|
Russ-Silsby J, Lee Y, Rajesh V, Amoli M, Mirhosseini NA, Godbole T, Johnson MB, Ibarra DE, Sun H, Krentz NAJ, Wakeling MN, Flanagan SE, Hattersley AT, Gloyn AL, De Franco E. Complete Loss of PAX4 causes Transient Neonatal Diabetes in Humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.01.25324926. [PMID: 40236391 PMCID: PMC11998800 DOI: 10.1101/2025.04.01.25324926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Gene discovery studies in individuals with diabetes diagnosed within 6 months of life (neonatal diabetes, NDM) can provide unique insights into the development and function of human pancreatic beta-cells. We describe the identification of homozygous PAX4 loss-of-function variants in 2 unrelated individuals with NDM: a p.(Arg126*) stop-gain variant and a c.-352_104del deletion affecting the first 4 PAX4 exons. We confirmed the p.(Arg126*) variant causes nonsense mediated decay in CRISPR-edited human induced pluripotent stem cell (iPSC)-derived pancreatic endoderm cells. Integrated analysis of CUT&RUN and RNA-sequencing in PAX4-depleted islet cell models identified genes directly regulated by PAX4 involved in both pancreatic islet development and glucose-stimulated insulin secretion. Both probands had transient NDM which remitted in early infancy but relapsed between the ages of 2 and 7 years, demonstrating that in contrast to mouse models, PAX4 is not essential for the development of human pancreatic beta-cells.
Collapse
Affiliation(s)
- James Russ-Silsby
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Yunkyeong Lee
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Varsha Rajesh
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Mahsa Amoli
- Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Iran
| | | | | | - Matthew B. Johnson
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Dora E. Ibarra
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Han Sun
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Nicole A. J. Krentz
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Matthew N. Wakeling
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Sarah E. Flanagan
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Andrew T. Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Anna L. Gloyn
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, School of Medicine, Stanford University, Stanford, CA, USA
| | - Elisa De Franco
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| |
Collapse
|
23
|
Ribezzi D, Català P, Pignatelli C, Citro A, Levato R. Bioprinting and synthetic biology approaches to engineer functional endocrine pancreatic constructs. Trends Biotechnol 2025:S0167-7799(25)00090-3. [PMID: 40185667 DOI: 10.1016/j.tibtech.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/17/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
Diabetes is a complex disease affecting over 500 million people worldwide. Traditional approaches, such as insulin delivery, are mainstay treatments, but do not cure the disease. Recent advances in biofabrication and synthetic biology offer new hope for the development of tissue constructs recapitulating salient organ functions. Here, we discuss recent progress in bioprinting a functional endocrine pancreas, ranging from cell sources to main advances in biomaterials. We review innovative areas for the development of this field, with a particular focus on the convergence of synthetic biology and cell engineering with bioprinting, which opens new avenues for developing advanced in vitro models and regenerative, transplantable grafts, with the potential to provide independence from exogenous insulin administration.
Collapse
Affiliation(s)
- Davide Ribezzi
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pere Català
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
24
|
Rajaei B, Garcia AM, Juksar J, Doppenberg JB, Paz-Barba M, Boot F, de Vos W, Mulder AA, Lambregtse F, Daleman L, de Leeuw AE, Nieveen MC, Engelse MA, Rabelink T, de Koning EJP, Carlotti F. Clinically compliant enrichment of human pluripotent stem cell-derived islets. Sci Transl Med 2025; 17:eadl4390. [PMID: 40173261 DOI: 10.1126/scitranslmed.adl4390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 08/28/2024] [Accepted: 03/10/2025] [Indexed: 04/04/2025]
Abstract
Human pluripotent stem cell-derived islet (SC-islet) transplantation is a promising β cell replacement therapy for patients with type 1 diabetes, offering a potential unlimited cell supply. Yet, the heterogeneity of the final cell product containing non-target cell types has relevant implications for SC-islet function, transplant volume, and cell product safety. Here, we present a clinically compliant, full three-dimensional differentiation protocol that includes a purification step of endocrine cell-rich clusters, relying on the principle of isopycnic centrifugation (density gradient separation). Enriched SC-islets displayed signs of functionality in vitro and in vivo. In contrast with antibody-based single-cell sorting approaches, this method does not destroy the islet cytoarchitecture associated with alterations of islet function and cell loss. Furthermore, it is fast, is easily scalable to large cell volumes, and can be applied during cell manufacturing. This method may also contribute to the generation of improved cell-based therapies for regenerative medicine purposes beyond the SC-islet field.
Collapse
Affiliation(s)
- Bahareh Rajaei
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Amadeo Muñoz Garcia
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Juri Juksar
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Jason B Doppenberg
- LUMC Transplant Center, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Miriam Paz-Barba
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Fransje Boot
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Willemijn de Vos
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Aat A Mulder
- Department of Cell and Chemistry Biology, Electron Microscopy Facility, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Ferdy Lambregtse
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Lizanne Daleman
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Anne E de Leeuw
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Maaike C Nieveen
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Marten A Engelse
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Ton Rabelink
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden 2333ZA, Netherlands
| |
Collapse
|
25
|
Imada T, Sasaki S, Yamaguchi H, Ueda A, Kawamori D, Katakami N, Shimomura I. Imeglimin, unlike metformin, does not perturb differentiation of human induced pluripotent stem cells towards pancreatic β-like cells and rather enhances gain in β cell identity gene sets. J Diabetes Investig 2025; 16:584-597. [PMID: 39829307 PMCID: PMC11970301 DOI: 10.1111/jdi.14410] [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: 07/23/2024] [Revised: 12/26/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
AIMS/INTRODUCTION Metformin treatment for hyperglycemia in pregnancy (HIP) beneficially improves maternal glucose metabolism and reduces perinatal complications. However, metformin could impede pancreatic β cell development via impaired mitochondrial function. A new anti-diabetes drug imeglimin, developed based on metformin, improves mitochondrial function. Here we examine the effect of imeglimin on β cell differentiation using human induced pluripotent stem cell (iPSC)-derived pancreatic islet-like spheroid (SC-islet) models. MATERIALS AND METHODS Human iPSCs are differentiated into SC-islets by three-dimensional culture with and without imeglimin or metformin. Differentiation efficiencies of SC-islets were analyzed by flow cytometry, immunostaining, quantitative PCR, and insulin secretion assay. RNA sequencing and oxygen consumption rate were obtained for further characterization of SC-islets. SC-islets were cultured with proinflammatory cytokines, in part mimicking the uterus environment in HIP. RESULTS Metformin perturbed SC-islet differentiation while imeglimin did not alter it. Furthermore, imeglimin enhanced the gene expressions of β cell lineage markers. Maintenance of mitochondrial function and optimization of TGF-β and Wnt signaling were considered potential mechanisms for augmented β cell maturation by imeglimin. In the presence of proinflammatory cytokines, imeglimin ameliorated β cell differentiation impaired by cytokines and metformin. CONCLUSIONS Imeglimin does not perturb differentiation of SC-islet cells and rather enhances gain in β cell identity gene sets in contrast to metformin. This may lead to the improvement of in vitro β cell differentiation protocols.
Collapse
Affiliation(s)
- Tasuku Imada
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Shugo Sasaki
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Hiroki Yamaguchi
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Ayaka Ueda
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Dan Kawamori
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
- Medical Education Center, Faculty of MedicineOsaka UniversityOsakaJapan
- Postgraduate Medical Training CenterOsaka University Hospital, Osaka UniversityOsakaJapan
| | - Naoto Katakami
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Iichiro Shimomura
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| |
Collapse
|
26
|
Piemonti L. The Last Mile in Beta-Cell Replacement Therapy for Type 1 Diabetes: Time to Grow Up. Transpl Int 2025; 38:14565. [PMID: 40236754 PMCID: PMC11998595 DOI: 10.3389/ti.2025.14565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
Beta cell replacement therapy for type 1 diabetes (T1D) is undergoing a transformative shift, driven by advances in stem cell biology, gene editing, and tissue engineering. While islet transplantation has demonstrated proof-of-concept success in restoring endogenous insulin production, its clinical impact remains limited by donor scarcity, immune rejection, and procedural complexities. The emergence of stem cell-derived beta-like cells represents a paradigm shift, with initial clinical trials showing promising insulin secretion in vivo. However, translating these breakthroughs into scalable, widely accessible treatments poses significant challenges. Drawing parallels to space exploration, this paper argues that while scientific feasibility has been demonstrated, true accessibility remains elusive. Without a strategic shift, beta cell therapy risks becoming an elite intervention, restricted by cost and infrastructure. Lessons from gene and cell therapies for rare diseases highlight the dangers of unsustainable pricing and limited market viability. To bridge the "last mile" a Quality by Design approach is proposed, emphasizing scalability, ease of use, and economic feasibility from the outset. By emphasizing practical implementation over academic achievements, corporate interests, market economics, or patent constraints, beta cell therapy can progress from proof-of-concept to a viable, widely accessible treatment.
Collapse
Affiliation(s)
- Lorenzo Piemonti
- Unit of Regenerative Medicine and Organ Transplants, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
27
|
Juksar J, Mijdam R, Bosman S, van Oudenaarden A, Carlotti F, de Koning EJP. Effects of Neurogenin 3 Induction on Endocrine Differentiation and Delamination in Adult Human Pancreatic Ductal Organoids. Transpl Int 2025; 38:13422. [PMID: 40236756 PMCID: PMC11996654 DOI: 10.3389/ti.2025.13422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
Diabetes mellitus is characterized by the loss of pancreatic insulin-secreting β-cells in the Islets of Langerhans. Understanding the regenerative potential of human islet cells is relevant in the context of putative restoration of islet function after damage and novel islet cell replacement therapies. Adult human pancreatic tissue can be cultured as three-dimensional organoids with the capacity for long-term expansion and the promise of endocrine cell formation. Here, we characterize the endocrine differentiation potential of human adult pancreatic organoids. Because exocrine-to-endocrine differentiation is dependent on the expression of Neurogenin 3 (NEUROG3), we first generated NEUROG3-inducible organoid lines. We show that doxycycline-induced NEUROG3 expression in the organoids leads to the formation of chromogranin A positive (CHGA+) endocrine progenitor cells. The efficiency of this differentiation was improved with the addition of thyroid hormone T3 and the AXL inhibitor R428. Further, compound screening demonstrated that modifying the pivotal embryonic endocrine pancreas signalling pathways driven by Notch, YAP, and EGFR led to increased NEUROG3 expression in organoids. In a similar fashion to embryonic development, adult ductal cells delaminated from the organoids after NEUROG3 induction. Thus, mechanisms in islet (re)generation including the initiation of endocrine differentiation and delamination can be achieved by NEUROG3 induction.
Collapse
Affiliation(s)
- Juri Juksar
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Rachel Mijdam
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, Netherlands
| | - Sabine Bosman
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, Netherlands
| | | | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Eelco J. P. de Koning
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
28
|
Berney T, Thaunat O, Berishvili E. Allogeneic Islet Transplantation: Chronicle of a Death Foretold? Transpl Int 2025; 38:14598. [PMID: 40236755 PMCID: PMC11998596 DOI: 10.3389/ti.2025.14598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
Innovative solutions have entered the routine management of patients with type 1 diabetes or are making the headlines and this is shaking the world of beta cell replacement therapies. Above all, allogeneic islet transplantation is enthusiastically doomed to extinction by the aficionados of "closed loop" artificial insulin delivery systems or those convinced of the imminent large scale availability of stem-cell derived insulin-producing tissues. This opinion paper will propose that neither will be a universal solution in the very near future and will argue that xenogeneic islet transplantation may be a serious outsider in the race for new therapies. In the meantime, the odds are in favor of allogeneic islet (and pancreas) transplantation remaining first line options in the treatment of complicated type 1 diabetes. There is no question that "closed loop" systems have already greatly improved the management of type 1 diabetes, but, while "unlimited" sources of insulin-producing cells are jockeying for approval as standard-of-care, these improvements are more likely to drive a shift of indications -from islet transplant alone to simultaneous islet-kidney transplantation- than to herald the demise of islet transplantation.
Collapse
Affiliation(s)
- Thierry Berney
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Faculty Diabetes Center, University of Geneva Medical Center, University of Geneva, Geneva, Switzerland
- Institute of Medical and Public Health Research, Ilia State University, Tbilisi, Georgia
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University Lyon, Lyon, France
- Lyon-Est Faculty of Medicine, Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Ekaterine Berishvili
- Faculty Diabetes Center, University of Geneva Medical Center, University of Geneva, Geneva, Switzerland
- Institute of Medical and Public Health Research, Ilia State University, Tbilisi, Georgia
- Department of Surgery, Laboratory of Tissue Engineering and Organ Regeneration, University of Geneva, Geneva, Switzerland
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
29
|
Maruoka A, Kimura A, Hattori F, Hitomi H, Osafune K, Shiojima I, Toyoda N. Expression of genes involved in thyroid hormone action in human induced pluripotent stem cells during differentiation to insulin-producing cells: Effects of iopanoic acid on differentiation. Mol Cell Endocrinol 2025; 599:112490. [PMID: 39921130 DOI: 10.1016/j.mce.2025.112490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
AIMS Type 3 iodothyronine deiodinase (Dio3) converts triiodothyronine (T3) to diiodothyronine, thereby reducing intracellular T3 levels. In this study, we investigated the potential roles of Dio3 in the differentiation of human pancreatic β cells, using β cells derived from human induced pluripotent stem cells (hiPSCs). MAIN METHODS hiPSCs were differentiated to β cells in a stepwise manner over 29 days. The differentiation medium was supplemented with B27, which contains T3 but not T4, instead of serum. The T3 levels in the differentiated cells were determined based on the amount of T3 supplied to the medium and the activity of Dio3 within the cells. Iopanoic acid (IOP) was used as the Dio3 inhibitor. KEY FINDINGS Dio3 expression is substantially altered during differentiation. IOP treatment reduced Dio3 activity on day 4 and increased T3 levels in the medium on day 29. To investigate the involvement of Dio3 during differentiation, we used IOP, in which cells differentiated in the presence of IOP (+IOP) were compared to those differentiated without IOP (-IOP). On day 29, the proportion of β cells expressing C-peptide, NKX6 homeobox 1, and both markers was considerably higher in the presence than in the absence of IOP. Furthermore, on day 29, the insulin content of differentiated + IOP cells was considerably higher than that of differentiated -IOP cells. CONCLUSIONS An increase in intracellular T3 content promoted via the inhibition of Dio3 activity by IOP from day 0-29 enhances the differentiation of hiPSCs to β cells.
Collapse
Affiliation(s)
- Azusa Maruoka
- Department of Medicine II, Kansai Medical University, Osaka, 5731010, Japan
| | - Azuma Kimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 6068507, Japan
| | - Fumiyuki Hattori
- iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, 5731010, Japan
| | - Hirofumi Hitomi
- iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, 5731010, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 6068507, Japan
| | - Ichiro Shiojima
- Department of Medicine II, Kansai Medical University, Osaka, 5731010, Japan
| | - Nagaoki Toyoda
- Department of Medicine II, Kansai Medical University, Osaka, 5731010, Japan.
| |
Collapse
|
30
|
Liu P, Ponnienselvan K, Nyalile T, Oikemus S, Joynt AT, Iyer S, Kelly K, Guo D, Kyawe PP, Vanderleeden E, Redick SD, Huang L, Chen Z, Lee JM, Schiffer CA, Harlan DM, Wang JP, Emerson CP, Lawson ND, Watts JK, Sontheimer EJ, Luban J, Wolfe SA. Increasing intracellular dNTP levels improves prime editing efficiency. Nat Biotechnol 2025; 43:539-544. [PMID: 39322763 DOI: 10.1038/s41587-024-02405-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 08/27/2024] [Indexed: 09/27/2024]
Abstract
In primary cell types, intracellular deoxynucleotide triphosphate (dNTP) levels are tightly regulated in a cell cycle-dependent manner. We report that prime editing efficiency is increased by mutations that improve the enzymatic properties of Moloney murine leukemia virus reverse transcriptase and treatments that increase intracellular dNTP levels. In combination, these modifications produce substantial increases in precise editing rates.
Collapse
Affiliation(s)
- Pengpeng Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Karthikeyan Ponnienselvan
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Thomas Nyalile
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sarah Oikemus
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Anya T Joynt
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sukanya Iyer
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Karen Kelly
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dongsheng Guo
- Department of Neurology, Wellstone Program, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Pyae P Kyawe
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Emma Vanderleeden
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sambra D Redick
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lei Huang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Zexiang Chen
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeong Min Lee
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - David M Harlan
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jennifer P Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Charles P Emerson
- Department of Neurology, Wellstone Program, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nathan D Lawson
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jonathan K Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Erik J Sontheimer
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
31
|
Huang H, Ye K, Jin S. Cell Seeding Strategy Influences Metabolism and Differentiation Potency of Human Induced Pluripotent Stem Cells Into Pancreatic Progenitors. Biotechnol J 2025; 20:e70022. [PMID: 40285386 PMCID: PMC12032514 DOI: 10.1002/biot.70022] [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: 02/10/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Human induced pluripotent stem cells (iPSCs) are an invaluable endless cell source for generating various therapeutic cells and tissues. However, their differentiation into specific cell lineages, such as definitive endoderm (DE) and pancreatic progenitor (PP), often suffers from poor reproducibility, due partially to their pluripotency. In this work, we investigated the impact of iPSC confluency during cell self-renewal and seeding density on cell metabolic activity, glycolysis to oxidative phosphorylation shift, and differentiation potential toward DE and PP lineages. Our findings demonstrated that cell seeding strategy influences cellular metabolic activity and the robustness of iPSC differentiation. iPSCs maintained at higher seeding density exhibited lower initial oxygen consumption rate (OCR) and metabolic activity. There is an optimal seeding density to ensure sufficient oxygen consumption during differentiation and to yield high expression of SOX17 in the DE lineage and high PDX1/NKX6.1 dual-positive cells in PPs. Interestingly, we found that cell confluency at the time of harvest has less impact on the efficacy of pancreatic lineage formation or metabolic activity. This study sheds light on the interplay between metabolic activity and iPSC lineage specification, offering new insights into the robustness of iPSC self-renewal and differentiation for creating human tissues.
Collapse
Affiliation(s)
- Hui Huang
- Department of Biomedical Engineering Thomas J. Watson College of Engineering and Applied Science Binghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
| | - Kaiming Ye
- Department of Biomedical Engineering Thomas J. Watson College of Engineering and Applied Science Binghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
- Center of Biomanufacturing for Regenerative MedicineBinghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
| | - Sha Jin
- Department of Biomedical Engineering Thomas J. Watson College of Engineering and Applied Science Binghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
- Center of Biomanufacturing for Regenerative MedicineBinghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
| |
Collapse
|
32
|
Sali S, Azzam L, Jaro T, Ali AAG, Mardini A, Al-Dajani O, Khattak S, Butler AE, Azeez JM, Nandakumar M. A perfect islet: reviewing recent protocol developments and proposing strategies for stem cell derived functional pancreatic islets. Stem Cell Res Ther 2025; 16:160. [PMID: 40165291 PMCID: PMC11959787 DOI: 10.1186/s13287-025-04293-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
The search for an effective cell replacement therapy for diabetes has driven the development of "perfect" pancreatic islets from human pluripotent stem cells (hPSCs). These hPSC-derived pancreatic islet-like β cells can overcome the limitations for disease modelling, drug development and transplantation therapies in diabetes. Nevertheless, challenges remain in generating fully functional and mature β cells from hPSCs. This review underscores the significant efforts made by researchers to optimize various differentiation protocols aimed at enhancing the efficiency and quality of hPSC-derived pancreatic islets and proposes methods for their improvement. By emulating the natural developmental processes of pancreatic embryogenesis, specific growth factors, signaling molecules and culture conditions are employed to guide hPSCs towards the formation of mature β cells capable of secreting insulin in response to glucose. However, the efficiency of these protocols varies greatly among different human embryonic stem cell (hESC) and induced pluripotent stem cell (hiPSC) lines. This variability poses a particular challenge for generating patient-specific β cells. Despite recent advancements, the ultimate goal remains to develop a highly efficient directed differentiation protocol that is applicable across all genetic backgrounds of hPSCs. Although progress has been made, further research is required to optimize the protocols and characterization methods that could ensure the safety and efficacy of hPSC-derived pancreatic islets before they can be utilized in clinical settings.
Collapse
Affiliation(s)
- Sujitha Sali
- King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Leen Azzam
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Taraf Jaro
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Ahmed Ali Gebril Ali
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Ali Mardini
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Omar Al-Dajani
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Shahryar Khattak
- King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Alexandra E Butler
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain.
| | - Juberiya M Azeez
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Manjula Nandakumar
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| |
Collapse
|
33
|
Batir-Marin D, Ștefan CS, Boev M, Gurău G, Popa GV, Matei MN, Ursu M, Nechita A, Maftei NM. A Multidisciplinary Approach of Type 1 Diabetes: The Intersection of Technology, Immunotherapy, and Personalized Medicine. J Clin Med 2025; 14:2144. [PMID: 40217595 PMCID: PMC11989447 DOI: 10.3390/jcm14072144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Type 1 diabetes (T1D) is a chronic autoimmune disorder characterized by the destruction of pancreatic β-cells, leading to absolute insulin deficiency. Despite advancements in insulin therapy and glucose monitoring, achieving optimal glycemic control remains a challenge. Emerging technologies and novel therapeutic strategies are transforming the landscape of T1D management, offering new opportunities for improved outcomes. Methods: This review synthesizes recent advancements in T1D treatment, focusing on innovations in continuous glucose monitoring (CGM), automated insulin delivery systems, smart insulin formulations, telemedicine, and artificial intelligence (AI). Additionally, we explore biomedical approaches such as stem cell therapy, gene editing, immunotherapy, gut microbiota modulation, nanomedicine-based interventions, and trace element-based therapies. Results: Advances in digital health, including CGM integration with hybrid closed-loop insulin pumps and AI-driven predictive analytics, have significantly improved real-time glucose management. AI and telemedicine have enhanced personalized diabetes care and patient engagement. Furthermore, regenerative medicine strategies, including β-cell replacement, CRISPR-based gene editing, and immunomodulatory therapies, hold potential for disease modification. Probiotics and microbiome-targeted therapies have demonstrated promising effects in maintaining metabolic homeostasis, while nanomedicine-based trace elements provide additional strategies to regulate insulin sensitivity and oxidative stress. Conclusions: The future of T1D management is shifting toward precision medicine and integrated technological solutions. While these advancements present promising therapeutic avenues, challenges such as long-term efficacy, safety, accessibility, and clinical validation must be addressed. A multidisciplinary approach, combining biomedical research, artificial intelligence, and nanotechnology, will be essential to translate these innovations into clinical practice, ultimately improving the quality of life for individuals with T1D.
Collapse
Affiliation(s)
- Denisa Batir-Marin
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania; (D.B.-M.); (N.-M.M.)
| | - Claudia Simona Ștefan
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania
| | - Monica Boev
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania; (D.B.-M.); (N.-M.M.)
| | - Gabriela Gurău
- Department of Morphological and Functional Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania;
- Clinic Laboratory Department, Clinical Hospital of Children Hospital “Sf. Ioan”, 800487 Galati, Romania
| | - Gabriel Valeriu Popa
- Department of Dental Medicine, Faculty of Medicine and Pharmacy Galați, “Dunărea de Jos” University, 800008 Galati, Romania; (G.V.P.); (M.N.M.)
| | - Mădălina Nicoleta Matei
- Department of Dental Medicine, Faculty of Medicine and Pharmacy Galați, “Dunărea de Jos” University, 800008 Galati, Romania; (G.V.P.); (M.N.M.)
| | - Maria Ursu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania; (M.U.); (A.N.)
| | - Aurel Nechita
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania; (M.U.); (A.N.)
| | - Nicoleta-Maricica Maftei
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania; (D.B.-M.); (N.-M.M.)
- Clinic Laboratory Department, Clinical Hospital of Children Hospital “Sf. Ioan”, 800487 Galati, Romania
| |
Collapse
|
34
|
Mbaye EHA, Scott EA, Burke JA. From Edmonton to Lantidra and beyond: immunoengineering islet transplantation to cure type 1 diabetes. FRONTIERS IN TRANSPLANTATION 2025; 4:1514956. [PMID: 40182604 PMCID: PMC11965681 DOI: 10.3389/frtra.2025.1514956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
Type 1 diabetes (T1D) is characterized by the autoimmune destruction of insulin-producing β cells within pancreatic islets, the specialized endocrine cell clusters of the pancreas. Islet transplantation has emerged as a β cell replacement therapy, involving the infusion of cadaveric islets into a patient's liver through the portal vein. This procedure offers individuals with T1D the potential to restore glucose control, reducing or even eliminating the need for exogenous insulin therapy. However, it does not address the underlying autoimmune condition responsible for T1D. The need for systemic immunosuppression remains the primary barrier to making islet transplantation a more widespread therapy for patients with T1D. Here, we review recent progress in addressing the key limitations of islet transplantation as a viable treatment for T1D. Concerns over systemic immunosuppression arise from its potential to cause severe side effects, including opportunistic infections, malignancies, and toxicity to transplanted islets. Recognizing the risks, the Edmonton protocol (2000) marked a shift away from glucocorticoids to prevent β cell damage specifically. This transition led to the development of combination immunosuppressive therapies and the emergence of less toxic immunosuppressive and anti-inflammatory drugs. More recent advances in islet transplantation derive from islet encapsulation devices, biomaterial platforms releasing immunomodulatory compounds or surface-modified with immune regulating ligands, islet engineering and co-transplantation with accessory cells. While most of the highlighted studies in this review remain at the preclinical stage using mouse and non-human primate models, they hold significant potential for clinical translation if a transdisciplinary research approach is prioritized.
Collapse
Affiliation(s)
- El Hadji Arona Mbaye
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
- Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | |
Collapse
|
35
|
Song H, Li J, Yang H, Kong B, Xu Y, Li X, Li H. Enhancement of functional insulin-producing cell differentiation from embryonic stem cells through MST1-silencing. Diabetol Metab Syndr 2025; 17:93. [PMID: 40108649 PMCID: PMC11924671 DOI: 10.1186/s13098-025-01666-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Islet β-cell transplantation offers a promising treatment for repairing pancreatic damage in diabetes, with the transcription factor pancreatic duodenal homeobox-1 (PDX1) being crucial for β-cell function and insulin secretion. Mammalian threonine protein kinase (MST1) is recognized for its role in regulating PDX1 during cell apoptosis, yet its function in embryonic stem cell (ESC) differentiation into insulin-producing cells (IPCs) remain underexplored. This study investigated the effect of MST1-silencing on the differentiation of ESC into IPCs. METHODS ESCs were transfected utilizing a recombinant MST1-silencing lentiviral vector (shMST1). qRT-PCR, immunofluorescence, flow cytometry, western blot and ELISA assays were performed to examine function of IPCs in vitro. Furthermore, these IPCs were transplanted into type 1 diabetic mellitus (T1DM) rats. Measuring the changes in blood glucose concentration of animals before and after IPCs transplantation. Intraperitoneal glucose tolerance test (IPGT) was used to determine the regulatory effect of IPCs transplantation on blood glucose stimulation and immunohistochemistry was used to detect the expression of pancreatic Insulin protein in T1DM rats. RESULTS It was observed that IPCs from the shMST1 group exhibited notably improvement in insulin secretion and glucose responsiveness, suggesting MST1 suppression may enhance IPC maturity. The rats demonstrated significant normalization of blood sugar levels and increased insulin levels, akin to non-diabetic controls. This implies that MST1-silencing not only augments IPC function in vitro but also their therapeutic efficacy in vivo. CONCLUSIONS The findings indicate that targeting MST1 offers a novel approach for deriving functionally mature IPCs from ESCs, potentially advancing cell replacement therapies for diabetes. This research underscores the importance of developing IPCs with competent insulin secretion for diabetes treatment in vitro.
Collapse
Affiliation(s)
- Hui Song
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiarui Li
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Haohao Yang
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Bin Kong
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
| | - Yu Xu
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiong Li
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750004, China.
| | - Hui Li
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China.
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
36
|
Wang Y, McGarrigle J, Cook J, Rios P, Monica GL, Chen Y, Wei W, Oberholzer J. The future of islet transplantation beyond the BLA approval: challenges and opportunities. FRONTIERS IN TRANSPLANTATION 2025; 4:1522409. [PMID: 40124184 PMCID: PMC11925927 DOI: 10.3389/frtra.2025.1522409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025]
Abstract
This opinion paper explores the path forward for islet transplantation as a cell therapy for type 1 diabetes, following the Biologics License Application (BLA) approval. The authors review key challenges and opportunities that lie ahead. After a brief overview of the history of human islet transplantation, the paper examines the FDA's regulatory stance on isolated islet cells and the requirements for obtaining a BLA. The authors discuss the significance of this approval and the critical steps necessary to broaden patient access, such as scaling up production, clinical integration, reimbursement frameworks, post-marketing surveillance, and patient education initiatives. The paper highlights that the approval of LANTIDRA as an allogeneic cell transplant for uncontrolled type 1 diabetes marks the beginning of new chapters in improving islet transplantation. The authors emphasize essential areas for development, including advancements in islet manufacturing, optimization of transplant sites, islet encapsulation, exploration of unlimited cell sources, and gene editing technologies. In conclusion, the future of islet transplantation beyond the BLA approval presents challenges and opportunities. While significant regulatory milestones have been reached, hurdles remain. Innovations in stem cell-derived islets, cell encapsulation, and gene editing show promise in enhancing graft survival, expanding the availability of transplantable cells, and reducing the reliance on immunosuppressive drugs. These advancements could pave the way for more accessible, durable, and personalized diabetes treatments.
Collapse
Affiliation(s)
- Yong Wang
- Clinic of Visceral and Transplant Surgery, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zürich, Switzerland
- CellTrans, Inc., Chicago, IL, United States
| | | | - Jenny Cook
- CellTrans, Inc., Chicago, IL, United States
| | - Peter Rios
- CellTrans, Inc., Chicago, IL, United States
| | | | - Yingying Chen
- Clinic of Visceral and Transplant Surgery, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zürich, Switzerland
| | - Wei Wei
- Clinic of Visceral and Transplant Surgery, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zürich, Switzerland
| | - Jose Oberholzer
- Clinic of Visceral and Transplant Surgery, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zürich, Switzerland
- CellTrans, Inc., Chicago, IL, United States
| |
Collapse
|
37
|
Waite EL, Tigue M, Yu M, Lahori D, Kelly K, May CL, Naji A, Roman J, Doliba N, Avrahami D, Nguyen-Ngoc KV, Sander M, Glaser B, Kaestner KH. The IsletTester Mouse: An Immunodeficient Model With Stable Hyperglycemia for the Study of Human Islets. Diabetes 2025; 74:332-342. [PMID: 39571094 PMCID: PMC11842601 DOI: 10.2337/db23-0887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/18/2024] [Indexed: 02/22/2025]
Abstract
The gold standard for assessing the function of human islets or β-like cells derived from stem cells involves their engraftment under the kidney capsule of hyperglycemic, immunodeficient mice. Current models, such as streptozotocin treatment of severely immunodeficient mice or the NRG-Akita strain, are limited due to unstable and variable hyperglycemia and/or high morbidity. To address these limitations, we developed the IsletTester mouse via CRISPR/Cas9-mediated gene editing of glucokinase (Gck), the glucose sensor of the β-cells, directly in NSG zygotes. IsletTester mice are heterozygous for an Arg345→stop mutation in Gck and present with stable random hyperglycemia (∼250 mg/dL [14 mmol/L]), normal lifespan, and fertility. We demonstrate the utility of this model through functional engraftment of both human islets and human embryonic stem cell-derived β-like cells. The IsletTester mouse will enable the study of human islet biology over time and under different physiological conditions and can provide a useful preclinical platform to determine the functionality of stem cell-derived islet products. ARTICLE HIGHLIGHTS Current mouse models for assessing islet function in vivo are limited due to unstable and variable hyperglycemia and/or high morbidity. We derived the IsletTester mouse to address these limitations. Leveraging a previously characterized glucokinase mutation and CRISPR/Cas9 technology, we successfully developed a moderately hyperglycemic and immunodeficient mouse model for the in vivo assessment of islet function. Our IsletTester mouse has stable, moderate hyperglycemia that can be corrected with primary human islets or stem cell-derived insulin-producing cells. The IsletTester mouse provides a reliable, easy-to-use platform for the preclinical assessment of stem cell-derived islet products or islet-targeted drugs.
Collapse
Affiliation(s)
- Eric L. Waite
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mark Tigue
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ming Yu
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Deeksha Lahori
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kai Kelly
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Catherine Lee May
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ali Naji
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Jeffrey Roman
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nicolai Doliba
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Dana Avrahami
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Kim-Vy Nguyen-Ngoc
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA
| | - Maike Sander
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA
- Max Delbruck Center, Berlin, Germany
| | - Benjamin Glaser
- Department of Endocrinology and Metabolism, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Klaus H. Kaestner
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
38
|
Jacques K, Coles BLK, van der Kooy D. Pancreatic stem cells originate during the pancreatic progenitor developmental stage. Front Cell Dev Biol 2025; 13:1521411. [PMID: 40040790 PMCID: PMC11876382 DOI: 10.3389/fcell.2025.1521411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Previously isolated adult pancreatic precursors called pancreatic multipotent progenitors (which make both pancreatic endocrine and exocrine cell types) originate from the Pancreatic Duodenal Homeobox 1 (PDX1) pancreatic developmental lineage. The embryonic time point at which adult pancreatic multipotent progenitor cells emerge has not been established. We have employed the use of two models: a human embryonic stem cell (hESC) to beta-cell cytokine-induced differentiation protocol and a mouse lineage tracing model during early development to isolate clonal pancreatic spheres. The results show that insulin-positive clonal spheres can be isolated as early as the pancreatic endoderm stage as well as the pancreatic progenitor stage during the hESC to beta-cell lineage differentiation model and that they can be isolated only as early as the pancreatic progenitor stage during mouse embryogenesis. Further, pancreatic clonal sphere-forming cells isolated from the pancreatic progenitor stage in embryonic mice display multipotentiality, and those isolated at a later gestational age demonstrate self-renewal ability. These findings suggest that pancreatic precursors isolated from mouse embryonic time points have stem cell properties and that the pancreatic progenitor stage in hESC development may be the optimal time to capture and expand these stem cells and make large numbers of beta cells.
Collapse
Affiliation(s)
- Krystal Jacques
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Brenda L. K. Coles
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Derek van der Kooy
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Campo F, Neroni A, Pignatelli C, Pellegrini S, Marzinotto I, Valla L, Manenti F, Policardi M, Lampasona V, Piemonti L, Citro A. Bioengineering of a human iPSC-derived vascularized endocrine pancreas for type 1 diabetes. Cell Rep Med 2025; 6:101938. [PMID: 39922198 PMCID: PMC11866511 DOI: 10.1016/j.xcrm.2025.101938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 11/18/2024] [Accepted: 01/13/2025] [Indexed: 02/10/2025]
Abstract
Intrahepatic islet transplantation in patients with type 1 diabetes is limited by donor availability and lack of engraftment. Alternative β cell sources and transplantation sites are needed. We demonstrate the feasibility to repurpose a decellularized lung as an endocrine pancreas for β cell replacement. We bioengineer an induced pluripotent stem cell (iPSC)-based version, fabricating a human iPSC-based vascularized endocrine pancreas (iVEP) using iPSC-derived β cells (iPSC-derived islets [SC-islets]) and endothelial cells (iECs). SC-islets and iECs are aggregated into vascularized iβ spheroids (ViβeSs), and over 7 days of culture, spheroids integrate into the bioengineered vasculature, generating a functional, perfusable human endocrine organ. In vitro, the vascularized extracellular matrix (ECM) sustained SC-islet engraftment and survival with a significantly preserved β cell mass and a physiologic insulin release. In vivo, iVEP restores normoglycemia in diabetic NSG mice. We report a human iVEP providing a controlled in vitro insulin-secreting phenotype and in vivo function.
Collapse
Affiliation(s)
- 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
| | - Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Pellegrini
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Marzinotto
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Libera Valla
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
| | - Fabio Manenti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Policardi
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vito Lampasona
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, 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
|
40
|
Yano T, Shimaya Y, Enomoto T, Kiho T, Komoriya S, Nakashima R, Shiraki N, Kume S. A small molecule K-3 promotes PDX1 expression and potentiates the differentiation of pluripotent stem cells into insulin-producing pancreatic β cells. Stem Cells 2025; 43:sxae075. [PMID: 39556137 DOI: 10.1093/stmcls/sxae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
Insulin-producing pancreatic β-like cells derived from human pluripotent stem cells (PSCs) are anticipated as a novel cell source for cell replacement therapy for patients with diabetes. Here, we describe the identification of small molecule compounds that promote the differentiation of the PSCs into insulin-producing cells by high throughput screening with a chemical library composed of 55 000 compounds. The initial hit compound K-1 and one derivative K-3 increased the proportion of PSC-derived insulin-positive endocrine cells and their glucose-stimulated insulin secretory (GSIS) functions. K-3 preferentially acts on stage 3 pancreatic progenitor cells and increases the population expressing high levels of PDX1. As a result, the ratios of the PSC-derived PDX1/NKX6.1 double-positive endocrine progenitor and INS/NKX6.1 double-positive mono-hormonal endocrine cells were increased. K-3 enhances the expression of functional pancreatic β cell markers and affects biological processes concerning organ development. K-3 also increased the yield of endocrine cells at the end of stage 5. The novel compound is a beneficial new tool for efficiently generating PSC-derived insulin-producing cells with high functionality and differentiation efficiency.
Collapse
Affiliation(s)
- Tatsuya Yano
- Daiichi Sankyo Co., Ltd., Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yukihiro Shimaya
- Department of Life Science and Technology, School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Kanagawa 226-8501, Japan
| | - Takayuki Enomoto
- Department of Life Science and Technology, School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Kanagawa 226-8501, Japan
- Bioscience Center, Research Infrastructure Management Center, Institute of Science Tokyo, Yokohama, Kanagawa 226-8501, Japan
| | - Toshihiro Kiho
- Daiichi Sankyo Co., Ltd., Shinagawa-ku, Tokyo 140-8710, Japan
| | | | | | - Nobuaki Shiraki
- Department of Life Science and Technology, School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Kanagawa 226-8501, Japan
| | - Shoen Kume
- Department of Life Science and Technology, School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Kanagawa 226-8501, Japan
| |
Collapse
|
41
|
Hogrebe NJ, Schmidt MD, Augsornworawat P, Gale SE, Shunkarova M, Millman JR. Depolymerizing F-actin accelerates the exit from pluripotency to enhance stem cell-derived islet differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.21.618465. [PMID: 39484596 PMCID: PMC11526947 DOI: 10.1101/2024.10.21.618465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In this study, we demonstrate that cytoskeletal state at the onset of directed differentiation is critical for the specification of human pluripotent stem cells (hPSCs) to all three germ layers. In particular, a polymerized actin cytoskeleton facilitates directed ectoderm differentiation, while depolymerizing F-actin promotes mesendoderm lineages. Applying this concept to a stem cell-derived islet (SC-islet) differentiation protocol, we show that depolymerizing F-actin with latrunculin A (latA) during the first 24 hours of definitive endoderm formation facilitates rapid exit from pluripotency and alters Activin/Nodal, BMP, JNK-JUN, and WNT pathway signaling dynamics. These signaling changes influence downstream patterning of the gut tube, leading to improved pancreatic progenitor identity and decreased expression of markers associated with other endodermal lineages. Continued differentiation generates islets containing a higher percentage of β cells that exhibit improved maturation, insulin secretion, and ability to reverse hyperglycemia. Furthermore, this latA treatment reduces enterochromaffin cells in the final cell population and corrects differentiations from hPSC lines that otherwise fail to consistently produce pancreatic islets, highlighting the importance of cytoskeletal signaling at the onset of directed differentiation.
Collapse
Affiliation(s)
- Nathaniel J. Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mason D. Schmidt
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Punn Augsornworawat
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sarah E. Gale
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mira Shunkarova
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey R. Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA
| |
Collapse
|
42
|
Mima A, Kimura A, Ito R, Hatano Y, Tsujimoto H, Mae SI, Yamane J, Fujibuchi W, Uza N, Toyoda T, Seno H, Osafune K. Mechanistic elucidation of human pancreatic acinar development using single-cell transcriptome analysis on a human iPSC differentiation model. Sci Rep 2025; 15:4668. [PMID: 39920294 PMCID: PMC11806057 DOI: 10.1038/s41598-025-88690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
Few effective treatments have been developed for intractable pancreatic exocrine disorders due to the lack of suitable disease models using human cells. Pancreatic acinar cells differentiated from human induced pluripotent stem cells (hiPSCs) have the potential to solve this issue. In this study, we aimed to elucidate the developmental mechanisms of pancreatic exocrine acinar lineages to establish a directed differentiation method for pancreatic acinar cells from hiPSCs. hiPSC-derived pancreatic endoderm cells were spontaneously differentiated into both pancreatic exocrine and endocrine tissues by implantation into the renal subcapsular space of NOD/SCID mice. Single-cell RNA-seq analysis of the retrieved grafts confirmed the differentiation of pancreatic acinar lineage cells and identified REG4 as a candidate marker for pancreatic acinar progenitor cells. Furthermore, differential gene expression analysis revealed upregulated pathways, including cAMP-related signals, involved in the differentiation of hiPSC-derived pancreatic acinar lineage cells in vivo, and we found that a cAMP activator, forskolin, facilitates the differentiation from hiPSC-derived pancreatic endoderm into pancreatic acinar progenitor cells in our in vitro differentiation culture. Therefore, this platform contributes to our understanding of the developmental mechanisms of pancreatic acinar lineage cells and the establishment of differentiation methods for acinar cells from hiPSCs.
Collapse
Affiliation(s)
- Atsushi Mima
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Azuma Kimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ryo Ito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yu Hatano
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Junko Yamane
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Wataru Fujibuchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
43
|
Kim M, Cho S, Hwang DG, Shim IK, Kim SC, Jang J, Jang J. Bioprinting of bespoke islet-specific niches to promote maturation of stem cell-derived islets. Nat Commun 2025; 16:1430. [PMID: 39920133 PMCID: PMC11805982 DOI: 10.1038/s41467-025-56665-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Pancreatic islets are densely packed cellular aggregates containing various hormonal cell types essential for blood glucose regulation. Interactions among these cells markedly affect the glucoregulatory functions of islets along with the surrounding niche and pancreatic tissue-specific geometrical organization. However, stem cell (SC)-derived islets generated in vitro often lack the three-dimensional extracellular microenvironment and peri-vasculature, which leads to the immaturity of SC-derived islets, reducing their ability to detect glucose fluctuations and insulin release. Here, we bioengineer the in vivo-like pancreatic niches by optimizing the combination of pancreatic tissue-specific extracellular matrix and basement membrane proteins and utilizing bioprinting-based geometrical guidance to recreate the spatial pattern of islet peripheries. The bioprinted islet-specific niche promotes coordinated interactions between islets and vasculature, supporting structural and functional features resembling native islets. Our strategy not only improves SC-derived islet functionality but also offers significant potential for advancing research on islet development, maturation, and diabetic disease modeling, with future implications for translational applications.
Collapse
Affiliation(s)
- Myungji Kim
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Seungyeun Cho
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dong Gyu Hwang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - In Kyong Shim
- Asan Institute for Life Science, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Song Cheol Kim
- Asan Institute for Life Science, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Jiwon Jang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinah Jang
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Mu-u-min RBA, Diane A, Allouch A, Al-Siddiqi HH. Immune Evasion in Stem Cell-Based Diabetes Therapy-Current Strategies and Their Application in Clinical Trials. Biomedicines 2025; 13:383. [PMID: 40002796 PMCID: PMC11853723 DOI: 10.3390/biomedicines13020383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Human pancreatic islet transplantation shows promise for long-term glycemic control in diabetes patients. A shortage of healthy donors and the need for continuous immunosuppressive therapy complicates this. Enhancing our understanding of the immune tolerance mechanisms related to graft rejection is crucial to generate safer transplantation strategies. This review will examine advancements in immune protection strategies for stem cell-derived islet therapy and discuss key clinical trials involving stem cell-derived β-cells and their protective strategies against the host immune system. Methods: A comprehensive literature search was performed on peer-reviewed publications on Google Scholar, Pubmed, and Scopus up to September 2024 to extract relevant studies on the various strategies of immune evasion of stem cell-derived β-cells in humans. The literature search was extended to assimilate all relevant clinical studies wherein stem cell-derived β-cells are transplanted to treat diabetes. Results: Our analysis highlighted the importance of human pluripotent stem cells (hPSCs) as a potentially unlimited source of insulin-producing β-cells. These cells can be transplanted as an effective source of insulin in diabetes patients if they can be protected against the host immune system. Various strategies of immune protection, such as encapsulation and genetic manipulation, are currently being studied and clinically tested. Conclusions: Investigating immune tolerance in hPSC-derived islets may help achieve a cure for diabetes without relying on exogenous insulin. Although reports of clinical trials show promise in reducing insulin dependency in patients, their safety and efficacy needs to be further studied to promote their use as a long-term solution to cure diabetes.
Collapse
Affiliation(s)
- Razik Bin Abdul Mu-u-min
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar; (A.D.); (H.H.A.-S.)
| | - Abdoulaye Diane
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar; (A.D.); (H.H.A.-S.)
| | - Asma Allouch
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar;
| | - Heba Hussain Al-Siddiqi
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar; (A.D.); (H.H.A.-S.)
| |
Collapse
|
45
|
Schaaf C, Sussel L. A Cure for Type 1 Diabetes: Are We There Yet? Diabetes Technol Ther 2025. [PMID: 39911033 DOI: 10.1089/dia.2024.0498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Type 1 diabetes (T1D) affects over 2 million people in the United States and has no known cure. The discovery and first use of insulin in humans 102 years ago marked a revolutionary course of treatment for the disease, and although the formulations and delivery systems have advanced, insulin administration remains the standard of care today. While improved treatment options represent notable progress in T1D management, finding a functional cure for the disease remains the ultimate goal. Approaches to curing T1D have historically focused on blunting the autoimmune response, although sustained effects of immune modulation have proven elusive. Islet transplant therapies have also proven effective, although a lack of available donor tissue and the need for immunosuppression to prevent both host-graft rejection and the autoimmune response have reserved such treatments for those who already require immunosuppressive regimens for other reasons or undergo severe hypoglycemic events in conjunction with hypoglycemic unawareness. With the advent of human stem cell research, the focus has shifted toward generating an abundance of allogeneic, functional beta-like cells that can be transplanted into the patients. Immunoisolation devices have also shown some promise as a method of preventing immune rejection and the autoimmune destruction of transplanted cells. Finally, advances in new immune therapies, if used in the early stages of T1D progression, have proven to delay the onset of diabetes. Stem cell-based therapies are a promising approach to curing T1D. The ongoing clinical trials show some success, although they currently require immunosuppressant agents. Encapsulation devices provide a method of immunoisolation that does not require immunosuppression; however, the devices tested thus far eventually lead to cell death and fibrotic tissue growth. Substantial research efforts are underway to develop new approaches to protect the stem cell-derived beta cells upon transplantation.
Collapse
Affiliation(s)
- Christopher Schaaf
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Center, Denver, Colorado, USA
| | - Lori Sussel
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Center, Denver, Colorado, USA
| |
Collapse
|
46
|
Schulze HG, Rangan S, Vardaki MZ, Blades MW, Turner RFB, Piret JM. Demixing and Analysis of Complex Biological Raman Hyperspectra Based on Peak Fitting, Amplitude Trend Clustering, and Spectrum Reconstruction. APPLIED SPECTROSCOPY 2025:37028241311296. [PMID: 39894915 DOI: 10.1177/00037028241311296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
To better interpret the Raman spectra from mammalian cells, it is often desirable to reduce their complexity by decomposing them into the spectral contributions from individual macromolecules or types of macromolecules. Diverse methods exist for demixing complex spectra, each with different benefits and drawbacks. However, some methods require a library of component spectra that might not be available, while others are hampered by noise and peak congestion that includes many proximal overlapping peaks. Through rapid fitting of individual peaks in every spectrum of a Raman hyperspectral data set, we have obtained individual peak parameters from which we determined the trends for all the peak amplitudes. We then grouped similar trends with k-means clustering. Then we used the peak parameters of all the peaks in a given cluster to reconstruct a spectrum representative of that cluster. This method produced spectra that were less distorted by unrelated overlapping peaks or noise, were less congested than those in the hyperspectral set, and thereby improved peak identification and macromolecule recognition. We have demonstrated the application of the method with Raman spectra from a perchlorate-polystyrene model system and extended it to complex spectra from methanol-fixed mammalian cells. We were able to recover independent spectra of perchlorate and polystyrene in the model system and spectra pertaining to individual macromolecular types (proteins, nucleic acids, lipids) from the mammalian cell data. We discuss how imperfections in spectral preprocessing and peak fitting can adversely affect the results. In summary, we have provided a proof-of-concept for a novel mixture resolution method with different attributes than extant ones.
Collapse
Affiliation(s)
| | - Shreyas Rangan
- Michael Smith Laboratories, The University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Martha Z Vardaki
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Michael W Blades
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Robin F B Turner
- Michael Smith Laboratories, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Electrical and Computer Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| | - James M Piret
- Michael Smith Laboratories, The University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
47
|
Farhat C, Xega V, Liu JL. A stepwise approach to deriving functional β-cells from human embryonic or induced pluripotent stem cells. MEDICAL REVIEW (2021) 2025; 5:23-34. [PMID: 39974557 PMCID: PMC11834748 DOI: 10.1515/mr-2024-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/05/2024] [Indexed: 02/21/2025]
Abstract
Our understanding of β-cell differentiation from pluripotent stem cells (PSCs) is rapidly evolving. Although progress has been made, challenges remain, particularly in achieving glucose-stimulated insulin secretion (GSIS). Human embryonic stem cells (hESCs) are valuable due to their pluripotent ability. A fixed protocol targeting master regulatory genes initiates stem cells into pancreatic lineage commitment. Due to the observations that a single stem cell can differentiate into multiple cell types depending on various factors and conditions, non-linear differentiation pathways exist. Co-expression of key factors remains essential for successful β-cell differentiation. The mature β-cell marker MAFA plays a critical role in maintaining the differentiation state and preventing dedifferentiation. Recapitulating pancreatic islet clustering enhances physiological responses, offering potential avenues for diabetes treatment. On the other hand, several enhanced differentiation protocols from induced pluripotent stem cells (iPSCs) have improved the functional insulin producing β-cells generated. These findings, with their potential to revolutionize diabetes treatment, highlight the complexity of β-cell differentiation and guide further advancements in regenerative medicine.
Collapse
Affiliation(s)
- Clara Farhat
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, Canada
- Department of Medicine, Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Viktoria Xega
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, Canada
- Department of Medicine, Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Jun-Li Liu
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, Canada
- Department of Medicine, Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| |
Collapse
|
48
|
Blaszczyk K, Jedrzejak AP, Ziojla N, Shcheglova E, Szarafin K, Jankowski A, Beamish CA, Chmielowiec J, Sabek OM, Balasubramanyam A, Patel S, Borowiak M. SPOCK2 controls the proliferation and function of immature pancreatic β-cells through MMP2. Exp Mol Med 2025; 57:131-150. [PMID: 39741186 PMCID: PMC11799530 DOI: 10.1038/s12276-024-01380-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 01/02/2025] Open
Abstract
Human pluripotent stem cell-derived β-cells (SC-β-cells) represent an alternative cell source for transplantation in diabetic patients. Although mitogens could in theory be used to expand β-cells, adult β-cells very rarely replicate. In contrast, newly formed β-cells, including SC-β-cells, display higher proliferative capacity and distinct transcriptional and functional profiles. Through bidirectional expression modulation and single-cell RNA-seq, we identified SPOCK2, an ECM protein, as an inhibitor of immature β-cell proliferation. Human β-cells lacking SPOCK2 presented elevated MMP2 expression and activity, leading to β-integrin-FAK-c-JUN pathway activation. Treatment with the MMP2 protein resulted in pronounced short- and long-term SC-β-cell expansion, significantly increasing glucose-stimulated insulin secretion in vitro and in vivo. These findings suggest that SPOCK2 mediates fetal β-cell proliferation and maturation. In summary, we identified a molecular mechanism that specifically regulates SC-β-cell proliferation and function, highlighting a unique signaling milieu of SC-β-cells with promise for the robust derivation of fully functional cells for transplantation.
Collapse
Affiliation(s)
- Katarzyna Blaszczyk
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, Poznan, 61-614, Poland
| | - Anna P Jedrzejak
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, Poznan, 61-614, Poland
| | - Natalia Ziojla
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, Poznan, 61-614, Poland
| | - Ekaterina Shcheglova
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, Poznan, 61-614, Poland
| | - Karolina Szarafin
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, Poznan, 61-614, Poland
| | - Artur Jankowski
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, Poznan, 61-614, Poland
| | - Christine A Beamish
- Department of Surgery, Methodist Research Institute, Houston, TX, 77030, USA
| | - Jolanta Chmielowiec
- Collegium Medicum, University of Warmia and Mazury, Aleja Warszawska 30, Olsztyn, 11-082, Poland
| | - Omaima M Sabek
- Department of Surgery, Methodist Research Institute, Houston, TX, 77030, USA
| | - Ashok Balasubramanyam
- Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sanjeet Patel
- Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, CA, 90033, USA
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, Poznan, 61-614, Poland.
- Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
49
|
Liu X, Zhang F, Chen D, Yin J, Bi Z, Chen L, Yan J, Dong Q, Peng W, Xu T, Guo Y, Lin H, Liu H. Generation of INS-jGCaMP7f knock-in Ca 2+ reporter human embryonic stem cell line, GZLe001-C, using CRISPR/Cas9-based gene targeting. Stem Cell Res 2025; 82:103633. [PMID: 39708407 DOI: 10.1016/j.scr.2024.103633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
As a member of the single-fluorophore genetically encoded calcium indicators (GECIs), jGCaMP7f is widely applied to investigate intracellular Ca2+ concentrations. Here, we established an INS-jGCaMP7f knock-in H1 human embryonic stem cell (hESC) line by integrating jGCaMP7f gene into insulin locus via CRISPR/Cas9 system. The reporter cell line not only effectively labelled the insulin-producing cells induced from hESC, but also reflected the cytosolic change of Ca2+ level in response to different stimuli. This reporter cell line is a valuable research tool for studying functional maturation of hESC-derived insulin-producing cells, conducting drug screenings, and exploring the mechanisms of diabetes.
Collapse
Affiliation(s)
- Xin Liu
- Department of Endocrinology and Metabolism, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi 830001, China; Guangzhou National Laboratory, Guangzhou 510005, China
| | - Feng Zhang
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Deqi Chen
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Jiaxiang Yin
- Guangzhou National Laboratory, Guangzhou 510005, China; School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, China
| | - Zirong Bi
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Lihua Chen
- Guangzhou National Laboratory, Guangzhou 510005, China; School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, China
| | - Jiawei Yan
- Guangzhou National Laboratory, Guangzhou 510005, China; School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, China
| | - Qifei Dong
- Guangzhou National Laboratory, Guangzhou 510005, China; Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wei Peng
- Guangzhou National Laboratory, Guangzhou 510005, China; Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Tao Xu
- Guangzhou National Laboratory, Guangzhou 510005, China; School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, China
| | - Yanying Guo
- Department of Endocrinology and Metabolism, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi 830001, China
| | - Haopeng Lin
- Guangzhou National Laboratory, Guangzhou 510005, China; School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, China
| | - Huisheng Liu
- Guangzhou National Laboratory, Guangzhou 510005, China; School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, China; Bioland Laboratory, Guangzhou 510005, China
| |
Collapse
|
50
|
Tran HT, Rodprasert W, Padeta I, Oontawee S, Purbantoro SD, Thongsit A, Siriarchavatana P, Srisuwatanasagul S, Egusa H, Osathanon T, Sawangmake C. Establishment of subcutaneous transplantation platform for delivering induced pluripotent stem cell-derived insulin-producing cells. PLoS One 2025; 20:e0318204. [PMID: 39883721 PMCID: PMC11781742 DOI: 10.1371/journal.pone.0318204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025] Open
Abstract
Potential trend of regenerative treatment for type I diabetes has been introduced for more than a decade. However, the technologies regarding insulin-producing cell (IPC) production and transplantation are still being developed. Here, we propose the potential IPC production protocol employing mouse gingival fibroblast-derived induced pluripotent stem cells (mGF-iPSCs) as a resource and the pre-clinical approved subcutaneous IPC transplantation platform for further clinical confirmation study. With a multi-step induction protocol, the functional and matured IPCs were generated by 13 days with a long-term survival capability. Further double encapsulation of mGF-iPSC-derived IPCs (mGF-iPSC-IPCs) could preserve the insulin secretion capacity and the transplantation potential of the generated IPCs. To address the potential on IPC transplantation, a 2-step subcutaneous transplantation procedure was established, comprising 1) vascularized subcutaneous pocket formation and 2) encapsulated IPC bead transplantation. The in vivo testing confirmed the safety and efficiency of the platform along with less inflammatory response which may help minimize tissue reaction and graft rejection. Further preliminary in vivo testing on subcutaneous IPC-bead transplantation in an induced type I diabetic mouse model showed beneficial trends on blood glucose control and survival rate sustainability of diabetic mice. Taken together, an established mGF-iPSC-IPC generation protocol in this study will be the potential backbone for developing the iPSC-derived IPC production employing human and animal cell resources. As well as the potential further development of IPC transplantation platform for diabetes treatment in human and veterinary practices using an established subcutaneous encapsulated IPC-bead transplantation platform presented in this study.
Collapse
Affiliation(s)
- Hong Thuan Tran
- Second Century Fund (C2F) Chulalongkorn University for Doctoral Scholarship, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, The International Graduate Program of Veterinary Science and Technology (VST), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Watchareewan Rodprasert
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Irma Padeta
- Second Century Fund (C2F) Chulalongkorn University for Doctoral Scholarship, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, The International Graduate Program of Veterinary Science and Technology (VST), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Saranyou Oontawee
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Steven dwi Purbantoro
- Second Century Fund (C2F) Chulalongkorn University for Doctoral Scholarship, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, The International Graduate Program of Veterinary Science and Technology (VST), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Anatcha Thongsit
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Parkpoom Siriarchavatana
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Medicine, Western University, Kanchanaburi, Thailand
| | - Sayamon Srisuwatanasagul
- Faculty of Veterinary Science, Department of Anatomy, Chulalongkorn University, Bangkok, Thailand
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Thanaphum Osathanon
- Faculty of Dentistry, Dental Stem Cell Biology Research Unit and Department of Anatomy, Chulalongkorn University, Bangkok, Thailand
- Faculty of Dentistry, Center of Excellence in Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chenphop Sawangmake
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Department of Pharmacology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|