1
|
Song H, Du X, Zhang Y, Liu W, Luo Y, Liu Y, Xue Y, Xu M, Lu J, Jia W, Du Y, Cao L, Lu J, Zhang W, He Z. Gelatin sponge patch grafting of microcryogel-based three-dimensional mesenchymal stem cells to alleviate acute liver failure. Biomaterials 2025; 321:123324. [PMID: 40253733 DOI: 10.1016/j.biomaterials.2025.123324] [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: 01/09/2025] [Revised: 03/24/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
The clinical application of human umbilical cord mesenchymal stem cells (hUCMSCs) in the treatment of liver failure faces challenges due to cell quality, short engraftment time, and limited efficacy. Here, gelatin microcryogel (GM) microcarriers with pore sizes ranging from 15 to 36 μm were tuned from mixed gelatin and glutaraldehyde to develop a 3D culture system of hUCMSCs with improved therapeutic effects. Bulk RNA sequencing and in vitro assays showed that 3D-hUCMSCs exhibited significant improvement in signaling pathways related to paracrine secretion and anti-inflammation. These 3D-hUCMSCs superior compared to 2D-hUCMSCs not only in terms of paracrine secretion, protection from oxidation, and resistance to mechanical force damage, but also had better liver function improvement effect than 2D-hUCMSCs when they were transplanted as single cells into liver injury mice. Furthermore, a gelatin sponge patch grafting (GSPG) strategy was developed to enable the direct engraftment of 3D-hUCMSCs within the GM microcarriers. The results showed that overall engraftment in the host liver was significantly improved, and the life span of transplanted hosts was extended. Our study provided a practical strategy to achieve high engraftment and long retraining time of 3D-hUCMSCs in rescuing acute liver failure with gelatin matrixes.
Collapse
Affiliation(s)
- Haimeng Song
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Xinyue Du
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Yuanyuan Zhang
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing, 100195, PR China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing, 100195, PR China
| | - Yi Luo
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Yuxin Liu
- Jinzhou Medical University, Jinzhou, Liaoning, 121001, PR China
| | - Yongjia Xue
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Mingyang Xu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Jizhen Lu
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Yanan Du
- School of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, PR China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 201619, PR China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 201619, PR China.
| | - Wencheng Zhang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China.
| | - Zhiying He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China.
| |
Collapse
|
2
|
Emerson AE, Lyons Q, Becker MW, Sepulveda K, Hiremath SC, Brady SR, Chilimba C, Weaver JD. Hydrogel injection molded complex macroencapsulation device geometry improves long-term cell therapy viability and function in the rat omentum transplant site. Biomaterials 2025; 317:123040. [PMID: 39754965 PMCID: PMC11788073 DOI: 10.1016/j.biomaterials.2024.123040] [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: 09/06/2024] [Revised: 12/08/2024] [Accepted: 12/21/2024] [Indexed: 01/06/2025]
Abstract
Insulin-secreting allogeneic cell therapies are a promising treatment for type 1 diabetes, with the potential to eliminate hypoglycemia and long-term complications of the disease. However, chronic systemic immunosuppression is necessary to prevent graft rejection, and the acute risks associated with immunosuppression limit the number of patients who can be treated with allogeneic cell therapies. Islet macroencapsulation in a hydrogel biomaterial is one proposed method to reduce or eliminate immune suppression; however, macroencapsulation devices suffer from poor oxygen transport and limited efficacy as they scale to large animal model preclinical studies and clinical trials. Hydrogel geometric device designs that optimize nutrient transport combined with methods to promote localized vasculogenesis may improve in vivo macroencapsulated cell viability and function. Here, we demonstrate with finite element modeling that a high surface area-to-volume ratio spiral geometry can increase macroencapsulated islet viability and function relative to a traditional cylindrical design, and we validate these observations in vitro under normoxic and physiological oxygen conditions. Finally, we evaluate macroencapsulated syngeneic islet survival and function in vivo in a diabetic rat omentum transplant model, and demonstrate that high surface area-to-volume hydrogel device designs improved macroencapsulated syngeneic islet function relative to traditional device designs.
Collapse
Affiliation(s)
- Amy E Emerson
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA
| | - Quincy Lyons
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA
| | - Matthew W Becker
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA
| | - Keven Sepulveda
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA
| | - Sarah R Brady
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA
| | - Chishiba Chilimba
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, 550 East Orange St., Tempe, AZ, 85281, USA.
| |
Collapse
|
3
|
Paz-Barba M, Peters IJ, de Graaf N, Carlotti F, de Koning EJ. Methylprednisolone Protects Human Pancreatic Beta Cells From Inflammation-induced Damage. Transplant Direct 2025; 11:e1770. [PMID: 40371054 PMCID: PMC12073852 DOI: 10.1097/txd.0000000000001770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 05/16/2025] Open
Abstract
Background Methylprednisolone is a glucocorticoid often used for immunosuppressive induction therapy or treatment of rejection in the context of organ transplantation and preservation of long-term function. In pancreas and islet transplantation, there is more reluctance to use high-dose methylprednisolone when there is suspicion of rejection, partly due to its hyperglycemic effects and doubts about the rescue of islet function. Here we investigated the functional and molecular effects of high-dose methylprednisolone on human pancreatic beta cells in an inflammatory environment, focusing on the nuclear factor kappa B and endoplasmic reticulum stress pathways. Methods We exposed primary human islets or human beta cells to proinflammatory cytokines in the presence or absence of methylprednisolone for 3 d and characterized its effects on beta-cell death, function, gene and protein expression, and secretion of inflammatory molecules. Results Methylprednisolone prevented cytokine-induced beta-cell failure and death (57% decrease in caspase 3/7 activation [P < 0.05]) after 72 h. This protective effect was associated with an 80% attenuation of the inflammatory cytokine gene IL-8 (80%, P < 0.01), the proapoptotic nuclear factor kappa B-related gene NFKB2 (26%, P < 0.05), and endoplasmic reticulum stress gene ATF3 (48%, P < 0.05) during cytokine treatment. Conclusions We propose that short-term treatment with methylprednisolone is beneficial for beta-cell health under inflammatory conditions which can be relevant in periprocedural pancreas or islet transplantation, and treatment of graft rejection.
Collapse
Affiliation(s)
- Miriam Paz-Barba
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Iris J.A. Peters
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Natascha de Graaf
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Eelco J.P. de Koning
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
4
|
Colpitts SJ, Jegatheeswaran S, Oakie A, Mashhouri S, Sachewsky N, Murshed H, Mathews JA, Reid KT, Misra PS, Fung VCW, Reichman TW, Nostro MC, Verchere CB, Levings MK, Crome SQ. Cell therapy with human IL-10-producing ILC2s enhances islet function and inhibits allograft rejection. Am J Transplant 2025:S1600-6135(25)00280-1. [PMID: 40412656 DOI: 10.1016/j.ajt.2025.05.023] [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: 11/20/2024] [Revised: 04/30/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Group 2 innate lymphoid cells (ILC2s) that produce IL-10 (IL-10+ILC2s) have demonstrated regulatory and tissue-protective properties in murine studies, but preclinical studies are lacking that explore the potential of human IL-10+ILC2s as a tolerance-promoting cell therapy for transplantation or autoimmunity. Here, we investigated whether human IL-10+ILC2s could enhance islet function and prevent allograft rejection in humanized mouse models of islet transplantation. In vitro, human IL-10+ILC2s did not display cytotoxicity towards allogeneic deceased-donor islets or stem cell-derived islet-like cells, and co-transplantation with IL-10+ILC2s significantly improved glucose control post-transplantation. Allogeneic IL10+ILC2s directly inhibited T cell-mediated cytotoxicity against islet-like cells in vitro, and in an antigen-specific transplant rejection model, prevented T cell-mediated rejection of deceased donor islet grafts. Effects were greater with allogeneic IL-10+ILC2s, as autologous cells did not inhibit T cell IFN-γ production or cytotoxic activity in vitro, and were not sufficient to prevent islet rejection in vivo.Collectively, these studies provide proof-of-principle that human IL-10+ILC2s have therapeutic potential for islet transplantation and type 1 diabetes, and support their use as an allogeneic regulatory cell therapy.
Collapse
Affiliation(s)
- Sarah J Colpitts
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Sinthuja Jegatheeswaran
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Siavash Mashhouri
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Nadia Sachewsky
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Humaira Murshed
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Jessica A Mathews
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Kyle T Reid
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Paraish S Misra
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Vivian C W Fung
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Trevor W Reichman
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - C Bruce Verchere
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Sarah Q Crome
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
5
|
Liu M, Deng H, Liu C, Wang L, Liao Z, Li D, Chen Y, Li J, Dong J, Sun X, Wang C, Huang L, Dong L, Xiao J. Islet transplantation in immunomodulatory nanoparticle-remodeled spleens. Sci Transl Med 2025; 17:eadj9615. [PMID: 40397715 DOI: 10.1126/scitranslmed.adj9615] [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: 07/26/2023] [Revised: 10/08/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025]
Abstract
Islet transplantation is a promising therapy for insulin-dependent diabetes. However, immune rejection and insufficient vascularization hinder the survival and function of transplanted islets. Here, we show effective engraftment of vascularized and functional mouse and rat islets transplanted into biomaterial-remodeled spleens of nonimmunosuppressed rodents and human islets transplanted into the remodeled spleens of nonhuman primates (NHPs) on varying degrees of immunosuppression. We found evidence that konjac glucomannan-modified silica nanoparticles (KSiNPs) remodeled the spleen into an extracellular matrix (ECM)-rich, immunosuppressive niche to support the survival of syngeneic or xenogeneic islets. Transplanted islets in the remodeled spleens showed improved engraftment, neovascularization, and functionality and restored normoglycemia in streptozotocin (STZ)-induced type 1 diabetic models in the mice and macaques, with stable insulin and C-peptide secretion in mice for 90 days and macaques for 28 days. KSiNP injection and islet transplantation into macaque spleens under B-ultrasound guidance were preclinically feasible. These findings highlight the safety and effectiveness of spleen tissue remodeling in supporting the survival and function of transplanted islets, providing a promising strategy for treating type 1 diabetes mellitus (T1DM).
Collapse
Affiliation(s)
- Mi Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Department of Wound Healing of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China
- Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, 315300, China
| | - Huiming Deng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Department of Wound Healing of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chunyan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Lintao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Zhongkai Liao
- Department of Organ Transplantation, Second Affiliated Hospital of Hainan Medical University, Haikou, 570216, China
| | - Desheng Li
- Department of Organ Transplantation, Second Affiliated Hospital of Hainan Medical University, Haikou, 570216, China
| | - Yan Chen
- Department of Oncology of the First Affiliated Hospital and Cancer Institute, Hainan Medical University, Haikou, 570102, China
| | - Jianhui Li
- Division of Hepatobiliary Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianhui Dong
- Institute of Transplantation Medicine, Second Affiliated Hospital of Guangxi Medical University; Guangxi Clinical Research Center for Organ Transplantation; Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning 530007, China
| | - Xuyong Sun
- Institute of Transplantation Medicine, Second Affiliated Hospital of Guangxi Medical University; Guangxi Clinical Research Center for Organ Transplantation; Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning 530007, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China
| | - Ling Huang
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, 571199, China
- Center for Pharmacovigilance of Hainan Province, Hainan Medical Products Administration, Haikou, 570216, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jian Xiao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Department of Wound Healing of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
6
|
Traini L, Negueruela J, Elvira B, St-Pierre-Wijckmans W, Vandenbempt V, Buss CE, Li A, Pérez-Chávez I, Ribeiro-Costa F, Nunes M, Messens J, Ezeriņa D, Hay DC, Bansal M, Gurzov EN. Genome editing of TXNIP in human pluripotent stem cells for the generation of hepatocyte-like cells and insulin-producing islet-like aggregates. Stem Cell Res Ther 2025; 16:225. [PMID: 40320524 PMCID: PMC12051322 DOI: 10.1186/s13287-025-04314-5] [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: 12/30/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Thioredoxin-interacting protein (TXNIP) plays a role in regulating endoplasmic reticulum (ER) and oxidative stress, which disrupt glucose homeostasis in diabetes. However, the impact of TXNIP deficiency on the differentiation and functionality of human stem cell-derived somatic metabolic cells remains unclear. METHODS We used CRISPR-Cas12a genome editing to generate TXNIP-deficient (TXNIP-/-) H1 human embryonic stem cells (H1-hESCs). These cells were differentiated into hepatocyte-like cells (HLCs) and stem-cell-derived insulin-producing islets (SC-islets). The maturation and functionality TXNIP-/- and TXNIP+/+ SC-islets were assessed by implantation under the kidney capsule of male or female NOD-SCID mice. RESULTS TXNIP deficiency significantly increased H1-hESC proliferation without affecting pluripotency, viability, or differentiation potential into HLCs and SC-islets. Bulk RNA-sequencing of thapsigargin-treated TXNIP-/- and TXNIP+/+ hESCs revealed differential expression of stress-responsive genes, with enriched apoptosis-related pathways in TXNIP+/+ cells, but minimal transcriptional changes specific to TXNIP deficiency. In HLCs, TXNIP deletion reduced albumin secretion and insulin signalling, as indicated by decreased AKT phosphorylation, while showing no differences in glycolytic activity or lipid metabolism markers. Under thapsigargin-induced ER stress, TXNIP-/- HLCs exhibited transiently reduced eIF2α phosphorylation and lower BiP expression, suggesting compromised adaptive responses to prolonged stress. SC-islets derived from TXNIP-/- hESCs showed comparable viability, endocrine cell composition, and cytokine responses to TXNIP+/+ islets. Following IFNα or IFNγ treatment, STAT1 phosphorylation was increased in TXNIP-/- SC-islets, indicating that IFN signalling remained intact despite TXNIP deficiency. Upon implantation into NOD-SCID mice, both TXNIP-/- and TXNIP+/+ SC-islets produced human C-peptide and responded to glucose stimulation. However, TXNIP-/- SC-islets did not demonstrate enhanced glycaemic control or glucose-stimulated insulin secretion compared to controls. CONCLUSIONS Our study demonstrates that TXNIP deficiency does not improve the differentiation or functionality of HLCs and SC-islets. We present the generation and characterisation of TXNIP-/- and TXNIP+/+ H1-hESCs, HLCs, and SC-islets as valuable models for future studies on the role of TXNIP in metabolic cell biology.
Collapse
Affiliation(s)
- Leonardo Traini
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Javier Negueruela
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Bernat Elvira
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Wadsen St-Pierre-Wijckmans
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Valerie Vandenbempt
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Carlos E Buss
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Ao Li
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Israel Pérez-Chávez
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Francisco Ribeiro-Costa
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Mariana Nunes
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Daria Ezeriņa
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - David C Hay
- Institute for Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Mayank Bansal
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium.
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium.
| |
Collapse
|
7
|
Lee K, Aviles Vargas A, Bottino R, Wang Y. Islet Transplantation: Microencapsulation, Nanoencapsulation, and Hypoimmune Engineering. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70016. [PMID: 40394888 PMCID: PMC12093044 DOI: 10.1002/wnan.70016] [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] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/14/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025]
Abstract
Islet transplantation represents a promising curative approach for type 1 diabetes by restoring glucose-responsive insulin secretion. However, the requirement for lifelong immunosuppression to prevent immune rejection can lead to significant side effects. Emerging strategies such as microencapsulation, nanoencapsulation, and hypoimmune engineering are being developed to protect transplanted islets from immune attack, thereby enhancing their viability and function. This review critically examines these innovative technologies, highlighting the methodologies, materials, experimental and clinical outcomes, as well as the challenges they face and potential solutions to overcome those challenges.
Collapse
Affiliation(s)
- Kyungsene Lee
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | - Ana Aviles Vargas
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | | | - Yong Wang
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| |
Collapse
|
8
|
Agarwal D, Naji A. A Cure Within for Type 1 Diabetes: Stem Cell-derived Islet Transplantation. Transplantation 2025; 109:757-758. [PMID: 39828916 DOI: 10.1097/tp.0000000000005337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Affiliation(s)
- Divyansh Agarwal
- Department of Surgery, Massachusetts General Hospital, Boston, MA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
9
|
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
|
10
|
Bal T. Scaffold-free endocrine tissue engineering: role of islet organization and implications in type 1 diabetes. BMC Endocr Disord 2025; 25:107. [PMID: 40259265 PMCID: PMC12010671 DOI: 10.1186/s12902-025-01919-y] [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: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic hyperglycemia disorder emerging from beta-cell (insulin secreting cells of the pancreas) targeted autoimmunity. As the blood glucose levels significantly increase and the insulin secretion is gradually lost, the entire body suffers from the complications. Although various advances in the insulin analogs, blood glucose monitoring and insulin application practices have been achieved in the last few decades, a cure for the disease is not obtained. Alternatively, pancreas/islet transplantation is an attractive therapeutic approach based on the patient prognosis, yet this treatment is also limited mainly by donor shortage, life-long use of immunosuppressive drugs and risk of disease transmission. In research and clinics, such drawbacks are addressed by the endocrine tissue engineering of the pancreas. One arm of this engineering is scaffold-free models which often utilize highly developed cell-cell junctions, soluble factors and 3D arrangement of islets with the cellular heterogeneity to prepare the transplant formulations. In this review, taking T1D as a model autoimmune disease, techniques to produce so-called pseudoislets and their applications are studied in detail with the aim of understanding the role of mimicry and pointing out the promising efforts which can be translated from benchside to bedside to achieve exogenous insulin-free patient treatment. Likewise, these developments in the pseudoislet formation are tools for the research to elucidate underlying mechanisms in pancreas (patho)biology, as platforms to screen drugs and to introduce immunoisolation barrier-based hybrid strategies.
Collapse
Affiliation(s)
- Tugba Bal
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, 34662, Turkey.
| |
Collapse
|
11
|
Deshpande R, Augustine T. Smart transplants: emerging role of nanotechnology and big data in kidney and islet transplantation, a frontier in precision medicine. Front Immunol 2025; 16:1567685. [PMID: 40264762 PMCID: PMC12011751 DOI: 10.3389/fimmu.2025.1567685] [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: 01/27/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Kidney and islet transplantation has revolutionized the management of renal failure and diabetes. Transplantation is considered as excellent therapeutic intervention for most suitable patients. While advancements in the surgical aspects, immunosuppression and outcomes have potentially plateaued, new technologies have developed which could enhance transplantation with benefits to patients and clinical teams alike. The science of nanotechnology and big data advancements are two such technologies, collectively paving the way for smarter transplantation solutions. Nanotechnology offers novel strategies to overcome critical challenges, including organ preservation, ischemia-reperfusion injury and immune modulation. Innovations such as nanoparticle-based drug delivery systems, biocompatible encapsulation technologies for islet transplants, and implantable artificial kidneys are redefining the standards of care. Meanwhile, big data analytics harness vast datasets to optimize donor-recipient matching, refine predictive models for post-transplant outcomes, and personalize therapeutic regimens. Integrating these technologies forms a synergistic framework where nanotechnology enhances therapeutic precision and big data provides actionable insights, enabling clinicians to adopt proactive, patient-specific strategies. By addressing unmet needs and leveraging the combined potential of nanotechnology and big data, this transformative approach promises to improve graft survival, functionality, and overall patient outcomes, marking a paradigm shift in transplantation medicine. These developments will also be accelerated with integration of the rapidly advancing science of artificial intelligence.
Collapse
Affiliation(s)
- Rajkiran Deshpande
- Department of Renal and Pancreas Transplantation and General Surgery, Manchester Royal Infirmary, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Titus Augustine
- Department of Renal and Pancreas Transplantation and General Surgery, Manchester Royal Infirmary, Manchester University Foundation Trust, Manchester, United Kingdom
- Department Faculty of Biology, Medicine and Health, Division of Diabetes, Endocrinology and Gastroenterology, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
12
|
Yuan Y, Wu Q, Huai G, Ma S, Zhao G. Protocol for renal subcapsular islet transplantation in diabetic mice to induce long-term immune tolerance. STAR Protoc 2025; 6:103729. [PMID: 40186861 PMCID: PMC12002982 DOI: 10.1016/j.xpro.2025.103729] [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: 11/20/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
Murine renal subcapsular islet transplantation presents a promising technique for diabetes treatment by addressing challenges such as immune rejection and reliance on immunosuppressive drugs. Here, we present a protocol for the isolation, purification, and transplantation of mouse pancreatic islets that overcomes these challenges. Specifically, we describe steps for inducing diabetes with streptozotocin, pancreatic perfusion and isolation, and islet cell purification. We then detail procedures for renal subcapsular islet transplantation, dual antibody therapy, and immune cell and graft monitoring. For complete details on the use and execution of this protocol, please refer to Liu et al.1.
Collapse
Affiliation(s)
- Yinglin Yuan
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qibin Wu
- Organ Transplant Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guoli Huai
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengyun Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
13
|
Tol MC, de Vries RHW, Engelse MA, Carlotti F, van Apeldoorn AA, de Koning EJP, Huurman VAL. Subcutaneous Implantation of Open Microwell Islet Delivery Devices in Pigs. Surg Innov 2025; 32:141-148. [PMID: 39670992 PMCID: PMC11894865 DOI: 10.1177/15533506241306491] [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] [Indexed: 12/14/2024]
Abstract
BackgroundIntraportal pancreatic islet transplantation is a treatment option for patients with severe beta cell failure and unstable glycemic control. However, this procedure is associated with loss of beta cells after intrahepatic transplantation. Islet delivery devices (IDDs) implanted at extrahepatic sites may support engraftment and improve survival of pancreatic islets. We assessed the surgical feasibility, tolerability and safety of implantation of open microwell devices at subcutaneous sites with varying friction in pigs.MethodsOpen, non-immunoisolating microwell islet delivery devices were made from polyvinylidene fluoride (PVDF). Empty (n = 26) and islet-seeded devices (n = 8) were implanted subcutaneously in 6 immunocompetent pigs in low-friction sites (abdomen and lateral hip) and high-friction sites (anterior neck) for 3 months. Retrieved grafts were analyzed histologically with haematoxylin and eosin, and Masson's Trichrome staining.ResultsIslet-seeding and transportation of IDDs was free from complications with minimal islet spillage. IDDs were implanted subcutaneously using standard surgical equipment, without complications during the surgeries. IDDs implanted in the neck and IDDs co-transplanted with human islets were expelled and retrieved after 10 days. Empty IDDs were removed after 3 months. The abdominal site showed reduced signs of inflammation as compared to the neck region, while similar tissue ingrowth and vascularization of devices were found in the two locations.ConclusionsOpen microwell IDDs can safely be implanted with standard surgical equipment and successful islet-loading can be performed. Low-friction sites are preferable over high-friction sites for subcutaneous implantation in the porcine model since these lead to the least amount of foreign body reaction.
Collapse
Affiliation(s)
- Maarten C. Tol
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Rick H. W. de Vries
- Department of Cell Biology – Inspired Tissue Engineering (cBITE), MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Marten A. Engelse
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Aart A. van Apeldoorn
- Department of Cell Biology – Inspired Tissue Engineering (cBITE), MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Eelco J. P. de Koning
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Volkert A. L. Huurman
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Lai F, Zhou K, Ma Y, Lv H, Wang W, Wang R, Xu T, Huang R. Single-cell RNA sequencing identifies endothelial-derived HBEGF as promoting pancreatic beta cell proliferation in mice via the EGFR-Kmt5a-H4K20me pathway. Diabetologia 2025; 68:835-853. [PMID: 39694915 PMCID: PMC11950091 DOI: 10.1007/s00125-024-06341-y] [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: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 12/20/2024]
Abstract
AIMS/HYPOTHESIS Pancreatic beta cell mass is dynamically regulated in response to increased physiological and pathological demands. Understanding the mechanisms that control physiological beta cell proliferation could provide valuable insights into novel therapeutic approaches to diabetes. Here, we aimed to analyse the intracellular and extracellular signalling pathways involved in regulating the physiological proliferation of beta cells using single-cell RNA-seq (scRNA-seq) and in vitro functional assays. METHODS Islets isolated from nulliparous mice, mice at different time points of gestation and mice at day 4 after delivery were analysed using scRNA-seq. Bioinformatics analyses of scRNA-seq data were performed to determine the heterogeneous transcriptomic characteristics of beta cells and to identify the proliferating subpopulation. CellChat was used to analyse cell-cell communication and identify the ligand-receptor pairs between beta cell subclusters as well as between non-beta cells and proliferating beta cells. In vitro functional assays were conducted in mouse and rat beta cell lines and isolated mouse primary islets to validate the role of Kmt5a- mono-methylation of histone H4 at lysine 20 (H4K20me) signalling and endothelial-derived heparin-binding EGF-like growth factor (HBEGF) in beta cell proliferation. RESULTS Of 43,724 endocrine and non-endocrine cells within islets analysed by scRNA-seq, 15,569 beta cells were clustered into eight distinct populations, each exhibiting unique heterogeneity. A proliferating beta cell subcluster was identified that highly expressed the histone methyltransferase Kmt5a. Activation of Kmt5a-H4K20me signalling upregulated the expression of Cdk1 and promoted beta cell proliferation. The crosstalk between endothelial cells and the proliferating beta cell subcluster, mediated by the HBEGF-EGF receptor (EGFR) ligand-receptor interaction, increased as beta cell mass expanded. HBEGF increased the expression levels of genes involved in the cell cycle and promoted beta cell proliferation by regulating the Kmt5a-H4K20me signalling pathway. CONCLUSIONS/INTERPRETATION Our study demonstrates that, under physiological conditions, endothelial-derived HBEGF regulates beta cell proliferation through the Kmt5a-H4K20me signalling pathway, which may serve as a potential target to promote beta cell expansion and treat diabetes. DATA AVAILABILITY The scRNA-seq and RNA-seq datasets are available from the Gene Expression Omnibus (GEO) using the accession numbers GSE278860 and GSE278861, respectively.
Collapse
Affiliation(s)
- Fengling Lai
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kaixin Zhou
- Guangzhou National Laboratory, Guangzhou, China
| | - Yingjie Ma
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Lv
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Weilin Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Rundong Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tao Xu
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Rong Huang
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
15
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [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] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
16
|
Peng X, Zhao L, Wang J, Zhang Y, Liu Z, Wang K, Zhang L. Melatonin Alleviates Oxidative Stress-Induced Mitochondrial Dysfunction Through Ameliorating NAD + Homeostasis of hDPSCs for Cell-Based Therapy. J Pineal Res 2025; 77:e70058. [PMID: 40391773 DOI: 10.1111/jpi.70058] [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: 03/26/2025] [Revised: 03/29/2025] [Accepted: 04/30/2025] [Indexed: 05/22/2025]
Abstract
Human dental pulp stem cells (hDPSCs) exhibit amazing therapeutic abilities in a variety of diseases due to their remarkable self-renewal capacity and multi-differentiation potential. However, their therapeutic potential could be weakened by various factors such as oxidative stress in cell survival microenvironment In Vivo. Here, we explored the protective effect and mechanism of melatonin (Mel) on hDPSCs transplanted in a type 1 diabetes mellitus (T1DM) rat model. Nicotinamide adenine dinucleotide (NAD+) metabolism and mitochondrial function were remarkably impaired in T1DM rats caused by oxidative stress, while the combination of Mel and post-hDPSCs transplantation could rebalance NAD+ homeostasis through regulating NAMPT-NAD+-SIRT1 axis. Furthermore, Mel significantly reduced intracellular and mitochondrial reactive oxygen species, and alleviated cell senescence and apoptosis of hDPSCs exposed to hydrogen peroxide through ameliorating NAD+ depletion and mitochondrial dysfunction. The protective role of Mel could be extremely essential to stem cells in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Xiu Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiale Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yinmo Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zihan Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kun Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
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
|
18
|
Zushi N, Takuma M, Endo A, Suzuki M, Wu Y, Shiraki N, Kume S, Fujie T. Multilayered Freestanding Porous Polycarbonate Nanosheets with Directed Protein Permeability for Cell-Encapsulated Devices. ACS APPLIED BIO MATERIALS 2025; 8:1963-1971. [PMID: 39951110 PMCID: PMC11921017 DOI: 10.1021/acsabm.4c01446] [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: 10/04/2024] [Revised: 01/25/2025] [Accepted: 02/02/2025] [Indexed: 03/18/2025]
Abstract
Implantable pancreatic β cell-encapsulated devices are required for the treatment of type 1 diabetes. Such devices should enable a semipermeable membrane to release insulin in response to glucose levels while avoiding immune reactions. Micrometer-thick track-etched porous polycarbonate (PC) membranes have been used for this purpose. However, the immediate release of insulin remains a challenge in the development of such semipermeable membranes. Herein, we attempted to develop a freestanding polymeric ultrathin film (nanosheet) with a porous structure that can be used in a cell-encapsulated device. Specifically, we fabricated a nonbiodegradable, porous PC nanosheet to enhance molecular permeability. The nanosheet was multistacked to ensure the controlled permeability of proteins of various molecular weights, such as insulin and IgG. The porous PC nanosheet was prepared by gravure coating using a blend solution comprising PC and polystyrene (PS) to induce macro-phase separation of the PC and PS. When the PC:PS weight ratio of the mixture was reduced to 3:1, we succeeded in fabricating a porous PC nanosheet (thickness: 100 nm, diameter: < 2.5 μm). A triple layer of such porous nanosheets with various pore sizes demonstrated 10 times less protein clogging, 10 times higher insulin permeability, and comparable IgG-blocking capability compared with commercially available porous PC membranes (thickness: 10 μm). Finally, we demonstrated that a cell-encapsulated device equipped with the multilayered porous PC nanosheet as a permeable membrane preserved the glucose response level of insulin-producing cells before, during, and after the cell-encapsulation process. We believe that cell-encapsulated devices equipped with such porous PC nanosheets will enable immediate insulin release in response to changes in glucose levels.
Collapse
Affiliation(s)
- Nanami Zushi
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Megumi Takuma
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Atena Endo
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Mahiro Suzuki
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Yumeng Wu
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Nobuaki Shiraki
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Shoen Kume
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Toshinori Fujie
- School
of Life Science and Technology, Institute of Science Tokyo, B-50, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
- Research
Center for Autonomous Systems Materialogy (ASMat), Institute of Integrated
Research (IIR), Institute of Science Tokyo, R3-23, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
| |
Collapse
|
19
|
de Bont DFA, Mohammed SG, de Vries RHW, Paulino da Silva Filho O, Vaithilingam V, Jetten MJ, Engelse MA, de Koning EJP, van Apeldoorn AA. Supporting islet function in a PVDF membrane based macroencapsulation delivery device by solvent non-solvent casting using PVP. PLoS One 2025; 20:e0298114. [PMID: 40073008 PMCID: PMC11902058 DOI: 10.1371/journal.pone.0298114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/08/2024] [Indexed: 03/14/2025] Open
Abstract
Type 1 diabetic (T1D) patients are life-long dependent on insulin therapy to keep their blood glucose levels under control. An alternative cell-based therapy for exogenous insulin injections is clinical islet transplantation (CIT). Currently the widespread application of CIT is limited, due to risks associated with the life-long use of immunosuppressive drugs to prevent rejection of donor cells. An immunoprotective macroencapsulation device can protect allogeneic islet cells against the host immune system and allow exploring extrahepatic transplantation sites. We report on the characterization and creation of porous polyvinylidene fluoride (PVDF) membrane-based devices intended for islet and beta-cell transplantation. We hypothesize that by incorporating polyvinyl-pyrrolidone (PVP) into a PVDF solution the permeability of PVDF membranes for insulin and glucose can be improved by solvent-non solvent casting to create submicrometer porous films. We show that the use of water-soluble PVP, can significantly increase glucose diffusion through these membranes while still having the ability to block immune cells from migrating through these membranes. Human donor islets loaded into devices made from these thin PVDF/PVP membranes showed a 92 ± 4% viability after 8 days similar to their free-floating counterparts. The glucose responsiveness of human donor islets encapsulated inside PVDF/PVP membrane-based devices was significantly improved compared to islets seeded in devices made from PVDF membranes without PVP, with a stimulation index of 3.2 for PVDF/PVP devices and 1.3 for PVDF-alone devices at day 8. Our data show that by addition of PVP as pore forming agent during membrane fabrication at a specific ratio the diffusion characteristics can be tuned such that human islet function in these closed macrodevices, can be kept at the same level as non-encapsulated islets, while the membrane can still serve as a protective barrier preventing the entry of primary human macrophages and damaging beta cells.
Collapse
Affiliation(s)
- Denise F. A. de Bont
- Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Sami G. Mohammed
- Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Rick H. W. de Vries
- Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Omar Paulino da Silva Filho
- Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Vijayaganapathy Vaithilingam
- Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Marlon J. Jetten
- Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Marten A. Engelse
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eelco J. P. de Koning
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Hubrecht Institute, Utrecht, The Netherlands
| | - Aart A. van Apeldoorn
- Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
20
|
Ma K, Han H, Bao Y, Chen R, Yang Y, Shao W. The Function of B and T Lymphocyte Attenuator and Its Role in Transplantation. APMIS 2025; 133:e70012. [PMID: 40040475 DOI: 10.1111/apm.70012] [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: 12/02/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025]
Abstract
Immune checkpoints are important molecules that regulate the immune response, preventing its overactivation from causing tissue damage and autoimmune diseases. B and T lymphocyte attenuator (BTLA) plays an important role in regulating the activation and suppression of the immune response as part of a bidirectional signaling complex. The BTLA and its ligand herpesvirus entry mediator (HVEM) interaction transmits inhibitory signals that suppress the biological activity of T cells, B cells, and DCs. In addition, BTLA-HVEM can affect the induction of Treg cells, further suggesting its important role in immune regulation. Organ transplantation is the ultimate treatment option for many patients with end-stage organ failure. Transplant rejection can cause damage to the transplanted organ, which seriously affects the prognosis of patients. Therefore, we would like to explore the potential application value of the BTLA-HVEM interaction to exert an immunosuppressive function and thus attenuate transplant rejection. We first reviewed the structure and function of BTLA and HVEM, then summarized their research progress in organ transplantation, and further explored the directions of potential future applications and the challenges of current BTLA-HVEM applications.
Collapse
MESH Headings
- Humans
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/genetics
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/immunology
- Receptors, Tumor Necrosis Factor, Member 14/chemistry
- Organ Transplantation
- Graft Rejection/immunology
- Graft Rejection/prevention & control
- Animals
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Kai Ma
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Heqiao Han
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yuchen Bao
- Medical School of Tianjin University, Tianjin, China
| | - Rongtao Chen
- Medical School of Tianjin University, Tianjin, China
| | - Yixuan Yang
- Medical School of Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
- Medical School of Tianjin University, Tianjin, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China
| |
Collapse
|
21
|
Wan J, Xu Y, Qi T, Xue X, Li Y, Huang M, Guo Y, Guo Q, Lu Y, Huang Y. AG73-GelMA/AlgMA hydrogels provide a stable microenvironment for the generation of pancreatic progenitor organoids. J Nanobiotechnology 2025; 23:149. [PMID: 40016740 PMCID: PMC11866579 DOI: 10.1186/s12951-025-03266-5] [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: 11/06/2024] [Accepted: 02/21/2025] [Indexed: 03/01/2025] Open
Abstract
Patient specific induced pluripotent stem cells (iPSCs) derived β cells represent an effective means for disease modeling and autologous diabetes cell replacement therapy. In this study, an AG73-5%gelatin methacryloyl (GelMA) /2% alginate methacrylate (AlgMA) hydrogel was employed to generate pancreatic progenitor (PP) organoids and improve stem cell-derived β (SC-β) cell differentiation protocol. The laminin-derived homolog AG73, which mimics certain cell‒matrix interactions, facilitates AKT signaling pathway activation to promote PDX1+/NKX6.1+ PP organoid formation and effectively modulates subsequent epithelial-mesenchymal transition (EMT) in the endocrine lineage. The 5%GelMA/2%AlgMA hydrogel mimics the physiological stiffness of the pancreas, providing the optimal mechanical stress and spatial structure for PP organoid differentiation. The Syndecan-4 (SDC4)-ITGAV complex plays a pivotal role in the early stages of pancreatic development by facilitating the formation of SOX9+/PDX1+ bipotent PPs. Our findings demonstrate that AG73-GelMA/AlgMA hydrogel-derived SC-β cells exhibit enhanced insulin secretion and accelerated hyperglycemia reversal in vivo. This study presents a cost-effective, stable, and efficient alternative for the comprehensive 3D culture of SC-β cells in vitro by mitigating the uncertainties associated with conventional culture methods.
Collapse
Affiliation(s)
- Jian Wan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yang Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Tianmu Qi
- Medical School of Nantong University, Nantong, China
| | - Xiaoxia Xue
- Department of Nephrology, Rugao Hospital of Traditional Chinese Medicine, Nantong, China
| | - Yuxi Li
- Medical School of Nantong University, Nantong, China
| | - Minjie Huang
- Medical School of Nantong University, Nantong, China
| | - Yuchen Guo
- Medical School of Nantong University, Nantong, China
| | - Qingsong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co- Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| |
Collapse
|
22
|
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
|
23
|
Langlois A, Cherfan J, Meugnier E, Rida A, Arous C, Peronet C, Hamdard H, Zarrouki B, Wehrle‐Haller B, Pinget M, Craige SM, Bouzakri K. DECORIN, a triceps-derived myokine, protects sorted β-cells and human islets against chronic inflammation associated with type 2 diabetes. Acta Physiol (Oxf) 2025; 241:e14267. [PMID: 39844653 PMCID: PMC11754997 DOI: 10.1111/apha.14267] [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: 09/05/2024] [Revised: 11/28/2024] [Accepted: 01/01/2025] [Indexed: 01/24/2025]
Abstract
AIM Pancreatic β-cells are susceptible to inflammation, leading to decreased insulin production/secretion and cell death. Previously, we have identified a novel triceps-derived myokine, DECORIN, which plays a pivotal role in skeletal muscle-to-pancreas interorgan communication. However, whether DECORIN can directly impact β-cell function and susceptibility to inflammation remains unexplored. METHODS The effect of DECORIN was assessed in sorted human and rat β-cell and human islets from healthy and type 2 diabetes (T2D) donors. We assessed glucose-stimulated insulin secretion (GSIS) and cytokine-mediated cell death. We then challenged sorted β-cells and human islets with inflammatory cytokines commonly associated with diabetes, such as tumor necrosis factor-α (TNF-α) alone or in combination with interleukin1-β (IL1-β) and interferon-γ (cytomix). RESULTS DECORIN enhanced cell spreading and the localization of phosphorylated FAK at adhesions, promoting GSIS under basal conditions. It also increased insulin granule docking adhesion length and countered the inhibitory effects of TNF-α on adhesion and actin remodeling at the β-cell surface, resulting in preserved GSIS. DECORIN protected from cell death in sorted β-cells and islets challenged with TNF-α alone or TNF-α + cytomix. Interestingly, DECORIN increased both insulin content and secretion in human islets from T2D individuals. Additionally, DECORIN treatment reversed the impaired gene expression caused by T2D and enhanced the expression of genes essential for islet function and metabolism. CONCLUSION Collectively, we have shown that DECORIN had a beneficial effect on human islets, protecting them from inflammation-induced cell death. In T2D islets, DECORIN restores islet function and reverses the expression of T2D-associated genes. Based on our data, we propose that DECORIN is a promising therapeutic target for diabetes-associated inflammation and diabetes itself.
Collapse
Affiliation(s)
- Allan Langlois
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Julien Cherfan
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Emmanuelle Meugnier
- CarMeN Laboratory, Inserm U1060, INRAE UMR1397, Univ‐LyonUniversité Claude Bernard Lyon‐1LyonFrance
| | - Ahmad Rida
- ILONOV, Boulevard René LericheStrasbourgFrance
| | - Caroline Arous
- Department of Cell Physiology and Metabolism, Centre Médical UniversitaireUniversity of GenevaGenevaSwitzerland
| | - Claude Peronet
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Harzo Hamdard
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Bader Zarrouki
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolic (CVRM), BioPharmaceuticals R&DGothenburgSweden
| | - Bernhard Wehrle‐Haller
- Department of Cell Physiology and Metabolism, Centre Médical UniversitaireUniversity of GenevaGenevaSwitzerland
| | - Michel Pinget
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
- ILONOV, Boulevard René LericheStrasbourgFrance
| | - Siobhan M. Craige
- Department of Human Nutrition, Foods, and ExerciseVirginia TechBlacksburgVirginiaUSA
| | - Karim Bouzakri
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
- ILONOV, Boulevard René LericheStrasbourgFrance
| |
Collapse
|
24
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
25
|
Mar S, Filatov E, Sasaki S, Mojibian M, Zhang D, Yang A, Nian C, Lynn FC. Tracking Insulin- and Glucagon-Expressing Cells In Vitro and In Vivo Using a Double-Reporter Human Embryonic Stem Cell Line. Diabetes 2025; 74:188-198. [PMID: 39561351 PMCID: PMC11755683 DOI: 10.2337/db24-0756] [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: 08/28/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
ARTICLE HIGHLIGHTS Differentiation protocols used to generate stem cell-derived islet cells yield heterogenous cell populations. We generated a human embryonic stem cell line that reports insulin- and glucagon-expressing cells in vitro and in vivo without altering their differentiation or function. We showed some insulin- and glucagon-expressing bihormonal cells are cell-autonomously fated to become α-like cells. This reporter cell line can be used to further study and improve stem cell-derived islet differentiation and transplantation.
Collapse
Affiliation(s)
- Samantha Mar
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ekaterina Filatov
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shugo Sasaki
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Majid Mojibian
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dahai Zhang
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Angela Yang
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cuilan Nian
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Francis C. Lynn
- BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Primavera R, Wang J, Buchwald P, Ganguly A, Patel S, Bettencourt L, Chetty S, Yarani R, Regmi S, Levitte S, Kevadiya B, Guindani M, Decuzzi P, Thakor AS. Controlled Nutrient Delivery to Pancreatic Islets Using Polydopamine-Coated Mesoporous Silica Nanoparticles. NANO LETTERS 2025; 25:939-950. [PMID: 39791700 DOI: 10.1021/acs.nanolett.4c03613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
In this study, we designed a nanoscale platform for sustained amino acid delivery to support transplanted pancreatic islets. The platform features mesoporous silica nanoparticles (MSNPs) loaded with glutamine (G), an essential amino acid required for islet survival and function, and coated with polydopamine (PD). We investigated various PD concentrations (0.5-2 mg/mL) and incubation times (0.5-2 h) to optimize G release, identifying that a PD concentration of 0.5 mg/mL incubated for 0.5 h yielded the best results to support islet viability and functionality ex vivo, particularly under inflammatory conditions. In syngeneic islet transplantation in STZ-diabetic mice, G alone provided only temporary benefits; however, PD-G-MSNPs significantly improved islet engraftment and function, with animals maintaining glycemic control for 30 days due to controlled G release. Our findings support the use of this nanoscale platform to provide essential nutrients like G to transplanted islets until they can establish their own blood and nutrient supply.
Collapse
Affiliation(s)
- Rosita Primavera
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Jing Wang
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Abantika Ganguly
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shaini Patel
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Lili Bettencourt
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shashank Chetty
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Shobha Regmi
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Steven Levitte
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Bhavesh Kevadiya
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Michele Guindani
- Department of Biostatistics, Jonathan and Karin Fielding School of Public Health, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Avnesh S Thakor
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| |
Collapse
|
27
|
Kim JE, Lee JW, Cha GD, Yoon JK. The Potential of Mesenchymal Stem Cell-Derived Exosomes to Treat Diabetes Mellitus. Biomimetics (Basel) 2025; 10:49. [PMID: 39851765 PMCID: PMC11760843 DOI: 10.3390/biomimetics10010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Diabetes mellitus (DM) is a fatal metabolic disease characterized by persistent hyperglycemia. In recent studies, mesenchymal stem cell (MSC)-derived exosomes, which are being investigated clinically as a cell-free therapy for various diseases, have gained attention due to their biomimetic properties that closely resemble natural cellular communication systems. These MSC-derived exosomes inherit the regenerative and protective effects from MSCs, inducing pancreatic β-cell proliferation and inhibiting apoptosis, as well as ameliorating insulin resistance by suppressing the release of various inflammatory cytokines. Consequently, MSC-derived exosomes have attracted attention as a novel treatment for DM as an alternative to stem cell therapy. In this review, we will introduce the potential of MSC-derived exosomes for the treatment of DM by discussing the studies that have used MSC-derived exosomes to treat DM, which have shown therapeutic effects in both type 1 and type 2 DM.
Collapse
Affiliation(s)
| | | | | | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea (G.D.C.)
| |
Collapse
|
28
|
Isenberg JS, Kandeel F. Can Islet Transplantation Possibly Reduce Mortality in Type 1 Diabetes. Cell Transplant 2025; 34:9636897241312801. [PMID: 39831598 PMCID: PMC11748148 DOI: 10.1177/09636897241312801] [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: 10/07/2024] [Revised: 11/21/2024] [Accepted: 12/22/2024] [Indexed: 01/22/2025] Open
Abstract
Islet transplantation (IT) is a successful natural cell therapy. But the benefits are known mostly to individuals with severe type 1 diabetes who undergo IT and the health care professionals that work to make the therapy available, reproducible, and safe. Data linking IT to overall survival in T1D might alter this situation and frame the therapy in a more positive light. Recent analysis of mortality in several cohorts suggests that IT has possible survival benefits when used alone or in conjunction with renal transplantation. Multi-center prospective studies with long-term follow-up of individuals that receive stand-alone IT versus individuals who qualify for but do not undergo the procedure would seem reasonable to undertake to confirm an IT survival benefit.
Collapse
Affiliation(s)
- Jeffrey S. Isenberg
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
29
|
Forgioni A, Watanabe M, Goto R, Harada T, Ota T, Shimamura T, Taketomi A. Anti-Inflammatory Effects of Ex Vivo-Generated Donor Antigen-Specific Immunomodulatory Cells on Pancreatic Islet Transplantation. Cell Transplant 2025; 34:9636897251317887. [PMID: 39981681 PMCID: PMC11843686 DOI: 10.1177/09636897251317887] [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: 10/15/2024] [Revised: 01/01/2025] [Accepted: 01/13/2025] [Indexed: 02/22/2025] Open
Abstract
Pancreatic islet transplantation (PITx) is a promising treatment option for patients with type 1 diabetes mellitus. Previously, we demonstrated that therapy with alloantigen-specific immunomodulatory cells (IMCs) generated ex vivo in the presence of anti-CD80 and CD86 monoclonal antibodies (mAbs), successfully induced tolerance following clinical liver transplantation. To extend IMC therapy to PITx, it is crucial to address the strong inflammatory and innate immune responses that occur immediately after PITx. In this study, we investigated the efficacy of IMCs in modulating macrophage activation and mitigating inflammatory damage of pancreatic islets. IMCs were induced using mouse splenocytes in the presence of anti-mouse anti-CD80 (RM80) and anti-CD86 (GL-1) mAbs. IMCs exerted donor-specific immunosuppressive effects in a mixed lymphocyte reaction. During lipopolysaccharide (LPS) stimulation, the addition of IMCs suppressed conversion to the M1 phenotype and promoted a shift toward the M2 phenotype, particularly under direct cell-cell contact conditions. Nitric oxide production, a hallmark of M1 polarized macrophages, was significantly reduced in LPS-stimulated RAW264 macrophages by IMC treatment. These findings were associated with reduced secretion of pro-inflammatory cytokines, tumoral necrosis factor α, and interleukin-6, and increased interleukin-10 production by macrophages. IMCs effectively prevented macrophage-mediated islet destruction after 12 h of co-culture with LPS-stimulated macrophages and significantly inhibited macrophage migration toward allogeneic islets in vitro. Intraportal co-infusion of IMCs with syngeneic islets in a mouse PITx model resulted in reduced messenger RNA (mRNA) expression of pro-inflammatory cytokines in the recipient liver. Immunohistochemical staining revealed a significantly lower number of F4/80+ macrophages at the transplantation site in IMCs-treated mice. These results demonstrate that IMCs modulate macrophage polarization, promoting a shift toward the M2 phenotype and protecting islets from macrophage-mediated damage. These effects combined with its intrinsic donor antigen-specific immunosuppressive capacity make IMC therapy a promising strategy for improving outcomes after PITx.
Collapse
Affiliation(s)
- Agustina Forgioni
- Department of Gastroenterological Surgery I, Hokkaido University, Sapporo, Japan
| | - Masaaki Watanabe
- Department of Transplant Surgery, Hokkaido University, Sapporo, Japan
| | - Ryoichi Goto
- Department of Gastroenterological Surgery I, Hokkaido University, Sapporo, Japan
| | - Takuya Harada
- Department of Gastroenterological Surgery I, Hokkaido University, Sapporo, Japan
| | - Takuji Ota
- Department of Gastroenterological Surgery I, Hokkaido University, Sapporo, Japan
| | - Tsuyoshi Shimamura
- Department of Organ Transplantation Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University, Sapporo, Japan
| |
Collapse
|
30
|
Wang X, Zeng Z, Li D, Wang K, Zhang W, Yu Y, Wang X. Advancements and Challenges in Immune Protection Strategies for Islet Transplantation. J Diabetes 2025; 17:e70048. [PMID: 39829227 PMCID: PMC11744047 DOI: 10.1111/1753-0407.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025] Open
Abstract
Pancreatic islet transplantation is a crucial treatment for managing type 1 diabetes (T1D) in clinical settings. However, the limited availability of human cadaveric islet donors and the need for ongoing administration of immunosuppressive agents post-transplantation hinder the widespread use of this treatment. Stem cell-derived islet organoids have emerged as an effective alternative to primary human islets. Nevertheless, implementing this cell replacement therapy still requires chronic immune suppression, which may result in life-long side effects. To address these challenges, innovations such as encapsulation devices, universal stem cells, and immunomodulatory strategies are being developed to mitigate immune rejection and prolong the function of the transplant. This review outlines the contemporary challenges in pancreatic β cell therapy, particularly immune rejection, and recent progress in immune-isolation devices, hypoimmunogenic stem cells, and immune regulation of transplants. A comprehensive evaluation of the advantages and limitations of these approaches will contribute to improved future clinical investigations. With these promising advancements, the application of pancreatic β cell therapy holds the potential to effectively treat T1D and benefit a larger population of T1D patients.
Collapse
Affiliation(s)
- Xue Wang
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Clinical Stem Cell Research CenterPeking University Third HospitalBeijingChina
| | - Ziyuan Zeng
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Clinical Stem Cell Research CenterPeking University Third HospitalBeijingChina
| | - Dayan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and RemodelingClinical Stem Cell Research Center, Peking University Third Hospital, Peking UniversityBeijingChina
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and RemodelingClinical Stem Cell Research Center, Peking University Third Hospital, Peking UniversityBeijingChina
- Beijing Advanced Center of Cellular Homeostasis and Aging‐Related DiseasesPeking UniversityBeijingChina
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance co. Ltd.BeijingChina
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Clinical Stem Cell Research CenterPeking University Third HospitalBeijingChina
- Beijing Advanced Center of Cellular Homeostasis and Aging‐Related DiseasesPeking UniversityBeijingChina
| | - Xi Wang
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Clinical Stem Cell Research CenterPeking University Third HospitalBeijingChina
- Beijing Advanced Center of Cellular Homeostasis and Aging‐Related DiseasesPeking UniversityBeijingChina
- Institute of Advanced Clinical Medicine, Peking UniversityBeijingChina
| |
Collapse
|
31
|
Moeun BN, Lemaire F, Smink AM, Ebrahimi Orimi H, Leask RL, de Vos P, Hoesli CA. Oxygenation and function of endocrine bioartificial pancreatic tissue constructs under flow for preclinical optimization. J Tissue Eng 2025; 16:20417314241284826. [PMID: 39866963 PMCID: PMC11758540 DOI: 10.1177/20417314241284826] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/02/2024] [Indexed: 01/28/2025] Open
Abstract
Islet transplantation and more recently stem cell-derived islets were shown to successfully re-establish glycemic control in people with type 1 diabetes under immunosuppression. These results were achieved through intraportal infusion which leads to early graft losses and limits the capacity to contain and retrieve implanted cells in case of adverse events. Extra-hepatic sites and encapsulation devices have been developed to address these challenges and potentially create an immunoprotective or immune-privileged environment. Many strategies have achieved reversal of hyperglycemia in diabetic rodents. So far, the results have been less promising when transitioning to humans and larger animal models due to challenges in oxygenation and insulin delivery. We propose a versatile in vitro perfusion system to culture and experimentally study the function of centimeter-scale tissues and devices for insulin-secreting cell delivery. The system accommodates various tissue geometries, experimental readouts, and oxygenation tensions reflective of potential transplantation sites. We highlight the system's applications by using case studies to explore three prominent bioartificial endocrine pancreas (BAP) configurations: (I) with internal flow, (II) with internal flow and microvascularized, and (III) without internal flow. Oxygen concentration profiles modeled computationally were analogous to viability gradients observed experimentally through live/dead endpoint measurements and in case I, time-lapse fluorescence imaging was used to monitor the viability of GFP-expressing cells in real time. Intervascular BAPs were cultured under flow for up to 3 days and BAPs without internal flow for up to 7 days, showing glucose-responsive insulin secretion quantified through at-line non-disruptive sampling. This system can complement other preclinical platforms to de-risk and optimize BAPs and other artificial tissue designs prior to clinical studies.
Collapse
Affiliation(s)
- Brenden N Moeun
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
| | - Florent Lemaire
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Richard L Leask
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| |
Collapse
|
32
|
Aljani B, Lindner A, Weigelt M, Zhao M, Sharma V, Bonifacio E, Jones P, Eugster A. Small RNA-Seq and real time rt-qPCR reveal islet miRNA released under stress conditions. Islets 2024; 16:2392343. [PMID: 39154325 PMCID: PMC11332650 DOI: 10.1080/19382014.2024.2392343] [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: 04/29/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
Replacement of beta cells through transplantation is a potential therapeutic approach for individuals with pancreas removal or poorly controllable type 1 diabetes. However, stress and death of beta cells pose significant challenges. Circulating miRNA has emerged as potential biomarkers reflecting early beta cell stress and death, allowing for timely intervention. The aim of this study was to identify miRNAs as potential biomarkers for beta cell health. Literature review combined with small RNA sequencing was employed to select islet-enriched miRNA. The release of those miRNA was assessed by RT-qPCR in vivo, using a streptozotocin induced diabetes mouse model and in vitro, through mouse and human islets exposed to varying degrees of hypoxic and cytokine stressors. Utilizing the streptozotocin induced model, we identified 18 miRNAs out of 39 candidate islet-enriched miRNA to be released upon islet stress in vivo. In vitro analysis of culture supernatants from cytokine and/or hypoxia stressed islets identified the release of 45 miRNAs from mouse and 8 miRNAs from human islets. Investigation into the biological pathways targeted by the cytokine- and/or hypoxia-induced miRNA suggested the involvement of MAPK and PI3K-Akt signaling pathways in both mouse and human islets. We have identified miRNAs associated with beta cell health and stress. The findings allowed us to propose a panel of 47 islet-related human miRNA that is potentially valuable for application in clinical contexts of beta cell transplantation and presymptomatic early-stage type 1 diabetes.
Collapse
Affiliation(s)
- Bssam Aljani
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Annett Lindner
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Marc Weigelt
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Min Zhao
- German Center for Environmental Health, Institute of Diabetes Research, Helmholtz Munich, Munich, Germany
| | - Virag Sharma
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ezio Bonifacio
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
- Faculty of Medicine, German Center for Diabetes Research (DZD), Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Peter Jones
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London, UK
| | - Anne Eugster
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
33
|
Sun G, Qi M, Sun O, Lizhar EM, Hussey D, Shi Y, Riggs AD. Multi-omics analysis of long-term cultured human islets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.25.626491. [PMID: 39763987 PMCID: PMC11703225 DOI: 10.1101/2024.12.25.626491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
β-cell dysfunction in pancreatic islets, characterized as either the loss of β-cell mass or the resistance of β-cell to glucose, is the leading cause of progression to diabetes. Islet transplantation became a promising approach to replenish functional β-cell mass. However, not much known about changes in islets used for transplantation after isolation. We have subjected human islets into long-term in vitro culture (LTC) and characterized those survived islets. While most of the dysregulated genes were downregulated during LTC, specific groups of mRNA or miRNA were upregulated, and they are involved in specific pathways. In general, α-cells and β-cells of LTC-islets have elevated expressions of MAFB and MAFA genes, respectively. We also found that exocrine cells were eliminated faster than endocrine cells, and β-cells were lost at a higher rate than α-cells. Interestingly, one specific group of cells that have characteristics of immature α-cells or β-cells, were enriched in LTC-islets, revealing the possibility of transdifferentiation of α-cells to β-cells, or dedifferentiation of β-cells to α -cells, under in vitro culture. Our results suggest that there are intrinsic cellular and molecular mechanisms in pancreatic cells that are associated with their maturity and correlated with their survival ability under unfavorable living conditions.
Collapse
Affiliation(s)
- Guihua Sun
- Department of Neurodegenerative Diseases, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Meirigeng Qi
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Olivia Sun
- Department of Diabetes & Cancer Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Elizabeth M. Lizhar
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Deborah Hussey
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Arthur D. Riggs
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
34
|
Krishnan SR, Langer R, Anderson DG. Materials approaches for next-generation encapsulated cell therapies. MRS COMMUNICATIONS 2024; 15:21-33. [PMID: 39958992 PMCID: PMC11825545 DOI: 10.1557/s43579-024-00678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/29/2024] [Indexed: 02/18/2025]
Abstract
Transplanted cells can act as living drug factories capable of secreting therapeutic proteins in vivo, with applications in the treatment of Type 1 diabetes (T1D), blood borne disease, vision disorders, and degenerative neural disease, potentially representing functional cures for chronic conditions. However, attack from the host immune system represents a major challenge, requiring chronic immunosuppression to enable long-lived cell transplantation in vivo. Encapsulating cells in engineered biomaterials capable of excluding components of the host immune system while allowing for the transport of therapeutic proteins, oxygen, nutrients, metabolites, and waste products represents a potential solution. However, the foreign-body response can lead to isolation from native vasculature and hypoxia leading to cell death. In this prospective article, we highlight materials-based solutions to three important challenges in the field: (i) improving biocompatibility and reducing fibrosis; (ii) enhancing transport of secreted protein drugs and key nutrients and oxygen via engineered, semipermeable membranes; and (iii) improving oxygenation. These efforts draw on several disciplines in materials' research, including polymer science, surfaces, membranes, biomaterials' microfabrication, and flexible electronics. If successful, these efforts could lead to new therapies for chronic disease and are a rich space for both fundamental materials' discovery and applied translational science. Graphical Abstract
Collapse
Affiliation(s)
- Siddharth R. Krishnan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA USA
| | - Daniel G. Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA USA
| |
Collapse
|
35
|
Zhang H, Wei Y, Wang Y, Liang J, Hou Y, Nie X, Hou J. Emerging Diabetes Therapies: Regenerating Pancreatic β Cells. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:644-656. [PMID: 39276101 DOI: 10.1089/ten.teb.2024.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
The incidence of diabetes mellitus (DM) is steadily increasing annually, with 537 million diabetic patients as of 2021. Restoring diminished β cell mass or impaired islet function is crucial in treating DM, particularly type 1 DM. However, the regenerative capacity of islet β cells, which primarily produce insulin, is severely limited, and natural regeneration is only observed in young rodents or children. Hence, there is an urgent need to develop advanced therapeutic approaches that can regenerate endogenous β cells or replace them with stem cell (SC)-derived or engineered β-like cells. Current strategies for treating insulin-dependent DM mainly include promoting the self-replication of endogenous β cells, inducing SC differentiation, reprogramming non-β cells into β-like cells, and generating pancreatic-like organoids through cell-based intervention. In this Review, we discuss the current state of the art in these approaches, describe associated challenges, propose potential solutions, and highlight ongoing efforts to optimize β cell or islet transplantation and related clinical trials. These effective cell-based therapies will generate a sustainable source of functional β cells for transplantation and lay strong foundations for future curative treatments for DM.
Collapse
Affiliation(s)
- Haojie Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yaxin Wei
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yubo Wang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jialin Liang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yifan Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
- Department of Urinary Surgery, Henan Provincial Research Center for the Prevention and Diagnosis of Prostate Diseases, Huaihe Hospital, Henan University, Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Department of Urinary Surgery, Henan Provincial Research Center for the Prevention and Diagnosis of Prostate Diseases, Huaihe Hospital, Henan University, Kaifeng, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
| |
Collapse
|
36
|
Pliner L, Laneret N, Roudaut M, Mogrovejo-Valdivia A, Vandenhaute E, Maubon N, Toillon RA, Karrout Y, Treizebre A, Annicotte JS. Mechanical and functional characterisation of a 3D porous biomimetic extracellular matrix to study insulin secretion from pancreatic β-cell lines. IN VITRO MODELS 2024; 3:205-218. [PMID: 39872697 PMCID: PMC11756461 DOI: 10.1007/s44164-024-00078-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 01/30/2025]
Abstract
Background Extracellular matrix (ECM) is a three-dimensional (3D) structure found around cells in the tissues of many organisms. It is composed mainly of fibrous proteins, such as collagen and elastin, and adhesive glycoproteins, such as fibronectin and laminin-as well as proteoglycans, such as hyaluronic acid. The ECM performs several essential functions, including structural support of tissues, regulation of cell communication, adhesion, migration, and differentiation by providing biochemical and biomechanical cues to the cells. Pancreatic β-cells have been previously shown to be responsive to the surrounding mechanical stress, impacting their insulin-secreting function. Purpose We aimed to derive a physiologically relevant in vitro model of pancreatic tissue by using an innovative synthesised porous ECM that mimics the native tissue microenvironment and mechanical properties. Methods Here we performed mechanical, physico-chemical and functional characterisation of a synthetic hydrogel ECM, composed of hyaluronic acid cross-linked with collagen types I and VI and modified with fibronectin. The hydrogel was used as a 3D cell culture scaffold for the MIN6 insulinoma cell line. Cell proliferation, viability, gene expression, and insulin secretion in response to glucose stimulus were assessed and contrasted with classic monolayer culture. Results The biomaterial exhibited a shear modulus of 815.37 kPa and a distinctive viscoelastic response. MIN6 cells showed a higher proliferation and viability rates and maintained insulin secretion in response to glucose stimulus and β-cell identity gene expression when cultured in the 3D hydrogel compared to monolayer culture. Conclusion Our study demonstrated the potential of this biomimetic hydrogel scaffold as an innovative matrix enabling better in vitro models to study disease physiopathology. Supplementary Information The online version contains supplementary material available at 10.1007/s44164-024-00078-z.
Collapse
Affiliation(s)
- Leonid Pliner
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, U1167 - RID-AGE - Facteurs de Risque Et Déterminants Moléculaires Des Maladies Liées Au Vieillissement, F-59000 Lille, France
| | - Nathan Laneret
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 – CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| | | | | | | | | | - Robert-Alain Toillon
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 – CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Youness Karrout
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - Anthony Treizebre
- Univ. Lille, CNRS, Univ Polytechnique Hauts-de-France, Junia, UMR 8520 - IEMN – Institut d’Électronique de Microélectronique et de Nanotechnologie, F-59000 Lille, France
| | - Jean-Sébastien Annicotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, U1167 - RID-AGE - Facteurs de Risque Et Déterminants Moléculaires Des Maladies Liées Au Vieillissement, F-59000 Lille, France
| |
Collapse
|
37
|
Alver CG, Dominguez-Bendala J, Agarwal A. Engineered tools to study endocrine dysfunction of pancreas. BIOPHYSICS REVIEWS 2024; 5:041303. [PMID: 39449867 PMCID: PMC11498943 DOI: 10.1063/5.0220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Pancreas, a vital organ with intricate endocrine and exocrine functions, is central to the regulation of the body's glucose levels and digestive processes. Disruptions in its endocrine functions, primarily regulated by islets of Langerhans, can lead to debilitating diseases such as diabetes mellitus. Murine models of pancreatic dysfunction have contributed significantly to the understanding of insulitis, islet-relevant immunological responses, and the optimization of cell therapies. However, genetic differences between mice and humans have severely limited their clinical translational relevance. Recent advancements in tissue engineering and microfabrication have ushered in a new era of in vitro models that offer a promising solution. This paper reviews the state-of-the-art engineered tools designed to study endocrine dysfunction of the pancreas. Islet on a chip devices that allow precise control of various culture conditions and noninvasive readouts of functional outcomes have led to the generation of physiomimetic niches for primary and stem cell derived islets. Live pancreatic slices are a new experimental tool that could more comprehensively recapitulate the complex cellular interplay between the endocrine and exocrine parts of the pancreas. Although a powerful tool, live pancreatic slices require more complex control over their culture parameters such as local oxygenation and continuous removal of digestive enzymes and cellular waste products for maintaining experimental functionality over long term. The combination of islet-immune and slice on chip strategies can guide the path toward the next generation of pancreatic tissue modeling for better understanding and treatment of endocrine pancreatic dysfunctions.
Collapse
Affiliation(s)
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ashutosh Agarwal
- Author to whom correspondence should be addressed:. Tel.: +1 305 243-8925
| |
Collapse
|
38
|
Ning X, Munir KM, Davis SN. Drugs stimulating insulin secretion in early clinical development for the treatment of type 1 diabetes: what's new? Expert Opin Investig Drugs 2024; 33:1199-1208. [PMID: 39645243 DOI: 10.1080/13543784.2024.2439501] [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: 01/25/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Type 1 diabetes is a chronic autoimmune condition characterized by the selective destruction of insulin-producing beta cells in the pancreas. The etiology of T1D is multifactorial, with a combination of genetic susceptibility and environmental triggers believed to underlie beta-cell destruction. Preserving and prolonging beta-cell function in T1D is a pivotal therapeutic objective that can mitigate disease progression and improve glycemic control. AREAS COVERED Insulin secretagogues have long been used in the management of type 2 diabetes, but do not have a significant beneficial effect in individuals with long-standing type 1 diabetes. Enhancement of beta-cell function early in the course of type 1 diabetes may offer important benefits in glycemic control and reduced hypoglycemia risk. Glucagon-like peptide-1 receptor agonists, glucokinase activators, free fatty acid receptor agonists, and glimins are drug classes which may offer benefit in enhancing insulin secretion in individuals with type 1 diabetes. EXPERT OPINION Drugs which enhance insulin secretion in individuals may offer clinical benefits to individuals with type 1 diabetes. However, the lack of beta-cell capacity introduces a challenge without regeneration of insulin-producing cells. Stem cell therapies combined with regulation of islet autoimmunity may offer the best prospect of increased insulin secretion in individuals with T1D.
Collapse
Affiliation(s)
- Xinyuan Ning
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Kashif M Munir
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Stephen N Davis
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Sevastianov VI, Ponomareva AS, Baranova NV, Belova AD, Kirsanova LA, Nikolskaya AO, Kuznetsova EG, Chuykova EO, Skaletskiy NN, Skaletskaya GN, Nemets EA, Basok YB, Gautier SV. A Tissue-Engineered Construct Based on a Decellularized Scaffold and the Islets of Langerhans: A Streptozotocin-Induced Diabetic Model. Life (Basel) 2024; 14:1505. [PMID: 39598303 PMCID: PMC11595861 DOI: 10.3390/life14111505] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Producing a tissue-engineered pancreas based on a tissue-specific scaffold from a decellularized pancreas, imitating the natural pancreatic tissue microenvironment and the islets of Langerhans, is one of the approaches to treating patients with type 1 diabetes mellitus (T1DM). The aim of this work was to investigate the ability of a fine-dispersed tissue-specific scaffold (DP scaffold) from decellularized human pancreas fragments to support the islets' survival and insulin-producing function when injected in a streptozotocin-induced diabetic rat model. The developed decellularization protocol allows us to obtain a scaffold with a low DNA content (33 [26; 38] ng/mg of tissue, p < 0.05) and with the preservation of GAGs (0.92 [0.84; 1.16] µg/mg, p < 0.05) and fibrillar collagen (273.7 [241.2; 303.0] µg/mg, p < 0.05). Rat islets of Langerhans were seeded in the obtained scaffolds. The rats with stable T1DM were treated by intraperitoneal injections of rat islets alone and islets seeded on the DP scaffold. The blood glucose level was determined for 10 weeks with a histological examination of experimental animals' pancreas. A more pronounced decrease in the recipient rats' glycemia was detected after comparing the islets seeded on the DP scaffold with the control injection (by 71.4% and 51.2%, respectively). It has been shown that the DP scaffold facilitates a longer survival and the efficient function of pancreatic islets in vivo and can be used to engineer a pancreas.
Collapse
Affiliation(s)
- Victor I. Sevastianov
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
- The Institute of Biomedical Research and Technology (IBRT), Autonomous Non-Profit Organization, 123557 Moscow, Russia
| | - Anna S. Ponomareva
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Natalia V. Baranova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Aleksandra D. Belova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Lyudmila A. Kirsanova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Alla O. Nikolskaya
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Eugenia G. Kuznetsova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Elizaveta O. Chuykova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
- The Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Nikolay N. Skaletskiy
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Galina N. Skaletskaya
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Evgeniy A. Nemets
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Yulia B. Basok
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Sergey V. Gautier
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
- The Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| |
Collapse
|
40
|
Chuang ST, Alcazar O, Watts B, Abdulreda MH, Buchwald P. Small-molecule inhibitors of the CD40-CD40L costimulatory interaction are effective in pancreatic islet transplantation and prevention of type 1 diabetes models. Front Immunol 2024; 15:1484425. [PMID: 39606229 PMCID: PMC11599200 DOI: 10.3389/fimmu.2024.1484425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
As part of our work to develop small-molecule inhibitors (SMIs) of the CD40-CD40L(CD154) costimulatory protein-protein interaction, here, we describe the ability of two of our most promising SMIs, DRI-C21041 and DRI-C21095, to prolong the survival and function of islet allografts in two murine models of islet transplantation (under the kidney capsule and in the anterior chamber of the eye) and to prevent autoimmune type 1 diabetes (T1D) onset in NOD mice. In both transplant models, a significant portion of islet allografts (50%-80%) remained intact and functional long after terminating treatment, suggesting the possibility of inducing operational immune tolerance via inhibition of the CD40-CD40L axis. SMI-treated mice maintained the structural integrity and function of their islet allografts with concomitant reduction in immune cell infiltration as evidenced by direct longitudinal imaging in situ. Furthermore, in female NODs, three-month SMI treatment reduced the incidence of diabetes from 80% to 60% (DRI-C21041) and 25% (DRI-C21095). These results (i) demonstrate the susceptibility of this TNF superfamily protein-protein interaction to small-molecule inhibition, (ii) confirm the in vivo therapeutic potential of these SMIs of a critical immune checkpoint, and (iii) reaffirm the therapeutic promise of CD40-CD40L blockade in islet transplantation and T1D prevention. Thus, CD40L-targeting SMIs could ultimately lead to alternative immunomodulatory therapeutics for transplant recipients and prevention of autoimmune diseases that are safer, less immunogenic, more controllable (shorter half-lives), and more patient-friendly (i.e., suitable for oral administration, which makes them easier to administer) than corresponding antibody-based interventions.
Collapse
Affiliation(s)
- Sung-Ting Chuang
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Oscar Alcazar
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Brandon Watts
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Midhat H. Abdulreda
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
41
|
Wang S, Du Y, Zhang B, Meng G, Liu Z, Liew SY, Liang R, Zhang Z, Cai X, Wu S, Gao W, Zhuang D, Zou J, Huang H, Wang M, Wang X, Wang X, Liang T, Liu T, Gu J, Liu N, Wei Y, Ding X, Pu Y, Zhan Y, Luo Y, Sun P, Xie S, Yang J, Weng Y, Zhou C, Wang Z, Wang S, Deng H, Shen Z. Transplantation of chemically induced pluripotent stem-cell-derived islets under abdominal anterior rectus sheath in a type 1 diabetes patient. Cell 2024; 187:6152-6164.e18. [PMID: 39326417 DOI: 10.1016/j.cell.2024.09.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/25/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024]
Abstract
We report the 1-year results from one patient as the preliminary analysis of a first-in-human phase I clinical trial (ChiCTR2300072200) assessing the feasibility of autologous transplantation of chemically induced pluripotent stem-cell-derived islets (CiPSC islets) beneath the abdominal anterior rectus sheath for type 1 diabetes treatment. The patient achieved sustained insulin independence starting 75 days post-transplantation. The patient's time-in-target glycemic range increased from a baseline value of 43.18% to 96.21% by month 4 post-transplantation, accompanied by a decrease in glycated hemoglobin, an indicator of long-term systemic glucose levels at a non-diabetic level. Thereafter, the patient presented a state of stable glycemic control, with time-in-target glycemic range at >98% and glycated hemoglobin at around 5%. At 1 year, the clinical data met all study endpoints with no indication of transplant-related abnormalities. Promising results from this patient suggest that further clinical studies assessing CiPSC-islet transplantation in type 1 diabetes are warranted.
Collapse
Affiliation(s)
- Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China.
| | - Yuanyuan Du
- School of Basic Medical Sciences, MOE Engineering Research Center of Regenerative Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100191, China; Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Boya Zhang
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Gaofan Meng
- School of Basic Medical Sciences, MOE Engineering Research Center of Regenerative Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100191, China; Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Zewen Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Soon Yi Liew
- School of Basic Medical Sciences, MOE Engineering Research Center of Regenerative Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100191, China; Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Zhengyuan Zhang
- School of Basic Medical Sciences, MOE Engineering Research Center of Regenerative Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100191, China
| | - Xiangheng Cai
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | | | - Wei Gao
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | | | - Jiaqi Zou
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Hui Huang
- Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Mingyang Wang
- Department of Ultrasound, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | | | - Xuelian Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Ting Liang
- Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Tengli Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Jiabin Gu
- Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Na Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Yanling Wei
- Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Xuejie Ding
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Yue Pu
- Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Yixiang Zhan
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Yu Luo
- Hangzhou Reprogenix Bioscience, Hangzhou, China
| | - Peng Sun
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Shuangshuang Xie
- Radiology Department, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Jiuxia Yang
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Yiqi Weng
- Department of Anesthesiology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Chunlei Zhou
- Department of Medical Laboratory, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Zhenglu Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Shuang Wang
- Department of Plastic and Burn, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Hongkui Deng
- School of Basic Medical Sciences, MOE Engineering Research Center of Regenerative Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100191, China; China Changping Laboratory, Beijing 102206, China.
| | - Zhongyang Shen
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, School of Medicine, Nankai University, NHC Key Laboratory for Critical Care Medicine, Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China.
| |
Collapse
|
42
|
Langlois A, Pinget M, Kessler L, Bouzakri K. Islet Transplantation: Current Limitations and Challenges for Successful Outcomes. Cells 2024; 13:1783. [PMID: 39513890 PMCID: PMC11544954 DOI: 10.3390/cells13211783] [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: 09/26/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Islet transplantation is a promising approach for treating patients with unstable T1DM. However, it is confronted with numerous obstacles throughout the various stages of the transplantation procedure. Significant progress has been made over the last 25 years in understanding the mechanisms behind the loss of functional islet mass and in developing protective strategies. Nevertheless, at present, two to three pancreases are still needed to treat a single patient, which limits the maximal number of patients who can benefit from islet transplantation. Thus, this publication provides an overview of recent scientific findings on the various issues affecting islet transplantation. Specifically, we will focus on the understanding of the mechanisms involved and the strategies developed to alleviate these problems from the isolation stage to the post-transplantation phase. Finally, we hope that this review will highlight new avenues of action, enabling us to propose pancreatic islet transplantation to a maximum number of patients with T1DM.
Collapse
Affiliation(s)
- Allan Langlois
- UR «Diabète et Thérapeutiques», Centre Européen d’Étude du Diabète, Université de Strasbourg, Boulevard René Leriche, 67200 Strasbourg, France; (A.L.); (M.P.)
| | - Michel Pinget
- UR «Diabète et Thérapeutiques», Centre Européen d’Étude du Diabète, Université de Strasbourg, Boulevard René Leriche, 67200 Strasbourg, France; (A.L.); (M.P.)
| | - Laurence Kessler
- Department of Endocrinology, Diabetes and Nutrition, University Hospital of Strasbourg, 67200 Strasbourg, France;
- Inserm UMR 1260, Nanomédicine Regenerative, University of Strasbourg, 67085 Strasbourg, France
| | - Karim Bouzakri
- UR «Diabète et Thérapeutiques», Centre Européen d’Étude du Diabète, Université de Strasbourg, Boulevard René Leriche, 67200 Strasbourg, France; (A.L.); (M.P.)
| |
Collapse
|
43
|
Moon JE, Lee YN, Jeong S, Jun HR, Hoang MH, Jo Y, Jang J, Shim IK, Kim SC. Enhancing differentiation and functionality of insulin-producing cells derived from iPSCs using esterified collagen hydrogel for cell therapy in diabetes mellitus. Stem Cell Res Ther 2024; 15:374. [PMID: 39443977 PMCID: PMC11515471 DOI: 10.1186/s13287-024-03971-2] [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: 03/25/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Islet transplantation is a recommended treatment for type 1 diabetes but is limited by donor organ shortage. This study introduces an innovative approach for improving the differentiation and functionality of insulin-producing cells (IPCs) from iPSCs using 3D spheroid formation and hydrogel matrix as an alternative pancreatic islet source. The extracellular matrix (ECM) is crucial for pancreatic islet functionality, but finding the ideal matrix for β-cell differentiation has been challenging. We aimed to advance IPC differentiation and maturation through an esterified collagen hydrogel, comparing its effectiveness with conventional basement membrane extract (BME) hydrogels. METHODS iPSCs were differentiated into IPCs using a small molecule-based sequential protocol, followed by spheroid formation in concave microwells. Rheological analysis, scanning electron microscopy, and proteomic profiling were used to characterize the chemical and physical properties of each matrix. IPCs, both in single-cell form and as spheroids, were embedded in either ionized collagen or BME hydrogels, which was followed by assessments of morphological changes, pancreatic islet-related gene expression, insulin secretion, and pathway activation using comprehensive analytical techniques. RESULTS Esterified collagen hydrogels markedly improved the structural integrity, insulin expression, and cell-cell interactions in IPC spheroids, forming densely packed insulin-expressing clusters, in contrast to the dispersed cells observed in BME cultures. Collagen hydrogel significantly enhanced the mRNA expression of crucial endocrine markers and maturation factors, with IPC spheroids showing accelerated differentiation from day 5, suggesting a faster differentiation compared to single cells in hydrogel encapsulation. Insulin secretion in response to glucose in collagen environments, with a GSIS index of 2.46 ± 0.05, exceeded those in 2D and BME, demonstrating superior pancreatic islet functionality. Pathway analysis highlighted enhanced insulin secretion capabilities, evidenced by the upregulation of genes like Secretogranin III and Chromogranin A in collagen cultures. In vivo transplantation results showed that collagen hydrogel enhanced cluster integrity, tissue integration, and insulin secretion compared to non-embedded IPCs and BME groups. CONCLUSION Esterified collagen hydrogels demonstrated superior efficacy over 2D and BME in promoting IPC differentiation and maturation, possibly through upregulation of the expression of key secretion pathway genes. Our findings suggest that using collagen hydrogels presents a promising approach to enhance insulin secretion efficiency in differentiating pancreatic β-cells, advancing cell therapy in diabetes cell therapy.
Collapse
Affiliation(s)
- Ji Eun Moon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Yu Na Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Sehui Jeong
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hye Ryeong Jun
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Minh Hien Hoang
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering (POSTECH), Pohang, 37673, Republic of Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering (POSTECH), Pohang, 37673, Republic of Korea
- Department of Mechanical Engineering (POSTECH), Pohang, 37673, Republic of Korea
| | - In Kyong Shim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Department of Biomedical Engineering, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Song Cheol Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Brain Korea 21 Project, Seoul, Korea.
| |
Collapse
|
44
|
Ray S, Palui R. Immunotherapy in type 1 diabetes: Novel pathway to the future ahead. World J Diabetes 2024; 15:2022-2035. [PMID: 39493558 PMCID: PMC11525730 DOI: 10.4239/wjd.v15.i10.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/26/2024] Open
Abstract
Since the discovery of insulin over 100 years ago, the focus of research in the management of type 1 diabetes (T1D) has centered around glycemic control and management of complications rather than the prevention of autoimmune destruction of pancreatic β cells. Fortunately, in recent years, there has been significant advancement in immune-targeted pharmacotherapy to halt the natural progression of T1D. The immune-targeted intervention aims to alter the underlying pathogenesis of T1D by targeting different aspects of the immune system. The immunotherapy can either antagonize the immune mediators like T cells, B cells or cytokines (antibody-based therapy), or reinduce self-tolerance to pancreatic β cells (antigen-based therapy) or stem-cell treatment. Recently, the US Food and Drug Administration approved the first immunotherapy teplizumab to be used only in stage 2 of T1D. However, the window of opportunity to practically implement this approved molecule in the selected target population is limited. In this Editorial, we briefly discuss the various promising recent developments in the field of immunotherapy research in T1D. However, further studies of these newer therapeutic agents are needed to explore their true potential for prevention or cure of T1D.
Collapse
Affiliation(s)
- Sayantan Ray
- Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar 751019, India
| | - Rajan Palui
- Department of Endocrinology, The Mission Hospital, Durgapur 713212, India
| |
Collapse
|
45
|
Wang LH, Marfil-Garza BA, Ernst AU, Pawlick RL, Pepper AR, Okada K, Epel B, Viswakarma N, Kotecha M, Flanders JA, Datta AK, Gao HJ, You YZ, Ma M, Shapiro AMJ. Inflammation-induced subcutaneous neovascularization for the long-term survival of encapsulated islets without immunosuppression. Nat Biomed Eng 2024; 8:1266-1284. [PMID: 38052996 DOI: 10.1038/s41551-023-01145-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/25/2023] [Indexed: 12/07/2023]
Abstract
Cellular therapies for type-1 diabetes can leverage cell encapsulation to dispense with immunosuppression. However, encapsulated islet cells do not survive long, particularly when implanted in poorly vascularized subcutaneous sites. Here we show that the induction of neovascularization via temporary controlled inflammation through the implantation of a nylon catheter can be used to create a subcutaneous cavity that supports the transplantation and optimal function of a geometrically matching islet-encapsulation device consisting of a twisted nylon surgical thread coated with an islet-seeded alginate hydrogel. The neovascularized cavity led to the sustained reversal of diabetes, as we show in immunocompetent syngeneic, allogeneic and xenogeneic mouse models of diabetes, owing to increased oxygenation, physiological glucose responsiveness and islet survival, as indicated by a computational model of mass transport. The cavity also allowed for the in situ replacement of impaired devices, with prompt return to normoglycemia. Controlled inflammation-induced neovascularization is a scalable approach, as we show with a minipig model, and may facilitate the clinical translation of immunosuppression-free subcutaneous islet transplantation.
Collapse
Affiliation(s)
- Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Braulio A Marfil-Garza
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrey, Mexico
| | - Alexander U Ernst
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Rena L Pawlick
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew R Pepper
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Kento Okada
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Boris Epel
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
- O2M Technologies, LLC, Chicago, IL, USA
| | | | | | | | - Ashim K Datta
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Hong-Jie Gao
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Ye-Zi You
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA.
| | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
46
|
Keymeulen B, De Groot K, Jacobs-Tulleneers-Thevissen D, Thompson DM, Bellin MD, Kroon EJ, Daniels M, Wang R, Jaiman M, Kieffer TJ, Foyt HL, Pipeleers D. Encapsulated stem cell-derived β cells exert glucose control in patients with type 1 diabetes. Nat Biotechnol 2024; 42:1507-1514. [PMID: 38012450 PMCID: PMC11471599 DOI: 10.1038/s41587-023-02055-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/05/2023] [Indexed: 11/29/2023]
Abstract
Clinical studies on the treatment of type 1 diabetes with device-encapsulated pancreatic precursor cells derived from human embryonic stem cells found that insulin output was insufficient for clinical benefit. We are conducting a phase 1/2, open-label, multicenter trial aimed at optimizing cell engraftment (ClinicalTrials.gov identifier: NCT03163511 ). Here we report interim, 1-year outcomes in one study group that received 2-3-fold higher cell doses in devices with an optimized membrane perforation pattern. β cell function was measured by meal-stimulated plasma C-peptide levels at 3-month intervals, and the effect on glucose control was assessed by continuous glucose monitoring (CGM) and insulin dosing. Of 10 patients with undetectable baseline C-peptide, three achieved levels ≥0.1 nmol l-1 from month 6 onwards that correlated with improved CGM measures and reduced insulin dosing, indicating a glucose-controlling effect. The patient with the highest C-peptide (0.23 nmol l-1) increased CGM time-in-range from 55% to 85% at month 12; β cell mass in sentinel devices in this patient at month 6 was 4% of the initial cell mass, indicating directions for improving efficacy.
Collapse
Affiliation(s)
- Bart Keymeulen
- Diabetes Research Center, Vrije Universiteit Brussel and Universitair Ziekenhuis Brussel, Brussels, Belgium.
| | - Kaat De Groot
- Diabetes Research Center, Vrije Universiteit Brussel and Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | - David M Thompson
- Division of Endocrinology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Melena D Bellin
- Department of Pediatrics and Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN, USA
| | | | | | | | | | - Timothy J Kieffer
- ViaCyte Inc., San Diego, CA, USA
- Department of Cellular and Physiological Sciences and Department of Surgery, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | | | - Daniel Pipeleers
- Diabetes Research Center, Vrije Universiteit Brussel and Universitair Ziekenhuis Brussel, Brussels, Belgium.
| |
Collapse
|
47
|
Yue L, Li J, Yao M, Song S, Zhang X, Wang Y. Cutting edge of immune response and immunosuppressants in allogeneic and xenogeneic islet transplantation. Front Immunol 2024; 15:1455691. [PMID: 39346923 PMCID: PMC11427288 DOI: 10.3389/fimmu.2024.1455691] [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: 06/27/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
As an effective treatment for diabetes, islet transplantation has garnered significant attention and research in recent years. However, immune rejection and the toxicity of immunosuppressive drugs remain critical factors influencing the success of islet transplantation. While immunosuppressants are essential in reducing immune rejection reactions and can significantly improve the survival rate of islet transplants, improper use of these drugs can markedly increase mortality rates following transplantation. Additionally, the current availability of islet organ donations fails to meet the demand for organ transplants, making xenotransplantation a crucial method for addressing organ shortages. This review will cover the following three aspects: 1) the immune responses occurring during allogeneic islet transplantation, including three stages: inflammation and IBMIR, allogeneic immune response, and autoimmune recurrence; 2) commonly used immunosuppressants in allogeneic islet transplantation, including calcineurin inhibitors (Cyclosporine A, Tacrolimus), mycophenolate mofetil, glucocorticoids, and Bortezomib; and 3) early and late immune responses in xenogeneic islet transplantation and the immune effects of triple therapy (ECDI-fixed donor spleen cells (ECDI-SP) + anti-CD20 + Sirolimus) on xenotransplantation.
Collapse
Affiliation(s)
- Liting Yue
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jisong Li
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingjun Yao
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Xiaoqin Zhang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
48
|
Jang D, Matthews K, Deng P, Berryman SG, Nian C, Duffy SP, Lynn FC, Ma H. Single cell glucose-stimulated insulin secretion assay using nanowell-in-microwell plates. LAB ON A CHIP 2024; 24:4232-4241. [PMID: 39212929 DOI: 10.1039/d4lc00413b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pancreatic β cells secrete insulin in response to elevated levels of glucose. Stem cell derived β (SCβ) cells aim to replicate this glucose-stimulated insulin secretion (GSIS) function, but current preparations cannot provide the same level of insulin as natural β cells. Here, we develop an assay to measure GSIS at the single cell level to investigate the functional heterogeneity of SCβ cells and donor-derived islet cells. Our assay involves randomly depositing single cells and insulin capture microbeads in open-top nanowells (40 × 40 × 55 μm3) fabricated on glass-bottom imaging microwell plates. Insulin secreted from single cells is captured on microbeads and then stained using a detection antibody. The nanowell microstructure limits diffusion of secreted insulin. The glass substrate provides an optically flat surface for quantitative microscopy to measure the concentration of secreted insulin. We used this approach to measure GSIS from SCβ cells and donor-derived islet cells after 15 minutes exposure to 3.3 mM and 16.7 mM glucose. Both cell types exhibited significant GSIS heterogeneity, where elite cells (<20%) produced the majority of the secreted insulin (55-78%). This assay provides an immediate readout of single cell glucose-stimulated insulin secretion in a flexible well plate-based format.
Collapse
Affiliation(s)
- Deasung Jang
- Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Kerryn Matthews
- Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Pan Deng
- Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Samuel G Berryman
- Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Cuilan Nian
- BC Children's Hospital Research Institute CFKF Diabetes Research Program, Vancouver, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Francis C Lynn
- BC Children's Hospital Research Institute CFKF Diabetes Research Program, Vancouver, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Hongshen Ma
- Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
49
|
Inagaki NF, Oki Y, Ikeda S, Tulakarnwong S, Shinohara M, Inagaki FF, Ohta S, Kokudo N, Sakai Y, Ito T. Transplantation of pancreatic beta-cell spheroids in mice via non-swellable hydrogel microwells composed of poly(HEMA- co-GelMA). Biomater Sci 2024; 12:4354-4362. [PMID: 38967234 DOI: 10.1039/d4bm00295d] [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: 07/06/2024]
Abstract
Pancreatic islet transplantation is an effective treatment for type I diabetes mellitus. However, many problems associated with pancreatic islet engraftment remain unresolved. In this study, we developed a hydrogel microwell device for islet implantation, fabricated by crosslinking gelatin-methacryloyl (GelMA) and 2-hydroxyethyl methacrylate (HEMA) in appropriate proportions. The fabricated hydrogel microwell device could be freeze-dried and restored by immersion in the culture medium at any time, allowing long-term storage and transport of the device for ready-to-use applications. In addition, due to its non-swelling properties, the shape of the wells of the device was maintained. Thus, the device allowed pancreatic β cell lines to form spheroids and increase insulin secretion. Intraperitoneal implantation of the β cell line-seeded GelMA/HEMA hydrogel microwell device reduced blood glucose levels in diabetic mice. In addition, they were easy to handle during transplantation and were removed from the transplant site without peritoneal adhesions or infiltration by inflammatory cells. These results suggest that the GelMA/HEMA hydrogel microwell device can go from spheroid and/or organoid fabrication to transplantation in a single step.
Collapse
Affiliation(s)
- Natsuko F Inagaki
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan.
| | - Yuichiro Oki
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan.
| | - Shunsuke Ikeda
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Sarun Tulakarnwong
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Marie Shinohara
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Fuyuki F Inagaki
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Seiichi Ohta
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
- Institute of Engineering Innovation, The University of Tokyo, Tokyo, Japan
| | - Norihiro Kokudo
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Taichi Ito
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
50
|
Toftdal MS, Christensen NP, Kadumudi FB, Dolatshahi-Pirouz A, Grunnet LG, Chen M. Mechanically reinforced hydrogel vehicle delivering angiogenic factor for beta cell therapy. J Colloid Interface Sci 2024; 667:54-63. [PMID: 38615623 DOI: 10.1016/j.jcis.2024.04.050] [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: 10/12/2023] [Revised: 03/18/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic disease affecting millions worldwide. Insulin therapy is currently the golden standard for treating T1DM; however, it does not restore the normal glycaemic balance entirely, which increases the risk of secondary complications. Beta-cell therapy may be a possible way of curing T1DM and has already shown promising results in the clinic. However, low retention rates, poor cell survival, and limited therapeutic potential are ongoing challenges, thus increasing the need for better cell encapsulation devices. This study aimed to develop a mechanically reinforced vascular endothelial growth factor (VEGF)-delivering encapsulation device suitable for beta cell encapsulation and transplantation. Poly(l-lactide-co-ε-caprolactone) (PLCL)/gelatin methacryloyl (GelMA)/alginate coaxial nanofibres were produced using electrospinning and embedded in an alginate hydrogel. The encapsulation device was physically and biologically characterised and was found to be suitable for INS-1E beta cell encapsulation, vascularization, and transplantation in terms of its biocompatibility, porosity, swelling ratio and mechanical properties. Lastly, VEGF was incorporated into the hydrogel and the release kinetics and functional studies revealed a sustained release of bioactive VEGF for at least 14 days, making the modified alginate system a promising candidate for improving the beta cell survival after transplantation.
Collapse
Affiliation(s)
- Mette Steen Toftdal
- Department of Biological and Chemical Engineering, Aarhus University, 8000 Aarhus C, Denmark; Department of Cell Formulation and Delivery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | | - Firoz Babu Kadumudi
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | | | - Lars Groth Grunnet
- Department of Cell Formulation and Delivery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Menglin Chen
- Department of Biological and Chemical Engineering, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|