1
|
Shalaby MS, Abdel-Reheim ES, Almanaa TN, Alhaber LA, Nabil A, Ahmed OM, Elwan M, Abdel-Moneim A. Therapeutic effects of mesenchymal stem cell conditioned media on streptozotocin-induced diabetes in Wistar rats. Regen Ther 2025; 28:1-11. [PMID: 39678398 PMCID: PMC11638607 DOI: 10.1016/j.reth.2024.11.004] [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: 05/21/2024] [Revised: 07/02/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Cell-based therapy is a new direction of treatment of diseases such as type 1 diabetes mellitus (T1DM); but unfortunately, its severe side effects include immunogenicity and tumor development. Using Mesenchymal stem cells conditioned medium (MSCs-CM) may be an alternative therapy to avoid stem cell risks, preserving effectiveness and demonstrating noticeably increased levels of cytokines, angiogenic factors, and growth factors that encourage and support regenerative processes. In the current work, we examined the effects of MSCs-CM injected in tail vein and pancreas directly compared with the standard antidiabetic drug, glimepiride in streptozotocin-induced type 1 diabetic rats. Fifty adults Male Wistar rats were allocated equally into five groups: normal, diabetic control and three diabetic groups treated respectively with glimepiride, MSCs-CM injected daily into tail vein (MSCs-CMT) and MSCs-CM injected directly in pancreas (MSCs-CMP); all treatments continued for 28 days. The treatments produced a significant improvement in blood glucose level and glycosylated hemoglobin A1c (HbA1c), serum insulin level and lipid panel, and pancreas apoptosis-related markers including B cell lymphoma-2 (Bcl-2) and vimentin. In addition, the treatments resulted in suppression in the oxidation stress and enhancement in the antioxidant, which were manifested by the suppressed lipid peroxidation and the increased antioxidant markers (glutathione, catalase and superoxide dismutase) in the pancreas. In association with the significant decrease in tumour necrosis factor-alpha (TNF-α) and interleukin-1beta (IL-1β) and a significant increase in interleukin-10 (IL-10) levels, the inflammatory mediator nuclear factor-kappa B (NF-κB) expression was significantly decreased by MSCs-CMT and MSCs-CMP. The histological amelioration of the pancreatic islet cells assured our study especially in MSCs-CMP group than MSCs-CMT which supports islet regeneration and elevated circulating insulin. These results imply that MSCs-CM infusion has therapeutic benefits in T1DM rats and may be a viable novel therapeutic approach; MSCs-CMP was shown to be more effective than glimepiride and MSCs-CMT. The mechanisms of antidiabtic actions may be mediated via the antioxidant, anti-apoptotic and anti-inflammatory effects.
Collapse
Affiliation(s)
- Marwa S. Shalaby
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| | - Eman S. Abdel-Reheim
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Taghreed N. Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Lama Abdulaziz Alhaber
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nabil
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| | - Osama M. Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Mariam Elwan
- Heath Affairs Directorate, Egyptian Ministry of Health, Mansoura, Egypt
| | - Adel Abdel-Moneim
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
2
|
Grattoni A, Korbutt G, Tomei AA, García AJ, Pepper AR, Stabler C, Brehm M, Papas K, Citro A, Shirwan H, Millman JR, Melero-Martin J, Graham M, Sefton M, Ma M, Kenyon N, Veiseh O, Desai TA, Nostro MC, Marinac M, Sykes M, Russ HA, Odorico J, Tang Q, Ricordi C, Latres E, Mamrak NE, Giraldo J, Poznansky MC, de Vos P. Harnessing cellular therapeutics for type 1 diabetes mellitus: progress, challenges, and the road ahead. Nat Rev Endocrinol 2025; 21:14-30. [PMID: 39227741 DOI: 10.1038/s41574-024-01029-0] [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] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a growing global health concern that affects approximately 8.5 million individuals worldwide. T1DM is characterized by an autoimmune destruction of pancreatic β cells, leading to a disruption in glucose homeostasis. Therapeutic intervention for T1DM requires a complex regimen of glycaemic monitoring and the administration of exogenous insulin to regulate blood glucose levels. Advances in continuous glucose monitoring and algorithm-driven insulin delivery devices have improved the quality of life of patients. Despite this, mimicking islet function and complex physiological feedback remains challenging. Pancreatic islet transplantation represents a potential functional cure for T1DM but is hindered by donor scarcity, variability in harvested cells, aggressive immunosuppressive regimens and suboptimal clinical outcomes. Current research is directed towards generating alternative cell sources, improving transplantation methods, and enhancing cell survival without chronic immunosuppression. This Review maps the progress in cell replacement therapies for T1DM and outlines the remaining challenges and future directions. We explore the state-of-the-art strategies for generating replenishable β cells, cell delivery technologies and local targeted immune modulation. Finally, we highlight relevant animal models and the regulatory aspects for advancing these technologies towards clinical deployment.
Collapse
Affiliation(s)
- Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| | - Gregory Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrew R Pepper
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Cherie Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
- Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Michael Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Klearchos Papas
- Department of Surgery, The University of Arizona, Tucson, AZ, USA
| | - Antonio Citro
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Haval Shirwan
- Department of Pediatrics, Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Juan Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Melanie Graham
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Michael Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Norma Kenyon
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Tejal A Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA, USA
- Brown University, School of Engineering, Providence, RI, USA
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Megan Sykes
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University, New York, NY, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jon Odorico
- UW Health Transplant Center, Madison, WI, USA
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
- Department of Surgery, University of California San Francisco, San Francisco, CA, US
- Gladstone Institute of Genomic Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Esther Latres
- Research Department, Breakthrough T1D, New York, NY, USA
| | | | - Jaime Giraldo
- Research Department, Breakthrough T1D, New York, NY, USA.
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
3
|
Kim BJ, Shin JS, Min BH, Kim JM, Park CG, Kang HJ, Hwang ES, Lee WW, Kim JS, Kim HJ, Kwon I, Kim JS, Kim GS, Moon J, Shin DY, Cho B, Yang HM, Kim SJ, Kim KW. Clinical Trial Protocol for Porcine Islet Xenotransplantation in South Korea. Diabetes Metab J 2024; 48:1160-1168. [PMID: 38772544 PMCID: PMC11621658 DOI: 10.4093/dmj.2023.0260] [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: 08/07/2023] [Accepted: 01/17/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGRUOUND Islet transplantation holds promise for treating selected type 1 diabetes mellitus patients, yet the scarcity of human donor organs impedes widespread adoption. Porcine islets, deemed a viable alternative, recently demonstrated successful longterm survival without zoonotic risks in a clinically relevant pig-to-non-human primate islet transplantation model. This success prompted the development of a clinical trial protocol for porcine islet xenotransplantation in humans. METHODS A single-center, open-label clinical trial initiated by the sponsor will assess the safety and efficacy of porcine islet transplantation for diabetes patients at Gachon Hospital. The protocol received approval from the Gachon Hospital Institutional Review Board (IRB) and the Korean Ministry of Food and Drug Safety (MFDS) under the Investigational New Drug (IND) process. Two diabetic patients, experiencing inadequate glycemic control despite intensive insulin treatment and frequent hypoglycemic unawareness, will be enrolled. Participants and their family members will engage in deliberation before xenotransplantation during the screening period. Each patient will receive islets isolated from designated pathogen-free pigs. Immunosuppressants and systemic infection prophylaxis will follow the program schedule. The primary endpoint is to confirm the safety of porcine islets in patients, and the secondary endpoint is to assess whether porcine islets can reduce insulin dose and the frequency of hypoglycemic unawareness. CONCLUSION A clinical trial protocol adhering to global consensus guidelines for porcine islet xenotransplantation is presented, facilitating streamlined implementation of comparable human trials worldwide.
Collapse
Affiliation(s)
- Byung-Joon Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | | | | | - Jong-Min Kim
- Department of Animal Health, Cheongju University College of Health and Medical Sciences, Cheongju, Korea
| | - Chung-Gyu Park
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University, Seoul, Korea
| | - Hee-Jung Kang
- Department of Laboratory Medicine, College of Medicine, Hallym University, Anyang, Korea
| | - Eung Soo Hwang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University, Seoul, Korea
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University, Seoul, Korea
| | - Jung-Sik Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University, Seoul, Korea
| | - Hyun Je Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University, Seoul, Korea
| | - Iov Kwon
- Department of Medical Education, Ewha Womans University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | - Kwang-Won Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| |
Collapse
|
4
|
Vasilchikova E, Ermakova P, Bogomolova A, Kashirina A, Lugovaya L, Tselousova J, Naraliev N, Kuchin D, Zagaynova E, Zagainov V, Kashina A. A Fresh Look at Islet Isolation from Rabbit Pancreases. Int J Mol Sci 2024; 25:10669. [PMID: 39408998 PMCID: PMC11477383 DOI: 10.3390/ijms251910669] [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: 08/27/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Islet transplantation represents a promising therapeutic approach for diabetes management, yet the isolation and evaluation of pancreatic islets remain challenging. This study focuses on the isolation of islets from rabbit pancreases, followed by a comprehensive assessment of their viability and functionality. We developed a novel method for isolating islet cells from the pancreas of adult rabbits. We successfully isolated viable islets, which were subsequently evaluated through a combination of viability assays, an insulin enzyme-linked immunosorbent assay (ELISA), and fluorescence lifetime imaging microscopy (FLIM). The viability assays indicated a high percentage of intact islets post-isolation, while the insulin ELISA demonstrated robust insulin secretion in response to glucose stimulation. FLIM provided insights into the metabolic state of the islets, revealing distinct fluorescence lifetime signatures correlating with functional viability. Our findings underscore the potential of rabbit islets as a model for studying islet biology and diabetes therapy, highlighting the efficacy of combining traditional assays with advanced imaging techniques for comprehensive functional assessments. This research contributes to the optimization of islet isolation protocols and enhances our understanding of islet functional activity dynamics in preclinical settings.
Collapse
Affiliation(s)
- Ekaterina Vasilchikova
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
- Federal State Educational Institution of Higher Educational Institution “National Research Nizhny Novgorod State University Named after N.I. Lobachevsky”, Nizhny Novgorod 603105, Russia
| | - Polina Ermakova
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
| | - Alexandra Bogomolova
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
| | - Alena Kashirina
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
| | - Liya Lugovaya
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
| | - Julia Tselousova
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
| | - Nasip Naraliev
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
- State Budgetary Healthcare Institution “Nizhny Novgorod Regional Clinical Oncology Dispensary”, Nizhny Novgorod 603126, Russia
| | - Denis Kuchin
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
- Nizhny Novgorod Regional Clinical Hospital Named after N.A. Semashko, Nizhny Novgorod 603005, Russia
| | - Elena Zagaynova
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
- Federal Scientific and Clinical Center for Physico-Chemical Medicine Named after Academician Yu. M. Lopukhin, Moscow 119334, Russia
| | - Vladimir Zagainov
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
- State Budgetary Healthcare Institution “Nizhny Novgorod Regional Clinical Oncology Dispensary”, Nizhny Novgorod 603126, Russia
| | - Alexandra Kashina
- Federal State Budgetary Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of Russia, Nizhny Novgorod 603082, Russia; (P.E.); (A.B.); (A.K.); (L.L.); (J.T.); (N.N.); (D.K.); (E.Z.); (V.Z.); (A.K.)
- Federal Scientific and Clinical Center for Physico-Chemical Medicine Named after Academician Yu. M. Lopukhin, Moscow 119334, Russia
| |
Collapse
|
5
|
Pilz J, Gloddek N, Lindheimer F, Lindner MJ, Puhr-Westerheide D, Ümütlü M, Cyran C, Seidensticker M, Lindner R, Kraetzl M, Renner S, Merkus D, Teupser D, Bartenstein P, Ziegler SI, Wolf E, Kemter E. Functional maturation and longitudinal imaging of intraportal neonatal porcine islet grafts in genetically diabetic pigs. Am J Transplant 2024; 24:1395-1405. [PMID: 38432328 DOI: 10.1016/j.ajt.2024.02.026] [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: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Allogeneic intraportal islet transplantation (ITx) has become an established treatment for patients with poorly controlled type 1 diabetes. However, the loss of viable beta-cell mass after transplantation remains a major challenge. Therefore, noninvasive imaging methods for long-term monitoring of the transplant fate are required. In this study, [68Ga]Ga-DOTA-exendin-4 positron emission tomography/computed tomography (PET/CT) was used for repeated monitoring of allogeneic neonatal porcine islets (NPI) after intraportal transplantation into immunosuppressed genetically diabetic pigs. NPI transplantation (3320-15,000 islet equivalents per kg body weight) led to a reduced need for exogenous insulin therapy and finally normalization of blood glucose levels in 3 out of 4 animals after 5 to 10 weeks. Longitudinal PET/CT measurements revealed a significant increase in standard uptake values in graft-bearing livers. Histologic analysis confirmed the presence of well-engrafted, mature islet clusters in the transplanted livers. Our study presents a novel large animal model for allogeneic intraportal ITx. A relatively small dose of NPIs was sufficient to normalize blood glucose levels in a clinically relevant diabetic pig model. [68Ga]Ga-DOTA-exendin-4 PET/CT proved to be efficacious for longitudinal monitoring of islet transplants. Thus, it could play a crucial role in optimizing ITx as a curative therapy for type 1 diabetes.
Collapse
Affiliation(s)
- Johanna Pilz
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nicol Gloddek
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Felix Lindheimer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Magdalena J Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | | | - Muzzafer Ümütlü
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Clemens Cyran
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Richard Lindner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Kraetzl
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Daphne Merkus
- Walter Brendel Center for Experimental Medicine (WBex), University Hospital, LMU Munich, Munich, Germany
| | - Daniel Teupser
- Department of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Sibylle I Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
6
|
Raoufinia R, Rahimi HR, Saburi E, Moghbeli M. Advances and challenges of the cell-based therapies among diabetic patients. J Transl Med 2024; 22:435. [PMID: 38720379 PMCID: PMC11077715 DOI: 10.1186/s12967-024-05226-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
Diabetes mellitus is a significant global public health challenge, with a rising prevalence and associated morbidity and mortality. Cell therapy has evolved over time and holds great potential in diabetes treatment. In the present review, we discussed the recent progresses in cell-based therapies for diabetes that provides an overview of islet and stem cell transplantation technologies used in clinical settings, highlighting their strengths and limitations. We also discussed immunomodulatory strategies employed in cell therapies. Therefore, this review highlights key progresses that pave the way to design transformative treatments to improve the life quality among diabetic patients.
Collapse
Affiliation(s)
- Ramin Raoufinia
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Ho BX, Teo AKK, Ng NHJ. Innovations in bio-engineering and cell-based approaches to address immunological challenges in islet transplantation. Front Immunol 2024; 15:1375177. [PMID: 38650946 PMCID: PMC11033429 DOI: 10.3389/fimmu.2024.1375177] [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: 01/23/2024] [Accepted: 03/11/2024] [Indexed: 04/25/2024] Open
Abstract
Human allogeneic pancreatic islet transplantation is a life-changing treatment for patients with severe Type 1 Diabetes (T1D) who suffer from hypoglycemia unawareness and high risk of severe hypoglycemia. However, intensive immunosuppression is required to prevent immune rejection of the graft, that may in turn lead to undesirable side effects such as toxicity to the islet cells, kidney toxicity, occurrence of opportunistic infections, and malignancies. The shortage of cadaveric human islet donors further limits islet transplantation as a treatment option for widespread adoption. Alternatively, porcine islets have been considered as another source of insulin-secreting cells for transplantation in T1D patients, though xeno-transplants raise concerns over the risk of endogenous retrovirus transmission and immunological incompatibility. As a result, technological advancements have been made to protect transplanted islets from immune rejection and inflammation, ideally in the absence of chronic immunosuppression, to improve the outcomes and accessibility of allogeneic islet cell replacement therapies. These include the use of microencapsulation or macroencapsulation devices designed to provide an immunoprotective environment using a cell-impermeable layer, preventing immune cell attack of the transplanted cells. Other up and coming advancements are based on the use of stem cells as the starting source material for generating islet cells 'on-demand'. These starting stem cell sources include human induced pluripotent stem cells (hiPSCs) that have been genetically engineered to avoid the host immune response, curated HLA-selected donor hiPSCs that can be matched with recipients within a given population, and multipotent stem cells with natural immune privilege properties. These strategies are developed to provide an immune-evasive cell resource for allogeneic cell therapy. This review will summarize the immunological challenges facing islet transplantation and highlight recent bio-engineering and cell-based approaches aimed at avoiding immune rejection, to improve the accessibility of islet cell therapy and enhance treatment outcomes. Better understanding of the different approaches and their limitations can guide future research endeavors towards developing more comprehensive and targeted strategies for creating a more tolerogenic microenvironment, and improve the effectiveness and sustainability of islet transplantation to benefit more patients.
Collapse
Affiliation(s)
- Beatrice Xuan Ho
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- BetaLife Pte Ltd, Singapore, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Natasha Hui Jin Ng
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
8
|
Kabakchieva P, Assyov Y, Gerasoudis S, Vasilev G, Peshevska-Sekulovska M, Sekulovski M, Lazova S, Miteva DG, Gulinac M, Tomov L, Velikova T. Islet transplantation-immunological challenges and current perspectives. World J Transplant 2023; 13:107-121. [PMID: 37388389 PMCID: PMC10303418 DOI: 10.5500/wjt.v13.i4.107] [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: 03/22/2023] [Revised: 05/16/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023] Open
Abstract
Pancreatic islet transplantation is a minimally invasive procedure aiming to reverse the effects of insulin deficiency in patients with type 1 diabetes (T1D) by transplanting pancreatic beta cells. Overall, pancreatic islet transplantation has improved to a great extent, and cellular replacement will likely become the mainstay treatment. We review pancreatic islet transplantation as a treatment for T1D and the immunological challenges faced. Published data demonstrated that the time for islet cell transfusion varied between 2 and 10 h. Approximately 54% of the patients gained insulin independence at the end of the first year, while only 20% remained insulin-free at the end of the second year. Eventually, most transplanted patients return to using some form of exogenous insulin within a few years after the transplantation, which imposed the need to improve immunological factors before transplantation. We also discuss the immunosuppressive regimens, apoptotic donor lymphocytes, anti-TIM-1 antibodies, mixed chimerism-based tolerance induction, induction of antigen-specific tolerance utilizing ethylene carbodiimide-fixed splenocytes, pretransplant infusions of donor apoptotic cells, B cell depletion, preconditioning of isolated islets, inducing local immunotolerance, cell encapsulation and immunoisolation, using of biomaterials, immunomodulatory cells, etc.
Collapse
Affiliation(s)
- Plamena Kabakchieva
- Clinic of Internal Diseases, Naval Hospital-Varna, Military Medical Academy, Varna 9010, Bulgaria
| | - Yavor Assyov
- Clinic of Endocrinology, Department of Internal Diseases, University Hospital "Alexandrovska", Medical University-Sofia, Sofia 1434, Bulgaria
| | | | - Georgi Vasilev
- Department of Neurology, Faculty of Medicine, Medical University of Plovdiv, Plovdiv 4000, Bulgaria
| | - Monika Peshevska-Sekulovska
- Department of Gastroenterology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Metodija Sekulovski
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
- Department of Anesthesiology and Intensive Care, University hospital Lozenetz, Sofia 1407, Bulgaria
| | - Snezhina Lazova
- Department of Pediatric, University Hospital "N. I. Pirogov", Sofia 1606, Bulgaria
- Department of Healthcare, Faculty of Public Health "Prof. Tsekomir Vodenicharov, MD, DSc", Medical University of Sofia, Sofia 1527, Bulgaria
| | | | - Milena Gulinac
- Department of General and Clinical Pathology, Medical University of Plovdiv, Plovdiv 4000, Bulgaria
| | - Latchezar Tomov
- Department of Informatics, New Bulgarian University, Sofia 1618, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
9
|
Ashe S, Hebrok M. Role of Cell-Based Therapies in T2D. Semin Nephrol 2023; 43:151432. [PMID: 37918206 DOI: 10.1016/j.semnephrol.2023.151432] [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] [Indexed: 11/04/2023]
Abstract
Type 2 diabetes mellitus (T2D) has become a global epidemic affecting the health of millions of people. T2D is a complex and multifactorial metabolic disease, largely characterized by a combination of impaired insulin secretion from β cells residing within the islets of the pancreas and peripheral insulin resistance. In this article, we discuss the current state and risk factors for T2D, conventional treatment options, and upcoming strategies, including progress in the areas of allogeneic and xenogeneic islet transplantation, with a major focus on stem cell-derived β cells and associated technologies.
Collapse
Affiliation(s)
- Sudipta Ashe
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA; TUM School of Medicine, Technical University Munich, Munich, Germany; Center for Organoid Systems, Technical University Munich, Garching, Germany; Institute for Diabetes and Organoid Technology, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; Munich Institute of Biomedical Engineering (MIBE), Technical University Munich, Munich, Germany; German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
10
|
Engineering Strategies of Islet Product for Endocrine Regeneration. ENGINEERED REGENERATION 2023. [DOI: 10.1016/j.engreg.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
11
|
Denner J. Xenotransplantation of pig islet cells: Potential adverse impact of virus infections on their functionality and insulin production. Xenotransplantation 2022; 30:e12789. [PMID: 36495163 DOI: 10.1111/xen.12789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/09/2022] [Accepted: 10/26/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Joachim Denner
- Institute of Virology Free University Berlin Berlin Germany
| |
Collapse
|
12
|
Tritschler S, Thomas M, Böttcher A, Ludwig B, Schmid J, Schubert U, Kemter E, Wolf E, Lickert H, Theis FJ. A transcriptional cross species map of pancreatic islet cells. Mol Metab 2022; 66:101595. [PMID: 36113773 PMCID: PMC9526148 DOI: 10.1016/j.molmet.2022.101595] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/20/2022] [Accepted: 09/03/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Pancreatic islets of Langerhans secrete hormones to regulate systemic glucose levels. Emerging evidence suggests that islet cells are functionally heterogeneous to allow a fine-tuned and efficient endocrine response to physiological changes. A precise description of the molecular basis of this heterogeneity, in particular linking animal models to human islets, is an important step towards identifying the factors critical for endocrine cell function in physiological and pathophysiological conditions. METHODS In this study, we used single-cell RNA sequencing to profile more than 50'000 endocrine cells isolated from healthy human, pig and mouse pancreatic islets and characterize transcriptional heterogeneity and evolutionary conservation of those cells across the three species. We systematically delineated endocrine cell types and α- and β-cell heterogeneity through prior knowledge- and data-driven gene sets shared across species, which altogether capture common and differential cellular properties, transcriptional dynamics and putative driving factors of state transitions. RESULTS We showed that global endocrine expression profiles correlate, and that critical identity and functional markers are shared between species, while only approximately 20% of cell type enriched expression is conserved. We resolved distinct human α- and β-cell states that form continuous transcriptional landscapes. These states differentially activate maturation and hormone secretion programs, which are related to regulatory hormone receptor expression, signaling pathways and different types of cellular stress responses. Finally, we mapped mouse and pig cells to the human reference and observed that the spectrum of human α- and β-cell heterogeneity and aspects of such functional gene expression are better recapitulated in the pig than mouse data. CONCLUSIONS Here, we provide a high-resolution transcriptional map of healthy human islet cells and their murine and porcine counterparts, which is easily queryable via an online interface. This comprehensive resource informs future efforts that focus on pancreatic endocrine function, failure and regeneration, and enables to assess molecular conservation in islet biology across species for translational purposes.
Collapse
Affiliation(s)
- Sophie Tritschler
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Technical University of Munich, School of Life Sciences Weihenstephan, 85354 Freising, Germany
| | - Moritz Thomas
- Technical University of Munich, School of Life Sciences Weihenstephan, 85354 Freising, Germany; Institute of AI for Health, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Barbara Ludwig
- Department of Medicine III, University Hospital Carl Gustav Carus, Technical University of Dresden, 01307 Dresden, Germany; Paul Langerhans Institute Dresden of Helmholtz Zentrum München, University Hospital Carl Gustav Carus, Technical University of Dresden, 01307 Dresden, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Janine Schmid
- Department of Medicine III, University Hospital Carl Gustav Carus, Technical University of Dresden, 01307 Dresden, Germany
| | - Undine Schubert
- Department of Medicine III, University Hospital Carl Gustav Carus, Technical University of Dresden, 01307 Dresden, Germany
| | - Elisabeth Kemter
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany
| | - Eckhard Wolf
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Chair for Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Technical University of Munich, Medical Faculty, 81675 Munich, Germany.
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Garching b. Munich, Germany.
| |
Collapse
|
13
|
Shahab M, Din NU, Shahab N. Genetically Engineered Porcine Organs for Human Xenotransplantation. Cureus 2022; 14:e29089. [PMID: 36249604 PMCID: PMC9556182 DOI: 10.7759/cureus.29089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 12/03/2022] Open
Abstract
Xenotransplantation holds a promising future for many patients, especially those with end-stage renal disease or uncontrollable serum glucose levels. Porcine organs are viewed as the perfect candidate for a source of xenografts. However, the recipient's immunity, incompatibility of biologic systems, and transfer of new pathogenic organisms are all obstacles to clinical xenotransplantation, in addition to the risk of zoonosis and xenoantigens. Genetic modification of pigs using clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated protein 9 (Cas9) resulted in the production of porcine endogenous retrovirus (PERV)-free offsprings with the consequent removal of many clinical complications post-transplantation. Such as minimizing both acute and chronic inflammation, in addition to suppressing rejection reactions, which may prolong graft survival. To build on these recent successes, it is important to look at the limits of genetic engineering and develop ways to advance the field of xenotransplantation and reverse xenotransplantation clinical applications forward. Still, significant problems remain with clinical human xenotransplantation; future work should focus on developing an ideal genetically engineered swine donor source that can improve long-term graft survival and suppress the immune system in a clinically useful way.
Collapse
|
14
|
Sordi V, Monaco L, Piemonti L. Cell Therapy for Type 1 Diabetes: From Islet Transplantation to Stem Cells. Horm Res Paediatr 2022; 96:658-669. [PMID: 36041412 DOI: 10.1159/000526618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 08/08/2022] [Indexed: 11/19/2022] Open
Abstract
The field of cell therapy of type 1 diabetes is a particularly interesting example in the scenario of regenerative medicine. In fact, β-cell replacement has its roots in the experience of islet transplantation, which began 40 years ago and is currently a rapidly accelerating field, with several ongoing clinical trials using β cells derived from stem cells. Type 1 diabetes is particularly suitable for cell therapy as it is a disease due to the deficiency of only one cell type, the insulin-producing β cell, and this endocrine cell does not need to be positioned inside the pancreas to perform its function. On the other hand, the presence of a double immunological barrier, the allogeneic one and the autoimmune one, makes the protection of β cells from rejection a major challenge. Until today, islet transplantation has taught us a lot, pioneering immunosuppressive therapies, graft encapsulation, tissue engineering, and test of different implant sites and has stimulated a great variety of studies on β-cell function. This review starts from islet transplantation, presenting its current indications and the latest published trials, to arrive at the prospects of stem cell therapy, presenting the latest innovations in the field.
Collapse
Affiliation(s)
- Valeria Sordi
- Diabetes Research Institute, San Raffaele Hospital, Milan, Italy,
| | - Laura Monaco
- Diabetes Research Institute, San Raffaele Hospital, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
15
|
Birdwhistell KE, Hurley DJ, Heins B, Peroni JF. Evaluation of equine xenogeneic mixed lymphocyte reactions using 5-ethynyl-2'-deoxyuridine (EdU). Vet Immunol Immunopathol 2022; 249:110430. [PMID: 35525064 DOI: 10.1016/j.vetimm.2022.110430] [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: 09/30/2021] [Revised: 04/29/2022] [Accepted: 05/01/2022] [Indexed: 11/19/2022]
Abstract
Allogeneic solid organ transplantation is currently the only treatment option for end stage organ disease. The shortage of available donor organs has driven efforts to utilize xenogeneic organs for transplantation. In vitro methods for evaluating immune-compatibility are a quick and low cost means of screening novel tissue products prior to more involved, expensive, and invasive live animal studies. Recently, a new analog of the DNA base thymidine, 5-ethynyl-2'-deoxyuridine (EdU), was developed. It may be used in a fast, efficient and specific means of evaluating cell proliferation via flow cytometry. This study was designed to test and optimize this platform for assessing equine xenogeneic one-way mixed lymphocyte reaction (MLR) to porcine stimulator cells. Furthermore, it was hypothesized that an enriched T-lymphocyte (T-cell) population would generate a stronger proliferative response to stimulation, and higher levels of cytokine production when compared to unfractionated peripheral blood mononuclear cells (PBMCs). PBMCs and T-cells were isolated from 3 horses and 4 pigs. Equine xenogeneic MLRs were set up using porcine allogeneic MLRs as a reference for clinically acceptable levels of cell proliferation. Equine T-cells showed significantly greater EdU incorporation in one-way xenogeneic MLRs than equine PBMCs. However, there was no significant difference in cell proliferation between porcine T-cell and PBMC as responders in allogenic one-way MLRs. Given the results of this study, we consider that enriched equine T-cells should be used in preference to unfractionated PBMCs when attempting to evaluate the equine xenogeneic response using the EdU assay as an indicator of suitability for transplant in vivo.
Collapse
Affiliation(s)
- Kate E Birdwhistell
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, 2200 College Station Rd, Athens, GA 30602, USA.
| | - David J Hurley
- Department of Population Health, University of Georgia College of Veterinary Medicine, 2200 College Station Rd, Athens, GA 30602, USA
| | - Bradley Heins
- Department of Population Health, University of Georgia College of Veterinary Medicine, 2200 College Station Rd, Athens, GA 30602, USA
| | - John F Peroni
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, 2200 College Station Rd, Athens, GA 30602, USA
| |
Collapse
|
16
|
Scheibner K, Lickert H. Awaking sleeping islets for a cure of diabetes. MED 2022; 3:279-280. [DOI: 10.1016/j.medj.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Human pluripotent stem-cell-derived islets ameliorate diabetes in non-human primates. Nat Med 2022; 28:272-282. [PMID: 35115708 DOI: 10.1038/s41591-021-01645-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem-cell-derived islets (hPSC-islets) are a promising cell resource for diabetes treatment1,2. However, this therapeutic strategy has not been systematically assessed in large animal models physiologically similar to humans, such as non-human primates3. In this study, we generated islets from human chemically induced pluripotent stem cells (hCiPSC-islets) and show that a one-dose intraportal infusion of hCiPSC-islets into diabetic non-human primates effectively restored endogenous insulin secretion and improved glycemic control. Fasting and average pre-prandial blood glucose levels significantly decreased in all recipients, accompanied by meal or glucose-responsive C-peptide release and overall increase in body weight. Notably, in the four long-term follow-up macaques, average hemoglobin A1c dropped by over 2% compared with peak values, whereas the average exogenous insulin requirement reduced by 49% 15 weeks after transplantation. Collectively, our findings show the feasibility of hPSC-islets for diabetic treatment in a preclinical context, marking a substantial step forward in clinical translation of hPSC-islets.
Collapse
|
18
|
Walker S, Appari M, Forbes S. Considerations and challenges of islet transplantation and future therapies on the horizon. Am J Physiol Endocrinol Metab 2022; 322:E109-E117. [PMID: 34927459 DOI: 10.1152/ajpendo.00310.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Islet transplantation is a treatment for selected adults with type 1 diabetes and severe hypoglycemia. Islets from two or more donor pancreases, a scarce resource, are usually required to impact glycemic control, but the treatment falls short of a cure. Islets are avascular when transplanted into the hypoxic liver environment and subjected to inflammatory insults, immune attack, and toxicity from systemic immunosuppression. The Collaborative Islet Transplant Registry, with outcome data on over 1,000 islet transplant recipients, has demonstrated that larger islet numbers transplanted and older age of recipients are associated with better outcomes. Induction with T-cell depleting agents and the TNF-α inhibitor etanercept and maintenance systemic immunosuppression with mTOR inhibitors in combination with calcineurin inhibitors also appear advantageous, but concerns remain over immunosuppressive toxicity. We discuss strategies and therapeutics that address specific challenges of islet transplantation, many of which are at the preclinical stage of development. On the horizon are adjuvant cell therapies with mesenchymal stromal cells and regulatory T cells that have been used in preclinical models and in humans in other contexts; such a strategy may enable reductions in immunosuppression in the early peri-transplant period when the islets are vulnerable to apoptosis. Human embryonic stem cell-derived islets are in early-phase clinical trials and hold the promise of an inexhaustible supply of insulin-producing cells; effective encapsulation of such cells or, silencing of the human leukocyte antigen (HLA) complex would eliminate the need for immunosuppression, enabling this therapy to be used in all those with type 1 diabetes.
Collapse
Affiliation(s)
- Sophie Walker
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Mahesh Appari
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Shareen Forbes
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Transplant Unit, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
- Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
19
|
Wan XX, Zhang DY, Khan MA, Zheng SY, Hu XM, Zhang Q, Yang RH, Xiong K. Stem Cell Transplantation in the Treatment of Type 1 Diabetes Mellitus: From Insulin Replacement to Beta-Cell Replacement. Front Endocrinol (Lausanne) 2022; 13:859638. [PMID: 35370989 PMCID: PMC8972968 DOI: 10.3389/fendo.2022.859638] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/16/2022] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease that attacks pancreatic β-cells, leading to the destruction of insulitis-related islet β-cells. Islet β-cell transplantation has been proven as a curative measure in T1DM. However, a logarithmic increase in the global population with diabetes, limited donor supply, and the need for lifelong immunosuppression restrict the widespread use of β-cell transplantation. Numerous therapeutic approaches have been taken to search for substitutes of β-cells, among which stem cell transplantation is one of the most promising alternatives. Stem cells have demonstrated the potential efficacy to treat T1DM by reconstitution of immunotolerance and preservation of islet β-cell function in recent research. cGMP-grade stem cell products have been used in human clinical trials, showing that stem cell transplantation has beneficial effects on T1DM, with no obvious adverse reactions. To better achieve remission of T1DM by stem cell transplantation, in this work, we explain the progression of stem cell transplantation such as mesenchymal stem cells (MSCs), human embryonic stem cells (hESCs), and bone marrow hematopoietic stem cells (BM-HSCs) to restore the immunotolerance and preserve the islet β-cell function of T1DM in recent years. This review article provides evidence of the clinical applications of stem cell therapy in the treatment of T1DM.
Collapse
Affiliation(s)
- Xin-Xing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Dan-Yi Zhang
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Md. Asaduzzaman Khan
- The Research Centre for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Sheng-Yuan Zheng
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Xi-Min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Rong-Hua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Kun Xiong, ; Rong-Hua Yang,
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China
- *Correspondence: Kun Xiong, ; Rong-Hua Yang,
| |
Collapse
|
20
|
Huang CP, Liu LC, Chang CC, Wu CC, Shyr CR. Intratumoral xenogeneic tissue-specific cell immunotherapy inhibits tumor growth by increasing antitumor immunity in murine triple negative breast and pancreatic tumor models. Cancer Lett 2021; 545:115478. [PMID: 35902043 DOI: 10.1016/j.canlet.2021.10.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/18/2021] [Accepted: 10/31/2021] [Indexed: 12/13/2022]
Abstract
Low immunogenicity in tumors and the immunosuppressive tumor microenvironment (TME) represent major obstacles to the full success of immunotherapy in cancer patients. A novel intratumoral xenogeneic tissue-specific cell immunotherapeutic approach could overcome the obstacles. Murine 4T1 triple negative breast cancer (TNBC) cells and Pan18 pancreatic ductal adenocarcinoma (PDAC) cells were used for establishing syngeneic graft tumor models to evaluate antitumor effect of intratumoral injection of xenogeneic tissue-specific cells. Responses to treatment were assessed by measuring tumor growth and tumor weight of the tumor-bearing mice. To investigate the mechanisms of action, tumor histology and immunohistochemistry and cytokine gene expression were measured. Splenic lymphocytes proliferation, cytokine production and cytotoxicity activities were also assessed. The findings showed that intratumoral injection of xenogeneic tissue-specific cells in monotherapy and combination with chemotherapy inhibit tumor growth. The therapeutic efficacy of intratumoral xenogeneic cells was significantly enhanced by the addition of cytotoxic chemotherapeutic agents. Mice that received combined treatment showed maximal attenuation in tumor growth rate. The antitumor immunity was explained by altered immune cell infiltration in tumors and immune cell functions. Our findings demonstrate that xenogeneic tissue-specific cells given intratumorally, provide a potent antitumor effect in murine breast and pancreatic tumor models by enhancing recruitment and activation of immune cells in tumors for local and systemic antitumor effects. Moreover, intratumoral xenogeneic cell treatment turns immunologically "cold" tumors to "hot" ones, generates systemic antitumor immunity, and synergizes with chemotherapy. Thus, the intratumoral xenogeneic tissue-specific cell immunotherapy may represent a useful therapeutic option to difficult-to-treat cancers.
Collapse
Affiliation(s)
- Chi-Ping Huang
- Department of Medicine, Department of Urology, College of Medicine, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Liang-Chih Liu
- Department of Medicine, Department of Surgery, College of Medicine, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Chih-Chun Chang
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Chun-Chie Wu
- Department of Medicine, Department of Urology, College of Medicine, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
21
|
Denner J. Porcine Endogenous Retroviruses and Xenotransplantation, 2021. Viruses 2021; 13:v13112156. [PMID: 34834962 PMCID: PMC8625113 DOI: 10.3390/v13112156] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/20/2021] [Indexed: 12/25/2022] Open
Abstract
Porcine endogenous retroviruses (PERVs) are integrated in the genome of all pigs, and some of them are able to infect human cells. Therefore, PERVs pose a risk for xenotransplantation, the transplantation of pig cells, tissues, or organ to humans in order to alleviate the shortage of human donor organs. Up to 2021, a huge body of knowledge about PERVs has been accumulated regarding their biology, including replication, recombination, origin, host range, and immunosuppressive properties. Until now, no PERV transmission has been observed in clinical trials transplanting pig islet cells into diabetic humans, in preclinical trials transplanting pig cells and organs into nonhuman primates with remarkable long survival times of the transplant, and in infection experiments with several animal species. Nevertheless, in order to prevent virus transmission to the recipient, numerous strategies have been developed, including selection of PERV-C-free animals, RNA interference, antiviral drugs, vaccination, and genome editing. Furthermore, at present there are no more experimental approaches to evaluate the full risk until we move to the clinic.
Collapse
Affiliation(s)
- Joachim Denner
- Department of Veterinary Medicine, Institute of Virology, Free University Berlin, 14163 Berlin, Germany
| |
Collapse
|
22
|
Abstract
Technological advancements in blood glucose monitoring and therapeutic insulin administration have improved the quality of life for people with type 1 diabetes. However, these efforts fall short of replicating the exquisite metabolic control provided by native islets. We examine the integrated advancements in islet cell replacement and immunomodulatory therapies that are coalescing to enable the restoration of endogenous glucose regulation. We highlight advances in stem cell biology and graft site design, which offer innovative sources of cellular material and improved engraftment. We also cover cutting-edge approaches for preventing allograft rejection and recurrent autoimmunity. These insights reflect a growing understanding of type 1 diabetes etiology, β cell biology, and biomaterial design, together highlighting therapeutic opportunities to durably replace the β cells destroyed in type 1 diabetes.
Collapse
Affiliation(s)
- Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, and Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- University of Florida Diabetes Institute, University of Florida, Gainesville, FL 32610, USA
| | - Holger A Russ
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cherie L Stabler
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
23
|
Kwak K, Park JK, Shim J, Ko N, Kim HJ, Lee Y, Kim JH, Alexander M, Lakey JRT, Kim H, Choi K. Comparison of islet isolation result and clinical applicability according to GMP-grade collagenase enzyme blend in adult porcine islet isolation and culture. Xenotransplantation 2021; 28:e12703. [PMID: 34176167 PMCID: PMC8459292 DOI: 10.1111/xen.12703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022]
Abstract
Background Porcine islet xenotransplantation is a promising treatment for type 1 diabetes as an alternative to human pancreatic islet transplantation and long‐term insulin therapy. Several research groups have explored porcine islets as an alternative to the inconsistent and chronic shortage of pancreases from human organ donors. Studies have confirmed successful transplant of porcine islets into non‐human primate models of diabetes; however, in most cases, they require more than one adult porcine donor to achieve sufficient viable islet mass for sustained function. The importance of GMP‐grade reagents includes the following: specific enzymes utilized in the pancreatic isolation process were identified as a key factor in successful human clinical islet transplantation trials using cadaveric islets. As xenotransplantation clinical research progresses, isolation reagents and digestion enzymes play a key role in the consistency of the product and ultimately the outcome of the islet xenotransplant. In this study, we evaluated several commercially available enzyme blends that have been used for islet isolation. We evaluated their impact on islet isolation yield and subsequent islet function as part of our plan to bring xenotransplantation into clinical xenotransplantation trials. Methods Adult porcine islets were isolated from 16 to 17‐month‐old Yucatan miniature pigs following standard rapid procurement. Pigs weighed on average 48.71 ± 2.85 kg, and the produced pancreases were 39.51 ± 1.80 grams (mean ± SEM). After ductal cannulation, we evaluated both GMP‐grade enzymes (Collagenase AF‐1 GMP grade and Liberase MTF C/T GMP grade) and compared with standard non‐GMP enzyme blend (Collagenase P). Islet quality control assessments including islet yield, islet size (IEQ), membrane integrity (acridine orange/propidium iodide), and functional viability (GSIS) were evaluated in triplicate on day 1 post‐islet isolation culture. Results Islet yield was highest in the group of adult pigs where Collagenase AF‐1 GMP grade was utilized. The mean islet yield was 16 586 ± 1391 IEQ/g vs 8302 ± 986 IEQ/g from pancreases isolated using unpurified crude Collagenase P. The mean islet size was higher in Collagenase AF‐1 GMP grade with neutral protease than in Collagenase P and Liberase MTF C/T GMP grade. We observed no significant difference between the experimental groups, but in vitro islet function after overnight tissue culture was significantly higher in Collagenase AF‐1 GMP grade with neutral protease and Liberase MTF C/T GMP grade than the crude control enzyme group. As expected, the GMP‐grade enzyme has significantly lower endotoxin levels than the crude control enzyme group when measured. Conclusions This study validates the importance of using specifically blended GMP grade for adult pig islet isolation for xenotransplantation trials and the ability to isolate a sufficient number of viable islets from one adult pig to provide a sufficient number for islets for a clinical islet transplantation. GMP‐grade enzymes are highly efficient in increasing islet yield, size, viability, and function at a lower and acceptable endotoxin level. Ongoing research transplants these islets into animal models of diabetes to validate in vivo function. Also, these defined and reproducible techniques using GMP‐grade enzymes allow for continuance of our plan to advance to xenotransplantation of isolated pig islets for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michael Alexander
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jonathan R T Lakey
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.,Department of Surgery, University of California Irvine, Orange, CA, USA
| | | | | |
Collapse
|
24
|
Boscari F, Avogaro A. Current treatment options and challenges in patients with Type 1 diabetes: Pharmacological, technical advances and future perspectives. Rev Endocr Metab Disord 2021; 22:217-240. [PMID: 33755854 PMCID: PMC7985920 DOI: 10.1007/s11154-021-09635-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes mellitus imposes a significant burden of complications and mortality, despite important advances in treatment: subjects affected by this disease have also a worse quality of life-related to disease management. To overcome these challenges, different new approaches have been proposed, such as new insulin formulations or innovative devices. The introduction of insulin pumps allows a more physiological insulin administration with a reduction of HbA1c level and hypoglycemic risk. New continuous glucose monitoring systems with better accuracy have allowed, not only better glucose control, but also the improvement of the quality of life. Integration of these devices with control algorithms brought to the creation of the first artificial pancreas, able to independently gain metabolic control without the risk of hypo- and hyperglycemic crisis. This approach has revolutionized the management of diabetes both in terms of quality of life and glucose control. However, complete independence from exogenous insulin will be obtained only by biological approaches that foresee the replacement of functional beta cells obtained from stem cells: this will be a major challenge but the biggest hope for the subjects with type 1 diabetes. In this review, we will outline the current scenario of innovative diabetes management both from a technological and biological point of view, and we will also forecast some cutting-edge approaches to reduce the challenges that hamper the definitive cure of diabetes.
Collapse
Affiliation(s)
- Federico Boscari
- Department of Medicine, Unit of Metabolic Diseases, University of Padova, Padova, Italy.
| | - Angelo Avogaro
- Department of Medicine, Unit of Metabolic Diseases, University of Padova, Padova, Italy
| |
Collapse
|
25
|
Huang CP, Yang CY, Shyr CR. Utilizing Xenogeneic Cells As a Therapeutic Agent for Treating Diseases. Cell Transplant 2021; 30:9636897211011995. [PMID: 33975464 PMCID: PMC8120531 DOI: 10.1177/09636897211011995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022] Open
Abstract
The utilization of biologically produced cells to treat diseases is a revolutionary invention in modern medicine after chemically synthesized small molecule drugs and biochemically made protein drugs. Cells are basic units of life with diverse functions in mature and developing organs, which biological properties could be utilized as a promising therapeutic approach for currently intractable and incurable diseases. Xenogeneic cell therapy utilizing animal cells other than human for medicinal purpose has been studied as a new way of treating diseases. Xenogeneic cell therapy is considered as a potential regenerative approach to fulfill current unmet medical needs because xenogeneic cells could be isolated from different animal organs and expanded ex vivo as well as maintain the characteristics of original organs, providing a versatile and plenty cell source for cell-based therapeutics beside autologous and allogeneic sources. The swine species is considered the most suitable source because of the similarity with humans in size and physiology of many organs in addition to the economic and ethical reasons plus the possibility of genetic modification. This review discusses the old proposed uses of xenogeneic cells such as xenogeneic pancreatic islet cells, hepatocytes and neuronal cells as a living drug for the treatment of degenerative and organ failure diseases. Novel applications of xenogeneic mesenchymal stroma cells and urothelial cells are also discussed. There are formidable immunological barriers toward successful cellular xenotransplantation in clinic despite major progress in the development of novel immunosuppression regimens and genetically multimodified donor pigs. However, immunological barriers could be turn into immune boosters by using xenogeneic cells of specific tissue types as a novel immunotherapeutic agent to elicit bystander antitumor immunity due to rejection immune responses. Xenogeneic cells have the potential to become a safe and efficacious option for intractable diseases and hard-to-treat cancers, adding a new class of cellular medicine in our drug armamentarium.
Collapse
Affiliation(s)
- Chi-Ping Huang
- Department of Urology, School of Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Chi-Yu Yang
- Animal Technology Research Center/Division of Animal Technology, Agriculture Technology Research Institute, Miaoli, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, Taiwan
| |
Collapse
|