1
|
Ali A, Kurome M, Kessler B, Kemter E, Wolf E. What Genetic Modifications of Source Pigs Are Essential and Sufficient for Cell, Tissue, and Organ Xenotransplantation? Transpl Int 2024; 37:13681. [PMID: 39697899 PMCID: PMC11652200 DOI: 10.3389/ti.2024.13681] [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: 08/21/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
Xenotransplantation of porcine organs has made remarkable progress towards clinical application. A key factor has been the generation of genetically multi-modified source pigs for xenotransplants, protected against immune rejection and coagulation dysregulation. While efficient gene editing tools and multi-cistronic expression cassettes facilitate sophisticated and complex genetic modifications with multiple gene knockouts and protective transgenes, an increasing number of independently segregating genetic units complicates the breeding of the source pigs. Therefore, an optimal combination of essential genetic modifications may be preferable to extensive editing of the source pigs. Here, we discuss the prioritization of genetic modifications to achieve long-term survival and function of xenotransplants and summarise the genotypes that have been most successful for xenogeneic heart, kidney, and islet transplantation. Specific emphasis is given to the choice of the breed/genetic background of the source pigs. Moreover, multimodal deep phenotyping of porcine organs after xenotransplantation into human decedents will be discussed as a strategy for selecting essential genetic modifications of the source pigs. In addition to germ-line gene editing, some of these modifications may also be induced during organ preservation/perfusion, as demonstrated recently by the successful knockdown of swine leukocyte antigens in porcine lungs during ex vivo perfusion.
Collapse
Affiliation(s)
- Asghar Ali
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
| | - Mayuko Kurome
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
| | - Barbara Kessler
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
| | - Elisabeth Kemter
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
2
|
Piemonti L, Citro A, Tomajer V, Partelli S, Caldara R. Pig Xenotransplantation in Beta Cell Replacement: Addressing Challenges and Harnessing Potential for Type 1 Diabetes Therapy. Transpl Int 2024; 37:13122. [PMID: 39512630 PMCID: PMC11540633 DOI: 10.3389/ti.2024.13122] [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: 04/11/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024]
Abstract
This opinion paper evaluates the potential of porcine islets as a promising alternative in beta cell replacement therapy for Type 1 Diabetes (T1D), juxtaposed with the current limitations of human donor islets. It analyzes the compatibility of pig islets with human glucose metabolism, their prospects as a limitless and high-quality source of beta cells, and the unique immunogenic challenges they present in xenotransplantation. Additionally, the paper discusses the regulatory and ethical considerations pertinent to the use of porcine islets. By synthesizing current research and expert perspectives, the paper highlights both the opportunities and significant barriers that need addressing to advance pig islets as a viable therapeutic option. The findings advocate for a balanced and forward-looking approach to the integration of pig islets in T1D treatment, underscoring the need for continued research and dialogue in this evolving field.
Collapse
Affiliation(s)
- Lorenzo Piemonti
- Clinic Unit of Regenerative Medicine and Organ Transplants and Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
- Diabetes Research Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- Clinic Unit of Regenerative Medicine and Organ Transplants and Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Valentina Tomajer
- Pancreatic Surgery, Pancreas Translational and Clinical Research Center, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Stefano Partelli
- Diabetes Research Institute, Università Vita-Salute San Raffaele, Milan, Italy
- Pancreatic Surgery, Pancreas Translational and Clinical Research Center, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Rossana Caldara
- Clinic Unit of Regenerative Medicine and Organ Transplants and Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
3
|
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
|
4
|
Grimus S, Sarangova V, Welzel PB, Ludwig B, Seissler J, Kemter E, Wolf E, Ali A. Immunoprotection Strategies in β-Cell Replacement Therapy: A Closer Look at Porcine Islet Xenotransplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401385. [PMID: 38884159 PMCID: PMC11336975 DOI: 10.1002/advs.202401385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/28/2024] [Indexed: 06/18/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by absolute insulin deficiency primarily due to autoimmune destruction of pancreatic β-cells. The prevailing treatment for T1DM involves daily subcutaneous insulin injections, but a substantial proportion of patients face challenges such as severe hypoglycemic episodes and poorly controlled hyperglycemia. For T1DM patients, a more effective therapeutic option involves the replacement of β-cells through allogeneic transplantation of either the entire pancreas or isolated pancreatic islets. Unfortunately, the scarcity of transplantable human organs has led to a growing list of patients waiting for an islet transplant. One potential alternative is xenotransplantation of porcine pancreatic islets. However, due to inter-species molecular incompatibilities, porcine tissues trigger a robust immune response in humans, leading to xenograft rejection. Several promising strategies aim to overcome this challenge and enhance the long-term survival and functionality of xenogeneic islet grafts. These strategies include the use of islets derived from genetically modified pigs, immunoisolation of islets by encapsulation in biocompatible materials, and the creation of an immunomodulatory microenvironment by co-transplanting islets with accessory cells or utilizing immunomodulatory biomaterials. This review concentrates on delineating the primary obstacles in islet xenotransplantation and elucidates the fundamental principles and recent breakthroughs aimed at addressing these challenges.
Collapse
Affiliation(s)
- Sarah Grimus
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| | - Victoria Sarangova
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Petra B. Welzel
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Barbara Ludwig
- Department of Medicine IIIUniversity Hospital Carl Gustav CarusTechnische Universität DresdenD‐01307DresdenGermany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the Technische Universität DresdenD‐01307DresdenGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
- DFG‐Center for Regenerative Therapies DresdenTechnische Universität DresdenD‐01307DresdenGermany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IVDiabetes Zentrum – Campus InnenstadtKlinikum der Ludwig‐Maximilians‐Universität MünchenD‐80336MunichGermany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Asghar Ali
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| |
Collapse
|
5
|
Turan A, Tarique M, Zhang L, Kazmi S, Ulker V, Tedla MG, Badal D, Yolcu ES, Shirwan H. Engineering Pancreatic Islets to Transiently Codisplay on Their Surface Thrombomodulin and CD47 Immunomodulatory Proteins as a Means of Mitigating Instant Blood-Mediated Inflammatory Reaction following Intraportal Transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1971-1980. [PMID: 38709159 PMCID: PMC11160431 DOI: 10.4049/jimmunol.2300743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/01/2024] [Indexed: 05/07/2024]
Abstract
Most pancreatic islets are destroyed immediately after intraportal transplantation by an instant blood-mediated inflammatory reaction (IBMIR) generated through activation of coagulation, complement, and proinflammatory pathways. Thus, effective mitigation of IBMIR may be contingent on the combined use of agents targeting these pathways for modulation. CD47 and thrombomodulin (TM) are two molecules with distinct functions in regulating coagulation and proinflammatory responses. We previously reported that the islet surface can be modified with biotin for transient display of novel forms of these two molecules chimeric with streptavidin (SA), that is, thrombomodulin chimeric with SA (SA-TM) and CD47 chimeric with SA (SA-CD47), as single agents with improved engraftment following intraportal transplantation. This study aimed to test whether islets can be coengineered with SA-TM and SA-CD47 molecules as a combinatorial approach to improve engraftment by inhibiting IBMIR. Mouse islets were effectively coengineered with both molecules without a detectable negative impact on their viability and metabolic function. Coengineered islets were refractory to destruction by IBMIR ex vivo and showed enhanced engraftment and sustained function in a marginal mass syngeneic intraportal transplantation model. Improved engraftment correlated with a reduction in intragraft innate immune infiltrates, particularly neutrophils and M1 macrophages. Moreover, transcripts for various intragraft procoagulatory and proinflammatory agents, including tissue factor, HMGB1 (high-mobility group box-1), IL-1β, IL-6, TNF-α, IFN-γ, and MIP-1α, were significantly reduced in coengineered islets. These data demonstrate that the transient codisplay of SA-TM and SA-CD47 proteins on the islet surface is a facile and effective platform to modulate procoagulatory and inflammatory responses with implications for both autologous and allogeneic islet transplantation.
Collapse
Affiliation(s)
- Ali Turan
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Mohammad Tarique
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Lei Zhang
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Shadab Kazmi
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Vahap Ulker
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Mebrahtu G Tedla
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Darshan Badal
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Esma S Yolcu
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Haval Shirwan
- Department of Pediatrics and Department of Molecular Microbiology and Immunology, NextGen Precision Health Institute, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| |
Collapse
|
6
|
Yuan Y, Cui Y, Zhao D, Yuan Y, Zhao Y, Li D, Jiang X, Zhao G. Complement networks in gene-edited pig xenotransplantation: enhancing transplant success and addressing organ shortage. J Transl Med 2024; 22:324. [PMID: 38566098 PMCID: PMC10986007 DOI: 10.1186/s12967-024-05136-4] [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: 12/26/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
The shortage of organs for transplantation emphasizes the urgent need for alternative solutions. Xenotransplantation has emerged as a promising option due to the greater availability of donor organs. However, significant hurdles such as hyperacute rejection and organ ischemia-reperfusion injury pose major challenges, largely orchestrated by the complement system, and activated immune responses. The complement system, a pivotal component of innate immunity, acts as a natural barrier for xenotransplantation. To address the challenges of immune rejection, gene-edited pigs have become a focal point, aiming to shield donor organs from human immune responses and enhance the overall success of xenotransplantation. This comprehensive review aims to illuminate strategies for regulating complement networks to optimize the efficacy of gene-edited pig xenotransplantation. We begin by exploring the impact of the complement system on the effectiveness of xenotransplantation. Subsequently, we delve into the evaluation of key complement regulators specific to gene-edited pigs. To further understand the status of xenotransplantation, we discuss preclinical studies that utilize gene-edited pigs as a viable source of organs. These investigations provide valuable insights into the feasibility and potential success of xenotransplantation, offering a bridge between scientific advancements and clinical application.
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
| | - Yuanyuan Cui
- 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
| | - Dayue 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
| | - Yuan 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
| | - Yanshuang Zhao
- Department of Pharmacy, The People's Hospital of Leshan, Leshan, China
| | - Danni Li
- Department of Pharmacy, Longquanyi District of Chengdu Maternity & Child Health Care Hospital, Chengdu, China
| | - Xiaomei Jiang
- 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
| | - 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
|
7
|
Eisenson DL, Iwase H, Chen W, Hisadome Y, Cui W, Santillan MR, Schulick AC, Gu D, Maxwell A, Koenig K, Sun Z, Warren D, Yamada K. Combined islet and kidney xenotransplantation for diabetic nephropathy: an update in ongoing research for a clinically relevant application of porcine islet transplantation. Front Immunol 2024; 15:1351717. [PMID: 38476227 PMCID: PMC10927755 DOI: 10.3389/fimmu.2024.1351717] [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: 12/06/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Combined islet and kidney xenotransplantation for the treatment of diabetic nephropathy represents a compelling and increasingly relevant therapeutic possibility for an ever-growing number of patients who would benefit from both durable renal replacement and cure of the underlying cause of their renal insufficiency: diabetes. Here we briefly review immune barriers to islet transplantation, highlight preclinical progress in the field, and summarize our experience with combined islet and kidney xenotransplantation, including both challenges with islet-kidney composite grafts as well as our recent success with sequential kidney followed by islet xenotransplantation in a pig-to-baboon model.
Collapse
Affiliation(s)
- Daniel L. Eisenson
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hayato Iwase
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Weili Chen
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yu Hisadome
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wanxing Cui
- Cell Therapy and Manufacturing, Medstar Georgetown University Hospital, Washington DC, United States
| | - Michelle R. Santillan
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alexander C. Schulick
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Du Gu
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amanda Maxwell
- Research Animal Resources, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kristy Koenig
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhaoli Sun
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniel Warren
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kazuhiko Yamada
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
8
|
Abstract
End-stage organ failure can result from various preexisting conditions and occurs in patients of all ages, and organ transplantation remains its only treatment. In recent years, extensive research has been done to explore the possibility of transplanting animal organs into humans, a process referred to as xenotransplantation. Due to their matching organ sizes and other anatomical and physiological similarities with humans, pigs are the preferred organ donor species. Organ rejection due to host immune response and possible interspecies infectious pathogen transmission have been the biggest hurdles to xenotransplantation's success. Use of genetically engineered pigs as tissue and organ donors for xenotransplantation has helped to address these hurdles. Although several preclinical trials have been conducted in nonhuman primates, some barriers still exist and demand further efforts. This review focuses on the recent advances and remaining challenges in organ and tissue xenotransplantation.
Collapse
Affiliation(s)
- Asghar Ali
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; , ,
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Elisabeth Kemter
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; , ,
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; , ,
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| |
Collapse
|
9
|
Zhao Y, Nicholson L, Wang H, Qian YW, Hawthorne WJ, Jimenez-Vera E, Gloss BS, Lai J, Thomas A, Chew YV, Burns H, Zhang GY, Wang YM, Rogers NM, Zheng G, Yi S, Alexander SI, O’Connell PJ, Hu M. Intragraft memory-like CD127hiCD4+Foxp3+ Tregs maintain transplant tolerance. JCI Insight 2024; 9:e169119. [PMID: 38516885 PMCID: PMC11063946 DOI: 10.1172/jci.insight.169119] [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: 02/14/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) play an essential role in suppressing transplant rejection, but their role within the graft and heterogeneity in tolerance are poorly understood. Here, we compared phenotypic and transcriptomic characteristics of Treg populations within lymphoid organs and grafts in an islet xenotransplant model of tolerance. We showed Tregs were essential for tolerance induction and maintenance. Tregs demonstrated heterogeneity within the graft and lymphoid organs of tolerant mice. A subpopulation of CD127hi Tregs with memory features were found in lymphoid organs, presented in high proportions within long-surviving islet grafts, and had a transcriptomic and phenotypic profile similar to tissue Tregs. Importantly, these memory-like CD127hi Tregs were better able to prevent rejection by effector T cells, after adoptive transfer into secondary Rag-/- hosts, than naive Tregs or unselected Tregs from tolerant mice. Administration of IL-7 to the CD127hi Treg subset was associated with a strong activation of phosphorylation of STAT5. We proposed that memory-like CD127hi Tregs developed within the draining lymph node and underwent further genetic reprogramming within the graft toward a phenotype that had shared characteristics with other tissue or tumor Tregs. These findings suggested that engineering Tregs with these characteristics either in vivo or for adoptive transfer could enhance transplant tolerance.
Collapse
Affiliation(s)
| | | | - Hannah Wang
- Centre for Transplant and Renal Research and
| | - Yi Wen Qian
- Centre for Transplant and Renal Research and
| | | | | | - Brian S. Gloss
- Scientific Platforms, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Joey Lai
- Scientific Platforms, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | | | - Yi Vee Chew
- Centre for Transplant and Renal Research and
| | | | - Geoff Y. Zhang
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - Natasha M. Rogers
- Centre for Transplant and Renal Research and
- Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | | | - Shounan Yi
- Centre for Transplant and Renal Research and
| | - Stephen I. Alexander
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | | | - Min Hu
- Centre for Transplant and Renal Research and
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
10
|
Goyal P, Malviya R. Stem Cell Therapy for the Management of Type 1 Diabetes: Advances and Perspectives. Endocr Metab Immune Disord Drug Targets 2024; 24:549-561. [PMID: 37861029 DOI: 10.2174/0118715303256582230919093535] [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: 04/05/2023] [Revised: 07/20/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Due to insulin resistance and excessive blood sugar levels, type 1 diabetes mellitus (T1DM) is characterized by pancreatic cell loss. This condition affects young people at a higher rate than any other chronic autoimmune disease. Regardless of the method, exogenous insulin cannot substitute for insulin produced by a healthy pancreas. An emerging area of medicine is pancreatic and islet transplantation for type 1 diabetics to restore normal blood sugar regulation. However, there are still obstacles standing in the way of the widespread use of these therapies, including very low availability of pancreatic and islets supplied from human organ donors, challenging transplantation conditions, high expenses, and a lack of easily accessible methods. Efforts to improve Type 1 Diabetes treatment have been conducted in response to the disease's increasing prevalence. Type 1 diabetes may one day be treated with stem cell treatment. Stem cell therapy has proven to be an effective treatment for type 1 diabetes. Recent progress in stem cell-based diabetes treatment is summarised, and the authors show how to isolate insulin-producing cells (IPCs) from a variety of progenitor cells.
Collapse
Affiliation(s)
- Priyanshi Goyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
11
|
Salvaris EJ, Fisicaro N, McIlfatrick S, Thomas A, Fuller E, Lew AM, Nottle MB, Hawthorne WJ, Cowan PJ. Characterisation of transgenic pigs expressing a human T cell-depleting anti-CD2 monoclonal antibody. Xenotransplantation 2023; 31:e12836. [PMID: 37961013 PMCID: PMC10909556 DOI: 10.1111/xen.12836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 10/03/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Pig islet xenotransplantation is a potential treatment for type 1 diabetes. We have shown that maintenance immunosuppression is required to protect genetically modified (GM) porcine islet xenografts from T cell-mediated rejection in baboons. Local expression of a depleting anti-CD2 monoclonal antibody (mAb) by the xenograft may provide an alternative solution. We have previously reported the generation of GGTA1 knock-in transgenic pigs expressing the chimeric anti-CD2 mAb diliximab under an MHC class I promoter (MHCIP). In this study, we generated GGTA1 knock-in pigs in which MHCIP was replaced by the β-cell-specific porcine insulin promoter (PIP), and compared the pattern of diliximab expression in the two lines. METHODS A PIP-diliximab knock-in construct was prepared and validated by transfection of NIT-1 mouse insulinoma cells. The construct was knocked into GGTA1 in wild type (WT) porcine fetal fibroblasts using CRISPR, and knock-in cells were used to generate pigs by somatic cell nuclear transfer (SCNT). Expression of the transgene in MHCIP-diliximab and PIP-diliximab knock-in pigs was characterised at the mRNA and protein levels using RT-qPCR, flow cytometry, ELISA and immunohistochemistry. Islets from MHCIP-diliximab and control GGTA1 KO neonatal pigs were transplanted under the kidney capsule of streptozotocin-diabetic SCID mice. RESULTS NIT-1 cells stably transfected with the PIP-diliximab knock-in construct secreted diliximab into the culture supernatant, confirming correct expression and processing of the mAb in β cells. PIP-diliximab knock-in pigs showed a precise integration of the transgene within GGTA1. Diliximab mRNA was detected in all tissues tested (spleen, kidney, heart, liver, lung, pancreas) in MHCIP-diliximab pigs, but was not detectable in PIP-diliximab pigs. Likewise, diliximab was present in the serum of MHCIP-diliximab pigs, at a mean concentration of 1.8 μg/mL, but was not detected in PIP-diliximab pig serum. An immunohistochemical survey revealed staining for diliximab in all organs of MHCIP-diliximab pigs but not of PIP-diliximab pigs. Whole genome sequencing (WGS) of a PIP-diliximab pig identified a missense mutation in the coding region for the dixilimab light chain. This mutation was also found to be present in the fibroblast knock-in clone used to generate the PIP-diliximab pigs. Islet xenografts from neonatal MHCIP-diliximab pigs restored normoglycemia in diabetic immunodeficient mice, indicating no overt effect of the transgene on islet function, and demonstrated expression of diliximab in situ. CONCLUSION Diliximab was widely expressed in MHCIP-diliximab pigs, including in islets, consistent with the endogenous expression pattern of MHC class I. Further investigation is required to determine whether the level of expression in islets from the MHCIP-diliximab pigs is sufficient to prevent T cell-mediated islet xenograft rejection. The unexpected absence of diliximab expression in the islets of PIP-diliximab pigs was probably due to a mutation in the transgene arising during the generation of the knock-in cells used for SCNT.
Collapse
Affiliation(s)
- Evelyn J. Salvaris
- Immunology Research CentreSt. Vincent's Hospital MelbourneFitzroyVictoriaAustralia
| | - Nella Fisicaro
- Immunology Research CentreSt. Vincent's Hospital MelbourneFitzroyVictoriaAustralia
| | - Stephen McIlfatrick
- Robinson Research Institute and School of BiomedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Adwin Thomas
- The Centre for Transplant & Renal ResearchWestmead Institute for Medical ResearchWestmeadNew South WalesAustralia
| | - Erin Fuller
- The Centre for Transplant & Renal ResearchWestmead Institute for Medical ResearchWestmeadNew South WalesAustralia
| | - Andrew M. Lew
- Walter and Eliza Hall InstituteDepartment of Medical Biology and Department of Microbiology & ImmunologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Mark B. Nottle
- Robinson Research Institute and School of BiomedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Wayne J. Hawthorne
- The Centre for Transplant & Renal ResearchWestmead Institute for Medical ResearchWestmeadNew South WalesAustralia
- Department of SurgeryWestmead HospitalSchool of Medical SciencesUniversity of SydneyWestmeadNew South WalesAustralia
| | - Peter J. Cowan
- Immunology Research CentreSt. Vincent's Hospital MelbourneFitzroyVictoriaAustralia
- Department of MedicineUniversity of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
12
|
Naqvi RA, Naqvi AR, Singh A, Priyadarshini M, Balamurugan AN, Layden BT. The future treatment for type 1 diabetes: Pig islet- or stem cell-derived β cells? Front Endocrinol (Lausanne) 2023; 13:1001041. [PMID: 36686451 PMCID: PMC9849241 DOI: 10.3389/fendo.2022.1001041] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/07/2022] [Indexed: 01/06/2023] Open
Abstract
Replacement of β cells is only a curative approach for type 1 diabetes (T1D) patients to avoid the threat of iatrogenic hypoglycemia. In this pursuit, islet allotransplantation under Edmonton's protocol emerged as a medical miracle to attain hypoglycemia-free insulin independence in T1D. Shortage of allo-islet donors and post-transplantation (post-tx) islet loss are still unmet hurdles for the widespread application of this therapeutic regimen. The long-term survival and effective insulin independence in preclinical studies have strongly suggested pig islets to cure overt hyperglycemia. Importantly, CRISPR-Cas9 technology is pursuing to develop "humanized" pig islets that could overcome the lifelong immunosuppression drug regimen. Lately, induced pluripotent stem cell (iPSC)-derived β cell approaches are also gaining momentum and may hold promise to yield a significant supply of insulin-producing cells. Theoretically, personalized β cells derived from a patient's iPSCs is one exciting approach, but β cell-specific immunity in T1D recipients would still be a challenge. In this context, encapsulation studies on both pig islet as well as iPSC-β cells were found promising and rendered long-term survival in mice. Oxygen tension and blood vessel growth within the capsules are a few of the hurdles that need to be addressed. In conclusion, challenges associated with both procedures, xenotransplantation (of pig-derived islets) and stem cell transplantation, are required to be cautiously resolved before their clinical application.
Collapse
Affiliation(s)
- Raza Ali Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Afsar Raza Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Amar Singh
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Medha Priyadarshini
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Appakalai N. Balamurugan
- Center for Clinical and Translational Research, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Brian T. Layden
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
13
|
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
|
14
|
Eisenson DL, Hisadome Y, Santillan MR, Yamada K. Progress in islet xenotransplantation: Immunologic barriers, advances in gene editing, and tolerance induction strategies for xenogeneic islets in pig-to-primate transplantation. FRONTIERS IN TRANSPLANTATION 2022; 1:989811. [PMID: 38390384 PMCID: PMC10883655 DOI: 10.3389/frtra.2022.989811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Islet transplantation has emerged as a curative therapy for diabetes in select patients but remains rare due to shortage of suitable donor pancreases. Islet transplantation using porcine islets has long been proposed as a solution to this organ shortage. There have already been several small clinical trials using porcine islets in humans, but results have been mixed and further trials limited by calls for more rigorous pre-clinical data. Recent progress in heart and kidney xenograft transplant, including three studies of pig-to-human xenograft transplant, have recaptured popular imagination and renewed interest in clinical islet xenotransplantation. This review outlines immunologic barriers to islet transplantation, summarizes current strategies to overcome these barriers with a particular focus on approaches to induce tolerance, and describes an innovative strategy for treatment of diabetic nephropathy with composite islet-kidney transplantation.
Collapse
Affiliation(s)
- Daniel L Eisenson
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Yu Hisadome
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, United States
| | | | - Kazuhiko Yamada
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
15
|
Pignatelli C, Campo F, Neroni A, Piemonti L, Citro A. Bioengineering the Vascularized Endocrine Pancreas: A Fine-Tuned Interplay Between Vascularization, Extracellular-Matrix-Based Scaffold Architecture, and Insulin-Producing Cells. Transpl Int 2022; 35:10555. [PMID: 36090775 PMCID: PMC9452644 DOI: 10.3389/ti.2022.10555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022]
Abstract
Intrahepatic islet transplantation is a promising β-cell replacement strategy for the treatment of type 1 diabetes. Instant blood-mediated inflammatory reactions, acute inflammatory storm, and graft revascularization delay limit islet engraftment in the peri-transplant phase, hampering the success rate of the procedure. Growing evidence has demonstrated that islet engraftment efficiency may take advantage of several bioengineering approaches aimed to recreate both vascular and endocrine compartments either ex vivo or in vivo. To this end, endocrine pancreas bioengineering is an emerging field in β-cell replacement, which might provide endocrine cells with all the building blocks (vascularization, ECM composition, or micro/macro-architecture) useful for their successful engraftment and function in vivo. Studies on reshaping either the endocrine cellular composition or the islet microenvironment have been largely performed, focusing on a single building block element, without, however, grasping that their synergistic effect is indispensable for correct endocrine function. Herein, the review focuses on the minimum building blocks that an ideal vascularized endocrine scaffold should have to resemble the endocrine niche architecture, composition, and function to foster functional connections between the vascular and endocrine compartments. Additionally, this review highlights the possibility of designing bioengineered scaffolds integrating alternative endocrine sources to overcome donor organ shortages and the possibility of combining novel immune-preserving strategies for long-term graft function.
Collapse
Affiliation(s)
- Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Campo
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessia Neroni
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
16
|
Hu M, Hawthorne WJ, Yi S, O’Connell PJ. Cellular Immune Responses in Islet Xenograft Rejection. Front Immunol 2022; 13:893985. [PMID: 35874735 PMCID: PMC9300897 DOI: 10.3389/fimmu.2022.893985] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
Porcine islets surviving the acute injury caused by humoral rejection and IBMIR will be subjected to cellular xenograft rejection, which is predominately mediated by CD4+ T cells and is characterised by significant infiltration of macrophages, B cells and T cells (CD4+ and CD8+). Overall, the response is different compared to the alloimmune response and more difficult to suppress. Activation of CD4+ T cells is both by direct and indirect antigen presentation. After activation they recruit macrophages and direct B cell responses. Although they are less important than CD4+ T cells in islet xenograft rejection, macrophages are believed to be a major effector cell in this response. Rodent studies have shown that xenoantigen-primed and CD4+ T cell-activated macrophages were capable of recognition and rejection of pancreatic islet xenografts, and they destroyed a graft via the secretion of various proinflammatory mediators, including TNF-α, reactive oxygen and nitrogen species, and complement factors. B cells are an important mediator of islet xenograft rejection via xenoantigen presentation, priming effector T cells and producing xenospecific antibodies. Depletion and/or inhibition of B cells combined with suppressing T cells has been suggested as a promising strategy for induction of xeno-donor-specific T- and B-cell tolerance in islet xenotransplantation. Thus, strategies that expand the influence of regulatory T cells and inhibit and/or reduce macrophage and B cell responses are required for use in combination with clinical applicable immunosuppressive agents to achieve effective suppression of the T cell-initiated xenograft response.
Collapse
Affiliation(s)
- Min Hu
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Wayne J. Hawthorne
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Shounan Yi
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Philip J. O’Connell
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Philip J. O’Connell,
| |
Collapse
|
17
|
Hawthorne WJ, Salvaris EJ, Chew YV, Burns H, Hawkes J, Barlow H, Hu M, Lew AM, Nottle MB, O’Connell PJ, Cowan PJ. Xenotransplantation of Genetically Modified Neonatal Pig Islets Cures Diabetes in Baboons. Front Immunol 2022; 13:898948. [PMID: 35784286 PMCID: PMC9243461 DOI: 10.3389/fimmu.2022.898948] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
Xenotransplantation using porcine donors is rapidly approaching clinical applicability as an alternative therapy for treatment of many end-stage diseases including type 1 diabetes. Porcine neonatal islet cell clusters (NICC) have normalised blood sugar levels for relatively short periods in the preclinical diabetic rhesus model but have met with limited success in the stringent baboon model. Here we report that NICC from genetically modified (GM) pigs deleted for αGal and expressing the human complement regulators CD55 and CD59 can cure diabetes long-term in immunosuppressed baboons, with maximum graft survival exceeding 22 months. Five diabetic baboons were transplanted intraportally with 9,673 – 56,913 islet equivalents (IEQ) per kg recipient weight. Immunosuppression consisted of T cell depletion with an anti-CD2 mAb, tacrolimus for the first 4 months, and maintenance with belatacept and anti-CD154; no anti-inflammatory treatment or cytomegalovirus (CMV) prophylaxis/treatment was given. This protocol was well tolerated, with all recipients maintaining or gaining weight. Recipients became insulin-independent at a mean of 87 ± 43 days post-transplant and remained insulin-independent for 397 ± 174 days. Maximum graft survival was 675 days. Liver biopsies showed functional islets staining for all islet endocrine components, with no evidence of the inflammatory blood-mediated inflammatory reaction (IBMIR) and minimal leukocytic infiltration. The costimulation blockade-based immunosuppressive protocol prevented an anti-pig antibody response in all recipients. In conclusion, we demonstrate that genetic modification of the donor pig enables attenuation of early islet xenograft injury, and in conjunction with judicious immunosuppression provides excellent long-term function and graft survival in the diabetic baboon model.
Collapse
Affiliation(s)
- Wayne J. Hawthorne
- The Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Department of Surgery, Westmead Hospital, School of Medical Sciences, University of Sydney, Westmead, NSW, Australia
- *Correspondence: Wayne J. Hawthorne,
| | - Evelyn J. Salvaris
- Immunology Research Centre, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Yi Vee Chew
- The Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Heather Burns
- The Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Joanne Hawkes
- The Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Helen Barlow
- Immunology Research Centre, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Min Hu
- The Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Andrew M. Lew
- Division of Immunology, Walter and Eliza Hall Institute, Melbourne, VIC, Australia
| | - Mark B. Nottle
- Department of Obstetrics and Gynaecology, University of Adelaide, Adelaide, SA, Australia
| | - Philip J. O’Connell
- The Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Peter J. Cowan
- Immunology Research Centre, St. Vincent’s Hospital, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Graham ML, Ramachandran S, Singh A, Moore MEG, Flanagan EB, Azimzadeh A, Burlak C, Mueller KR, Martins K, Anazawa T, Balamurugan AN, Bansal-Pakala P, Murtaugh MP, O’Brien TD, Papas KK, Spizzo T, Schuurman HJ, Hancock WW, Hering BJ. Clinically available immunosuppression averts rejection but not systemic inflammation after porcine islet xenotransplant in cynomolgus macaques. Am J Transplant 2022; 22:745-760. [PMID: 34704345 PMCID: PMC9832996 DOI: 10.1111/ajt.16876] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/30/2021] [Accepted: 10/19/2021] [Indexed: 01/25/2023]
Abstract
A safe, efficacious, and clinically applicable immunosuppressive regimen is necessary for islet xenotransplantation to become a viable treatment option for diabetes. We performed intraportal transplants of wild-type adult porcine islets in 25 streptozotocin-diabetic cynomolgus monkeys. Islet engraftment was good in 21, partial in 3, and poor in 1 recipient. Median xenograft survival was 25 days with rapamycin and CTLA4Ig immunosuppression. Adding basiliximab induction and maintenance tacrolimus to the base regimen significantly extended median graft survival to 147 days (p < .0001), with three animals maintaining insulin-free xenograft survival for 265, 282, and 288 days. We demonstrate that this regimen suppresses non-Gal anti-pig antibody responses, circulating effector memory T cell expansion, effector function, and infiltration of the graft. However, a chronic systemic inflammatory state manifested in the majority of recipients with long-term graft survival indicated by increased neutrophil to lymphocyte ratio, IL-6, MCP-1, CD40, and CRP expression. This suggests that this immunosuppression regimen fails to regulate innate immunity and resulting inflammation is significantly associated with increased incidence and severity of adverse events making this regimen unacceptable for translation. Additional studies are needed to optimize a maintenance regimen for regulating the innate inflammatory response.
Collapse
Affiliation(s)
- Melanie L. Graham
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Meghan E. G. Moore
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN
| | - E. Brian Flanagan
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Agnes Azimzadeh
- Department of Surgery, University of Maryland, Baltimore, MD
| | - Christopher Burlak
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Kate R. Mueller
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Kyra Martins
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Takayuki Anazawa
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Pratima Bansal-Pakala
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Michael P. Murtaugh
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Timothy D. O’Brien
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN
| | - Klearchos K. Papas
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Henk-J. Schuurman
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN,Spring Point Project, Minneapolis, MN
| | - Wayne W. Hancock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bernhard. J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| |
Collapse
|
19
|
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
|
20
|
Hawthorne WJ, Fuller E, Thomas A, Rao JS, Burlak C. Updateon xenotransplantation for May/June 2021. Xenotransplantation 2021; 28:e12710. [PMID: 34617623 DOI: 10.1111/xen.12710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Wayne J Hawthorne
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Department of Surgery, Westmead Clinical School, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
| | - Erin Fuller
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Adwin Thomas
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Joseph Sushil Rao
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA.,Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher Burlak
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Human islet transplantation has proven to be a highly effective treatment for patients with labile type 1 diabetes mellitus, which can free patients from daily glucose monitoring and insulin injections. However, the shortage of islet donors limits its' broad application. Porcine islet xenotransplantation presents a solution to the donor shortage and recent advances in genetic modification and immunosuppressive regimens provide renewed enthusiasm for the potential of this treatment. RECENT FINDINGS Advances in genetic editing technology are leading to multigene modified porcine islet donors with alterations in expression of known xenoantigens, modifications of their complement and coagulation systems, and modifications to gain improved immunological compatibility. Recent NHP-based trials of costimulation blockade using CD154 blockade show promising improvements in islet survival, whereas results targeting CD40 are less consistent. Furthermore, trials using IL-6 receptor antagonism have yet to demonstrate improvement in glucose control and suffer from poor graft revascularization. SUMMARY This review will detail the current status of islet xenotransplantation as a potential treatment for type I diabetes mellitus, focusing on recent advances in porcine xenogeneic islet production, assessment in nonhuman primate preclinical models, the outcome of human clinical trials and review barriers to translation of xenoislets to the clinic.
Collapse
|
22
|
Niu D, Ma X, Yuan T, Niu Y, Xu Y, Sun Z, Ping Y, Li W, Zhang J, Wang T, Church GM. Porcine genome engineering for xenotransplantation. Adv Drug Deliv Rev 2021; 168:229-245. [PMID: 32275950 DOI: 10.1016/j.addr.2020.04.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/28/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
The extreme shortage of human donor organs for treatment of patients with end-stage organ failures is well known. Xenotransplantation, which might provide unlimited organ supply, is a most promising strategy to solve this problem. Domestic pigs are regarded as ideal organ-source animals owing to similarity in anatomy, physiology and organ size to humans as well as high reproductive capacity and low maintenance cost. However, several barriers, which include immune rejection, inflammation and coagulative dysfunctions, as well as the cross-species transmission risk of porcine endogenous retrovirus, blocked the pig-to-human xenotransplantation. With the rapid development of genome engineering technologies and the potent immunosuppressive medications in recent years, these barriers could be eliminated through genetic modification of pig genome together with the administration of effective immunosuppressants. A number of candidate genes involved in the regulation of immune response, inflammation and coagulation have been explored to optimize porcine xenograft survival in non-human primate recipients. PERV inactivation in pigs has also been accomplished to firmly address the safety issue in pig-to-human xenotransplantation. Many encouraging preclinical milestones have been achieved with some organs surviving for years. Therefore, the clinical trials of some promising organs, such as islet, kidney and heart, are aimed to be launched in the near future.
Collapse
Affiliation(s)
- Dong Niu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, P.R. China
| | - Xiang Ma
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, P.R. China
| | - Taoyan Yuan
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, China
| | - Yifan Niu
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, Jiangsu 211300, China
| | - Yibin Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhongxin Sun
- Cosmetic & Plastic Surgery Department, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Weifen Li
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jufang Zhang
- Cosmetic & Plastic Surgery Department, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, China.
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, Jiangsu 211300, China.
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
23
|
Zammit NW, Seeberger KL, Zamerli J, Walters SN, Lisowski L, Korbutt GS, Grey ST. Selection of a novel AAV2/TNFAIP3 vector for local suppression of islet xenograft inflammation. Xenotransplantation 2020; 28:e12669. [PMID: 33316848 DOI: 10.1111/xen.12669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/24/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Neonatal porcine islets (NPIs) can restore glucose control in mice, pigs, and non-human primates, representing a potential abundant alternative islet supply for clinical beta cell replacement therapy. However, NPIs are vulnerable to inflammatory insults that could be overcome with genetic modifications. Here, we demonstrate in a series of proof-of-concept experiments the potential of the cytoplasmic ubiquitin-editing protein A20, encoded by the TNFAIP3 gene, as an NPI cytoprotective gene. METHODS We forced A20 expression in NPI grafts using a recombinant adenovirus 5 (Ad5) vector and looked for impact on TNF-stimulated NF-κB activation and NPI graft function. As adeno-associated vectors (AAV) are clinically preferred vectors but exhibit poor transduction efficacy in NPIs, we next screened a series of AAV serotypes under different transduction protocols for their ability achieve high transduction efficiency and suppress NPI inflammation without impacting NPI maturation. RESULTS Forcing the expression of A20 in NPI with Ad5 vector blocked NF-κB activation by inhibiting IκBα phosphorylation and degradation, and reduced the induction of pro-inflammatory genes Cxcl10 and Icam1. A20-expressing NPIs also exhibited superior functional capacity when transplanted into diabetic immunodeficient recipient mice, evidenced by a more rapid return to euglycemia and improved GTT compared to unmodified NPI grafts. We found AAV2 combined with a 14-day culture period maximized NPI transduction efficiency (>70% transduction rate), and suppressed NF-κB-dependent gene expression without adverse impact upon NPI maturation. CONCLUSION We report a new protocol that allows for high-efficiency genetic modification of NPIs, which can be utilized to introduce candidate genes without the need for germline engineering. This approach would be suitable for preclinical and clinical testing of beneficial molecules. We also report for the first time that A20 is cytoprotective for NPI, such that A20 gene therapy could aid the clinical development of NPIs for beta cell replacement.
Collapse
Affiliation(s)
- Nathan W Zammit
- Immunology Department, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| | | | - Jad Zamerli
- Immunology Department, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Stacey N Walters
- Immunology Department, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Leszek Lisowski
- Translational Vectorology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia.,Military Institute of Medicine, Laboratory of Molecular Oncology and Innovative Therapies, Warsaw, Poland
| | | | - Shane T Grey
- Immunology Department, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| |
Collapse
|
24
|
Nanno Y, Shajahan A, Sonon RN, Azadi P, Hering BJ, Burlak C. High-mannose type N-glycans with core fucosylation and complex-type N-glycans with terminal neuraminic acid residues are unique to porcine islets. PLoS One 2020; 15:e0241249. [PMID: 33170858 PMCID: PMC7654812 DOI: 10.1371/journal.pone.0241249] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/09/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Islet transplantation is an emerging treatment option for type 1 diabetes but its application is limited by the shortage of human pancreas donors. Characterization of the N- and O-glycan surface antigens that vary between human and genetically engineered porcine islet donors could shed light on targets of antibody mediated rejection. METHODS N- and O-glycans were isolated from human and adult porcine islets and analyzed using matrix-assisted laser-desorption time-of-flight mass spectrometry (MALDI-TOF-MS) and electrospray ionization mass spectrometry (ESI-MS/MS). RESULTS A total of 57 porcine and 34 human N-glycans and 21 porcine and 14 human O-glycans were detected from cultured islets. Twenty-eight of which were detected only from porcine islets, which include novel xenoantigens such as high-mannose type N-glycans with core fucosylation and complex-type N-glycans with terminal neuraminic acid residues. Porcine islets have terminal N-glycolylneuraminic acid (NeuGc) residue in bi-antennary N-glycans and sialyl-Tn O-glycans. No galactose-α-1,3-galactose (α-Gal) or Sda epitope were detected on any of the islets. CONCLUSIONS These results provide important insights into the potential antigenic differences of N- and O-glycan profiles between human and porcine islets. Glycan differences may identify novel gene targets for genetic engineering to generate superior porcine islet donors.
Collapse
Affiliation(s)
- Yoshihide Nanno
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States of America
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States of America
| | - Bernhard J. Hering
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Christopher Burlak
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
25
|
Walters EM, Burlak C. Xenotransplantation literature update, May/June2020. Xenotransplantation 2020; 27:e12638. [PMID: 32896009 DOI: 10.1111/xen.12638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/11/2019] [Indexed: 11/29/2022]
Affiliation(s)
| | - Christopher Burlak
- Department of Surgery, Schultz Diabetes Institutes, University of Minnesota School of Medicine, Minneapolis, MN, USA
| |
Collapse
|
26
|
Hawthorne WJ, Cowan PJ. Xenotransplantation in Australia: Development of the regulatory process. Xenotransplantation 2020; 27:e12603. [DOI: 10.1111/xen.12603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 01/19/2023]
Affiliation(s)
- Wayne J. Hawthorne
- Centre for Transplant and Renal Research The Westmead Institute for Medical ResearchWestmead Hospital Sydney NSW Australia
- Department of Surgery Western Clinical School University of Sydney Sydney NSW Australia
| | - Peter J. Cowan
- Immunology Research Centre St Vincent’s Hospital Melbourne Melbourne Vic. Australia
- Department of Medicine University of Melbourne Melbourne Vic. Australia
| |
Collapse
|
27
|
Thomas A, Hawthorne WJ, Burlak C. Xenotransplantation literature update, November/December 2019. Xenotransplantation 2020; 27:e12582. [PMID: 31984549 DOI: 10.1111/xen.12582] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 01/14/2020] [Indexed: 12/13/2022]
Abstract
The ever-increasing disparity between the lack of organ donors and patients on the transplant waiting list is increasing worldwide. For the past several decades xenotransplantation has led the way to correct this deficit and remains clearly the only feasible option to provide a means to meet the demand for patients in need of an organ transplant. Xenotransplantation's ability to provide a specifically designed unlimited supply of organs, suited to treat the various needs for transplant organs and cells, has recently been championed by successful pre-clinical trials that have run long-term in non-human primate studies. In this review we show how these improvements have come about due to long-term dedicated research and recent advances in biomedical engineering technology, such as genome editing tools including zinc finger nucleases, TALEN, and CRISPER/Cas9 which have paved the way for significant breakthroughs in improving xenograft outcomes through genetic modifications to the donor source pig. Other novel approaches include the development of decellularized porcine tissue, such as corneas which can now be transplanted into patients with the minimal need for immunosuppression or other side effects. Further genetic variants of the porcine genome are also now being optimized to abrogate rejection. The emergence of new modalities such as; mesenchymal stem cells, donor thymic vascularization, in vivo bioreactors, chemokine and cytokine therapies have come to show improvements in xenograft outcomes. Furthermore, new studies confirm the safety status of using porcine xenografts, verifying that with current technologies and approaches, the issue of PERV transmission is a moot point. These breakthroughs and technological advancements push the reality of xenotransplantation one step closer to the clinic.
Collapse
Affiliation(s)
- Adwin Thomas
- The Centre for Transplant & Renal Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Wayne J Hawthorne
- The Centre for Transplant & Renal Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,The Department of Surgery, University of Sydney, Westmead Hospital, Westmead, NSW, Australia
| | - Christopher Burlak
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
28
|
Samy KP, Gao Q, Davis RP, Song M, Fitch ZW, Mulvihill MS, MacDonald AL, Leopardi FV, How T, Williams KD, Devi GR, Collins BH, Luo X, Kirk AD. The role of human CD46 in early xenoislet engraftment in a dual transplant model. Xenotransplantation 2019; 26:e12540. [PMID: 31219218 PMCID: PMC6908747 DOI: 10.1111/xen.12540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/26/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Membrane cofactor protein CD46 attenuates the complement cascade by facilitating cleavage of C3b and C4b. In solid organ xenotransplantation, organs expressing CD46 have been shown to resist hyperacute rejection. However, the incremental value of human CD46 expression for islet xenotransplantation remains poorly defined. METHODS This study attempted to delineate the role of CD46 in early neonatal porcine islet engraftment by comparing Gal-knocked out (GKO) and hCD46-transgenic (GKO/CD46) islets in a dual transplant model. Seven rhesus macaques underwent dual transplant and were sacrificed at 1 hour (n = 4) or 24 hours (n = 3). Both hemilivers were recovered and fixed for immunohistochemistry (CD46, insulin, neutrophil elastase, platelet, IgM, IgG, C3d, C4d, CD68, Caspase 3). Quantitative immunohistochemical analysis was performed using the Aperio Imagescope. RESULTS Within 1 hour of intraportal infusion of xenografts, no differences were observed between the two types of islets in terms of platelet, antibody, or complement deposition. Cellular infiltration and islet apoptotic activity were also similar at 1 hour. At 24 hours, GKO/CD46 islets demonstrated significantly less platelet deposition (P = 0.01) and neutrophil infiltration (P = 0.01) compared to GKO islets. In contrast, C3d (P = 0.38) and C4d (P = 0.45) deposition was equal between the two genotypes. CONCLUSIONS Our findings suggest that expression of hCD46 on NPIs potentially provides a measurable incremental survival advantage in vivo by reducing early thrombo-inflammatory events associated with instant blood-mediated inflammatory reaction (IBMIR) following intraportal islet infusion.
Collapse
Affiliation(s)
- Kannan P Samy
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Qimeng Gao
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Robert Patrick Davis
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Mingqing Song
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Zachary W Fitch
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Michael S Mulvihill
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Andrea L MacDonald
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Frank V Leopardi
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Tam How
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Kyha D Williams
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Gayathri R Devi
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Bradley H Collins
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Xunrong Luo
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Allan D Kirk
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
29
|
Recent progress in porcine islet isolation, culture and engraftment strategies for xenotransplantation. Curr Opin Organ Transplant 2019; 23:633-641. [PMID: 30247169 DOI: 10.1097/mot.0000000000000579] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Xenotransplantation of porcine islets is a realistic option to restore β-cell function in type 1 diabetic patients. Among other factors, such as islet donor age (fetal, neonatal and adult) and genotype (wild type and genetically modified), choice of the transplantation site, and immune protection of the islets, efficient strategies for islet isolation, culture and engraftment are critical for the success of islet xenotransplantation. RECENT FINDINGS Neonatal porcine islets (NPIs) are immature at isolation and need to be matured in vitro or in vivo before they become fully functional. Recent developments include a scalable protocol for isolation of clinically relevant batches of NPIs and a stepwise differentiation protocol for directed maturation of NPIs. In addition, different sources of mesenchymal stem cells were shown to support survival and functional maturation of NPIs in vitro and in various transplantation models in vivo. SUMMARY A plethora of different culture media and supplements have been tested; however, a unique best culture system for NPIs is still missing. New insights, for example from single-cell analyses of islets or from stem cell differentiation toward β cells may help to optimize culture of porcine islets for xenotransplantation in an evidence-based manner.
Collapse
|
30
|
Wolf E, Kemter E, Klymiuk N, Reichart B. Genetically modified pigs as donors of cells, tissues, and organs for xenotransplantation. Anim Front 2019; 9:13-20. [PMID: 32002258 PMCID: PMC6951927 DOI: 10.1093/af/vfz014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Science, 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 Science, LMU Munich, Munich, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Science, LMU Munich, Munich, Germany
| | - Bruno Reichart
- Walter Brendel Center for Experimental Medicine, LMU Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany
| |
Collapse
|
31
|
Li X, Meng Q, Zhang L. Overcoming Immunobiological Barriers Against Porcine Islet Xenografts: What Should Be Done? Pancreas 2019; 48:299-308. [PMID: 30855426 DOI: 10.1097/mpa.0000000000001259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Porcine islets might represent an ideal solution to the severe shortage of living donor islets available for transplantation and thus have great potential for the treatment of diabetes. Although tremendous progress has been achieved through recent experiments, the immune response remains a major obstacle. This review first describes the 3 major pathways of rejection: hyperacute rejection mediated by preformed natural antibodies and complement, instant blood-mediated inflammatory reactions, and acute cell-mediated rejection. Furthermore, this review examines immune-related strategies, including major advances, which have been shown to extend the life and/or function of porcine islets in vitro and in vivo: (1) genetic modification to make porcine islets more compatible with the recipient, (2) optimization of the newly defined biological agents that have been shown to promote long-term survival of xenografts in nonhuman primates, and (3) development of novel immunoisolation technologies that maintain the long-term survival of islet xenografts without the use of systemic immunosuppressive drugs. Finally, the clinical application of porcine islet transplantation is presented. Even though less clinical information is available, experimental data indicate that porcine islet xenografts are likely to become a standard treatment for patients with type 1 diabetes in the future.
Collapse
Affiliation(s)
- Xinyu Li
- From the Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | |
Collapse
|
32
|
Hawthorne WJ, Burlak C. Xenotransplantation literature update, January/February 2018. Xenotransplantation 2018; 25:e12398. [PMID: 29654665 DOI: 10.1111/xen.12398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 03/24/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Wayne J Hawthorne
- Department of Surgery, Westmead Hospital, Sydney Medical School, University of Sydney, Westmead, NSW, Australia
| | - Christopher Burlak
- Department of Surgery, Schultz Diabetes Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Porcine islets represent a potentially attractive beta-cell source for xenotransplantation into patients with type 1 diabetes, who are not eligible to islet allo-transplantation due to a lack of suitable human donor organs. Recent progress in genetic engineering/gene editing of donor pigs provides new opportunities to overcome rejection of xeno-islets, to improve their engraftment and insulin secretion capacity, and to reduce the risk for transmission of porcine endogenous retroviruses. This review summarizes the current issues and progress in islet xenotransplantation with special emphasis on genetically modified/gene edited donor pigs. RECENT FINDINGS Attempts to overcome acute rejection of xeno-islets, especially after intraportal transplantation into the liver, include the genetic elimination of specific carbohydrate antigens such as αGal, Neu5Gc, and Sd(a) for which humans and-in part-non-human primates have natural antibodies that bind to these targets leading to activation of complement and coagulation. A complementary approach is the expression of one or more human complement regulatory proteins (hCD46, hCD55, hCD59). Transgenic attempts to overcome cellular rejection of islet xenotransplants include the expression of proteins that inhibit co-stimulation of T cells. Expression of glucagon-like peptide-1 and M3 muscarinic receptors has been shown to increase the insulin secretion of virally transduced porcine islets in vitro and it will be interesting to see the effects of these modifications in transgenic pigs and islet products derived from them. Genome-wide inactivation of porcine endogenous retrovirus (PERV) integrants by mutating their pol genes using CRISPR/Cas9 is a recent approach to reduce the risk for PERV transmission by xeno-islets. Genetic engineering/gene editing of xeno-islet donor pigs facilitated major progress towards clinical islet xenotransplantation. The required set of genetic modifications will depend on the source of islets (fetal/neonatal vs. adult), the mode of delivery (encapsulated vs. free), and the transplantation site.
Collapse
Affiliation(s)
- Elisabeth Kemter
- Gene Center, and Center for Innovative Medical Models (CiMM), LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Joachim Denner
- Robert Koch Institute, Nordufer 20, 13353, Berlin, Germany
| | - Eckhard Wolf
- Gene Center, and Center for Innovative Medical Models (CiMM), LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
34
|
Abstract
β cell replacement with either pancreas or islet transplantation has progressed immensely over the last decades with current 1- and 5-year insulin independence rates of approximately 85% and 50%, respectively. Recent advances are largely attributed to improvements in immunosuppressive regimen, donor selection, and surgical technique. However, both strategies are compromised by a scarce donor source. Xenotransplantation offers a potential solution by providing a theoretically unlimited supply of islets, but clinical application has been limited by concerns for a potent immune response against xenogeneic tissue. β cell clusters derived from embryonic or induced pluripotent stem cells represent another promising unlimited source of insulin producing cells, but clinical application is pending further advances in the function of the β cell like clusters. Exciting developments and rapid progress in all areas of β cell replacement prompted a lively debate by members of the young investigator committee of the International Pancreas and Islet Transplant Association at the 15th International Pancreas and Islet Transplant Association Congress in Melbourne and at the 26th international congress of The Transplant Society in Hong Kong. This international group of young investigators debated which modality of β cell replacement would predominate the landscape in 10 years, and their arguments are summarized here.
Collapse
|
35
|
Bottino R, Knoll MF, Knoll CA, Bertera S, Trucco MM. The Future of Islet Transplantation Is Now. Front Med (Lausanne) 2018; 5:202. [PMID: 30057900 PMCID: PMC6053495 DOI: 10.3389/fmed.2018.00202] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
Abstract
Milestones in the history of diabetes therapy include the discovery of insulin and successful methods of beta cell replacement including whole pancreas and islet cell transplantation options. While pancreas transplantation remains the gold standard for patients who have difficulty controlling their symptoms with exogenous insulin, islet allotransplantation is now able to provide similar results with some advantages that make it an attractive potential alternative. The Edmonton Protocol, which incorporated a large dose of islets from multiple donors with steroid-free immunosuppression helped to establish the modern era of islet transplantation almost 20 years ago. While islet allotransplantation is recognized around the world as a powerful clinical therapy for type 1 diabetes it is not yet recognized by the Federal Drug Administration of the United States. Large-scale clinical trials administered by the Clinical Islet Transplantation Consortium have recently demonstrated that the well-regulated manufacture of a human islet product transplanted into patients with difficult to control type 1 diabetes and with a history of severe hyperglycemic episodes can safely and efficaciously maintain glycemic balance and eliminate the most severe complications associated with diabetes. The results of these clinical trials have established a strong basis for licensure of clinical islet allotransplantation in the US. Recognition by the Federal Drug Administration would likely lead to third party reimbursement for islet allotransplantation as a therapeutic option in the United States and would make the treatment available to many more patients. The high costs of rampant diabetes justify the expense of the treatment, which is in-line with the costs of clinical pancreas transplantation. While much enthusiasm and hope is raised toward the development and optimization of stem cell therapy, the islet transplantation community should push toward licensure, if that means broader access of this procedure to patients who may benefit from it. Even as we prepare to take the first steps in that direction, we must acknowledge the new challenges that a shift from the experimental to clinical will bring. Clinical islet allotransplantation in the United States would be a game-changing event in the treatment of type 1 diabetes and also generate enthusiasm for continued research.
Collapse
Affiliation(s)
- Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
- College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Michael F. Knoll
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Carmela A. Knoll
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Suzanne Bertera
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Massimo M. Trucco
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
- College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
36
|
Park H, Haque MR, Park JB, Lee KW, Lee S, Kwon Y, Lee HS, Kim GS, Shin DY, Jin SM, Kim JH, Kang HJ, Byun Y, Kim SJ. Polymeric nano-shielded islets with heparin-polyethylene glycol in a non-human primate model. Biomaterials 2018; 171:164-177. [PMID: 29698867 DOI: 10.1016/j.biomaterials.2018.04.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 10/17/2022]
Abstract
Intraportal pancreatic islet transplantation incurs huge cell losses during its early stages due to instant blood-mediated inflammatory reactions (IBMIRs), which may also drive regulation of the adaptive immune system. Therefore, a method that evades IBMIR will improve clinical islet transplantation. We used a layer-by-layer approach to shield non-human primate (NHP) islets with polyethylene glycol (nano-shielded islets, NSIs) and polyethylene glycol plus heparin (heparin nano-shielded islets; HNSIs). Islets ranging from 10,000 to 20,000 IEQ/kg body weight were transplanted into 19 cynomolgus monkeys (n = 4, control; n = 5, NSI; and n = 10, HNSI). The mean C-peptide positive graft survival times were 68.5, 64 and 108 days for the control, NSI and HNSI groups, respectively (P = 0.012). HNSI also reduced the factors responsible for IBMIR in vitro. Based on these data, HNSIs in conjunction with clinically established immunosuppressive drug regimens will result in superior outcomes compared to those achieved with the current protocol for clinical islet transplantation.
Collapse
Affiliation(s)
- Hyojun Park
- Department of Surgery, VHS Medical Center, Seoul 05368, Republic of Korea
| | - Muhammad R Haque
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Berm Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyo Won Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Sanghoon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Yeongbeen Kwon
- Transplantation Research Center, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Han Sin Lee
- Transplantation Research Center, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Geun-Soo Kim
- Transplantation Research Center, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Du Yeon Shin
- Transplantation Research Center, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Sang-Man Jin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jae Hyeon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Hee Jung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang-si, Republic of Korea
| | - Youngro Byun
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea.
| | - Sung Joo Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; Transplantation Research Center, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea.
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Porcine islets are being extensively investigated as alternative sources of insulin-secreting cells for transplantation in insulin-dependent diabetic patients. The present review focuses on recent advances in porcine islet transplantation with particular emphasis on new transgenic pig models, islet encapsulation, and biosafety considerations. RECENT FINDINGS Genetic modifications aimed to reduce islet cell immunogenicity, to prolong their survival, and to improve their secretory function have been reported. Micro- and macroencapsulation of porcine islets should allow their use in the clinic with no or minimal immunosuppression. The risk of porcine endogenous retrovirus transmission is being re-evaluated since no evidence for infection was found in several clinical and preclinical studies. SUMMARY Pig islet xenotransplantation is still a serious contestant in the race for novel treatments for type I diabetes. Adequate pathogen screening, animal selection, and the establishment of microbiological, genetic, and potency release quality controls should increase safety and efficacy of future porcine islets transplantation clinical trials.
Collapse
|
38
|
Kim H, Kim H, Lee SK, Jin XL, Kim TJ, Park C, Lee JI, Kim HS, Hong SK, Yoon KC, Ahn SW, Lee KB, Yi NJ, Yang J, Lee KW, Hawthorne WJ, Suh KS. Memory T cells are significantly increased in rejected liver allografts of rhesus monkeys. Liver Transpl 2018; 24:256-268. [PMID: 29150986 PMCID: PMC5817407 DOI: 10.1002/lt.24983] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/18/2017] [Accepted: 10/29/2017] [Indexed: 12/17/2022]
Abstract
The rhesus monkey (RM) is an excellent preclinical model in kidney, heart, and islet transplantation that has provided the basis for new immunosuppressive protocols for clinical studies. However, there remain relatively few liver transplantation (LT) models in nonhuman primates. In this study, we analyzed the immune cell populations of peripheral blood mononuclear cells (PBMCs) and secondary lymphoid organs along with livers of normal RMs and compared them with those of rejected LT recipients following withdrawal of immunosuppression. We undertook 5 allogeneic ABO compatible orthotopic LTs in monkeys using 5 normal donor monkey livers. We collected tissues including lymph nodes, spleens, blood, and recipient livers, and we performed flow cytometric analysis using isolated immune cells. We found that CD4 or CD8 naïve T cells were normally seen at low levels, and memory T cells were seen at high levels in the liver rather than lymphoid organs or PBMC. However, regulatory cells such as CD4+ forkhead box P3+ T cells and CD8+ CD28- cells remained in high numbers in the liver, but not in the lymph nodes or PBMC. The comparison of CD4/8 T subpopulations in normal and rejected livers and the various tissues showed that naïve cells were dramatically decreased in the spleen, lymph node, and PBMCs of rejected LT monkeys, but rather, the memory CD4/8 T cells were increased in all tissues and PBMC. The normal liver has large numbers of CD4 regulatory T cells, CD8+ CD28-, and myeloid-derived suppressor cells, which are known immunosuppressive cells occurring at much higher levels than those seen in lymph node or peripheral blood. Memory T cells are dramatically increased in rejected liver allografts of RMs compared with those seen in normal RM tissues. Liver Transplantation 24 256-268 2018 AASLD.
Collapse
Affiliation(s)
- Hwajung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyeyoung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Sun-Kyung Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Xue-Li Jin
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae Jin Kim
- Division of Immunobiology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Chanho Park
- Division of Immunobiology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Jae-Il Lee
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyo-Sin Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Suk Kyun Hong
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyung Chul Yoon
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Sung Woo Ahn
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyoung-Bun Lee
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Nam-Joon Yi
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Jaeseok Yang
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea.,Transplantation Center, Seoul National University Hospital, Seoul, South Korea
| | - Kwang-Woong Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Wayne J Hawthorne
- Department of Surgery, University of Sydney at Westmead Hospital, Westmead, New South Wales, Australia
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
39
|
Wright K, Dziuk R, Mital P, Kaur G, Dufour JM. Xenotransplanted Pig Sertoli Cells Inhibit Both the Alternative and Classical Pathways of Complement-Mediated Cell Lysis While Pig Islets Are Killed. Cell Transplant 2018; 25:2027-2040. [PMID: 27305664 DOI: 10.3727/096368916x692032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Xenotransplantation has vast clinical potential but is limited by the potent immune responses generated against xenogeneic tissue. Immune-privileged Sertoli cells (SCs) survive xenotransplantation long term (≥90 days) without immunosuppression, making SCs an ideal model to identify xenograft survival mechanisms. Xenograft rejection includes the binding of natural and induced antibodies and the activation of the complement cascade. Using an in vitro cytotoxicity assay, wherein cells were cultured with human serum and complement, we demonstrated that neonatal pig SCs (NPSCs) are resistant to complement-mediated cell lysis and express complement inhibitory factors, membrane cofactor protein (MCP; CD46), and decay- accelerating factor (DAF; CD55) at significantly higher levels than neonatal pig islets (NPIs), which served as non-immune-privileged controls. After xenotransplantation into naive Lewis rats, NPSCs survived throughout the study, while NPIs were rejected within 9 days. Serum antibodies, and antibody and complement deposition within the grafts were analyzed. Compared to preformed circulating anti-pig IgM antibodies, no significant increase in IgM production against NPSCs or NPIs was observed, while IgM deposition was detected from day 6 onward in both sets of grafts. A late serum IgG response was detected in NPSC (days 13 and 20) and NPI (day 20) recipients. Consistently, IgG deposition was first detected at days 9 and 13 in NPSC and NPI grafts, respectively. Interestingly, C3 was deposited at days 1 and 3 in NPI grafts and only at day 1 in NPSC grafts, while membrane attack complex (MAC) deposition was only detected in NPI grafts (at days 1-4). Collectively, these data suggest NPSCs actively inhibit both the alternative and classical pathways of complement-mediated cell lysis, while the alternative pathway plays a role in rejecting NPIs. Ultimately, inhibiting the alternative pathway along with transplanting xenogeneic tissue from transgenic pigs (expressing human complement inhibitory factors) could prolong the survival of xenogeneic cells without immunosuppression.
Collapse
Affiliation(s)
- Kandis Wright
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Rachel Dziuk
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Payal Mital
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Gurvinder Kaur
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jannette M Dufour
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
40
|
Zhu H, Zhang X, He Y, Yu L, Lü Y, Pan K, Wang B, Chen G. [Research progress on the donor cell sources of pancreatic islet transplantation for treatment of diabetes mellitus]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:104-111. [PMID: 29806374 PMCID: PMC8414200 DOI: 10.7507/1002-1892.201707049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 12/13/2017] [Indexed: 11/03/2022]
Abstract
Objective To summarize the research progress on the source and selection of donor cells in the field of islet replacement therapy for diabetes mellitus. Methods Domestic and abroad literature concerning islet replacement therapy for diabetes mellitus, as well as donor source and donor selection was reviewed and analyzed thoroughly. Results The shortage of donor supply is still a major obstacle for the widely clinical application of pancreatic islet transplantation (PIT). Currently, in addition to the progress on the allogeneic/autologous donor islet supply, some remarkable achievements have been also attained in the application of xenogeneic islet (from pig donor), as well as islet like cells derived from stem cells and islet cell line, potentially enlarging the source of implantable cells. Conclusion Adequate and suitable donor cell supply is an essential prerequisite for widely clinical application of PIT therapy for type 1 diabetes mellitus (T1DM). Further perfection of organ donation system, together with development of immune-tolerance induction, gene and bioengineering technology etc. will possibly solve the problem of donor cell shortage and provide a basis for clinical application of cellular replacement therapy for T1DM.
Collapse
Affiliation(s)
- Haitao Zhu
- Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061, P.R.China;Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Xiaoge Zhang
- Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061, P.R.China
| | - Yayi He
- Department of Endocrinology, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Liang Yu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Yi Lü
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China;Research Institute of Advanced Surgical Technology and Engineering, Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Kaili Pan
- Department of Pediatrics (No. 2 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061, P.R.China
| | - Bo Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China;Department of Endocrinology, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061,
| | - Guoqiang Chen
- Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061,
| |
Collapse
|
41
|
Meier RPH, Muller YD, Balaphas A, Morel P, Pascual M, Seebach JD, Buhler LH. Xenotransplantation: back to the future? Transpl Int 2018; 31:465-477. [PMID: 29210109 DOI: 10.1111/tri.13104] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/05/2017] [Accepted: 11/26/2017] [Indexed: 12/26/2022]
Abstract
The field of xenotransplantation has fluctuated between great optimism and doubts over the last 50 years. The initial clinical attempts were extremely ambitious but faced technical and ethical issues that prompted the research community to go back to preclinical studies. Important players left the field due to perceived xenozoonotic risks and the lack of progress in pig-to-nonhuman-primate transplant models. Initial apparently unsurmountable issues appear now to be possible to overcome due to progress of genetic engineering, allowing the generation of multiple-xenoantigen knockout pigs that express human transgenes and the genomewide inactivation of porcine endogenous retroviruses. These important steps forward were made possible by new genome editing technologies, such as CRISPR/Cas9, allowing researchers to precisely remove or insert genes anywhere in the genome. An additional emerging perspective is the possibility of growing humanized organs in pigs using blastocyst complementation. This article summarizes the current advances in xenotransplantation research in nonhuman primates, and it describes the newly developed genome editing technology tools and interspecific organ generation.
Collapse
Affiliation(s)
- Raphael P H Meier
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | - Yannick D Muller
- Division of Clinical Immunology and Allergy, Department of Medical Specialties, University Hospitals and Medical Faculty, Geneva, Switzerland.,Transplantation Center, Lausanne University Hospital, Lausanne, Switzerland
| | - Alexandre Balaphas
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | - Philippe Morel
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | - Manuel Pascual
- Transplantation Center, Lausanne University Hospital, Lausanne, Switzerland
| | - Jörg D Seebach
- Division of Clinical Immunology and Allergy, Department of Medical Specialties, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Leo H Buhler
- Visceral and Transplant Surgery, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
42
|
Cooper DKC, Cowan P, Fishman JA, Hering BJ, Mohiuddin MM, Pierson RN, Sachs DH, Schuurman HJ, Dennis JU, Tönjes RR. Joint FDA‐IXA Symposium, September 20, 2017. Xenotransplantation 2017; 24. [PMID: 29193342 DOI: 10.1111/xen.12365] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Peter Cowan
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Jay A Fishman
- Infectious Disease Division and MGH Transplant Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bernhard J Hering
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Muhammad M Mohiuddin
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard N Pierson
- Department of Surgery, University of Maryland School of Medicine, Baltimore VA Medical Center, Baltimore, MD, USA
| | - David H Sachs
- Columbia University Medical Center, New York City, NY, USA.,Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | | | - John U Dennis
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ralf R Tönjes
- Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Division of Medical Biotechnology, Langen, Germany
| |
Collapse
|
43
|
Cowan PJ, Tector AJ. The Resurgence of Xenotransplantation. Am J Transplant 2017; 17:2531-2536. [PMID: 28397351 DOI: 10.1111/ajt.14311] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/21/2017] [Accepted: 04/01/2017] [Indexed: 01/25/2023]
Abstract
There has been an upsurge of interest in xenotransplantation in recent years. This resurgence can attributed to a combination of factors. First, there has been a dramatic improvement in efficacy in several preclinical models, with maximum xenograft survival times increasing to 950 days for islets, 945 days for hearts, and 310 days for kidneys. Second, the rapid development of genome editing technology (particularly the advent of clustered regularly interspaced short palindromic repeats/Cas9) has revolutionized the capacity to generate new donor pigs with multiple protective genetic modifications; what once took many years to achieve can now be performed in months, with much greater precision and scope. Third, the specter of porcine endogenous retrovirus (PERV) has receded significantly. There has been no evidence of PERV transmission in clinical trials and preclinical models, and improved screening methods and new options for the treatment or even elimination of PERV are now available. Balancing these positive developments are several remaining challenges, notably the heavy and often clinically inapplicable immunosuppression required to prevent xenograft rejection. Nonetheless, the potential for xenotransplantation as a solution to the shortage of human organs and tissues for transplantation continues to grow.
Collapse
Affiliation(s)
- P J Cowan
- Immunology Research Centre, St Vincent's Hospital Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia
| | - A J Tector
- School of Medicine, University of Alabama, Birmingham, AL
| |
Collapse
|
44
|
In Vivo Costimulation Blockade-Induced Regulatory T Cells Demonstrate Dominant and Specific Tolerance to Porcine Islet Xenografts. Transplantation 2017; 101:1587-1599. [PMID: 27653300 DOI: 10.1097/tp.0000000000001482] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Although islet xenotransplantation is a promising therapy for type 1 diabetes, its clinical application has been hampered by cellular rejection and the requirement for high levels of immunosuppression. The aim of this study was to determine the role of Foxp3 regulatory T (Treg) cells in costimulation blockade-induced dominant tolerance to porcine neonatal islet cell cluster (NICC) xenografts in mice. METHODS Porcine-NICC were transplanted under the renal capsule of BALB/c or C57BL/6 recipients and given a single dose of CTLA4-Fc at the time of transplant and 4doses of anti-CD154 mAb to day 6. Depletion of Foxp3Treg cell was performed in DEpletion of REGulatory T cells mice at day 80 posttransplantation. Foxp3Treg cell from spleens of treated BALB/c mice (tolerant Treg cell), and splenocytes were cotransferred into islet transplanted nonobese diabetic background with severe combined immunodeficiency mice to assess suppressive function. RESULTS In treated mice, increased numbers of Foxp3Treg cell were identified in the porcine-NICC xenografts, draining lymph node, and spleen. Porcine-NICC xenografts from treated mice expressed elevated levels of TGF-β, IL-10 and IFN-γ. Porcine-NICC xenograft tolerance was abrogated after depletion of Foxp3Treg cell. Tolerant Treg cell produced high levels of IL-10 and had diverse T cell receptor Vβ repertoires with an oligoclonal expansion in CDR3 of T cell receptor Vβ14. These tolerant Treg cells had the capacity to transfer dominant tolerance and specifically exhibited more potent regulatory function to porcine-NICC xenografts that naive Treg cell. CONCLUSIONS This study demonstrated that short-term costimulation blockade-induced dominant tolerance and that Foxp3Treg cell played an essential role in its maintenance. Foxp3Treg cells were activated and had more potent regulatory function in vivo than naive Treg cells.
Collapse
|
45
|
Nottle MB, Salvaris EJ, Fisicaro N, McIlfatrick S, Vassiliev I, Hawthorne WJ, O'Connell PJ, Brady JL, Lew AM, Cowan PJ. Targeted insertion of an anti-CD2 monoclonal antibody transgene into the GGTA1 locus in pigs using FokI-dCas9. Sci Rep 2017; 7:8383. [PMID: 28814758 PMCID: PMC5559588 DOI: 10.1038/s41598-017-09030-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/17/2017] [Indexed: 12/21/2022] Open
Abstract
Xenotransplantation from pigs has been advocated as a solution to the perennial shortage of donated human organs and tissues. CRISPR/Cas9 has facilitated the silencing of genes in donor pigs that contribute to xenograft rejection. However, the generation of modified pigs using second-generation nucleases with much lower off-target mutation rates than Cas9, such as FokI-dCas9, has not been reported. Furthermore, there have been no reports on the use of CRISPR to knock protective transgenes into detrimental porcine genes. In this study, we used FokI-dCas9 with two guide RNAs to integrate a 7.1 kilobase pair transgene into exon 9 of the GGTA1 gene in porcine fetal fibroblasts. The modified cells lacked expression of the αGal xenoantigen, and secreted an anti-CD2 monoclonal antibody encoded by the transgene. PCR and sequencing revealed precise integration of the transgene into one allele of GGTA1, and a small deletion in the second allele. The cells were used for somatic cell nuclear transfer to generate healthy male knock-in piglets, which did not express αGal and which contained anti-CD2 in their serum. We have therefore developed a versatile high-fidelity system for knocking transgenes into the pig genome for xenotransplantation purposes.
Collapse
Affiliation(s)
- Mark B Nottle
- Robinson Research Institute & Adelaide School of Medicine, University of Adelaide, Adelaide, Australia
| | - Evelyn J Salvaris
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
| | - Nella Fisicaro
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
| | - Stephen McIlfatrick
- Robinson Research Institute & Adelaide School of Medicine, University of Adelaide, Adelaide, Australia
| | - Ivan Vassiliev
- Robinson Research Institute & Adelaide School of Medicine, University of Adelaide, Adelaide, Australia
| | - Wayne J Hawthorne
- Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | | | - Jamie L Brady
- Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
| | - Andrew M Lew
- Walter and Eliza Hall Institute, Melbourne, Victoria, Australia.,Department of Microbiology & Immunology, University of Melbourne, Victoria, Australia
| | - Peter J Cowan
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Melbourne, Victoria, Australia. .,Department of Medicine, University of Melbourne, Victoria, Australia.
| |
Collapse
|
46
|
Mourad NI, Gianello P. Gene Editing, Gene Therapy, and Cell Xenotransplantation: Cell Transplantation Across Species. CURRENT TRANSPLANTATION REPORTS 2017; 4:193-200. [PMID: 28932650 PMCID: PMC5577055 DOI: 10.1007/s40472-017-0157-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Cell xenotransplantation has the potential to provide a safe, ethically acceptable, unlimited source for cell replacement therapies. This review focuses on genetic modification strategies aimed to overcome remaining hurdles standing in the way of clinical porcine islet transplantation and to develop neural cell xenotransplantation. RECENT FINDINGS In addition to previously described genetic modifications aimed to mitigate hyperacute rejection, instant blood-mediated inflammatory reaction, and cell-mediated rejection, new data showing the possibility of increasing porcine islet insulin secretion by transgenesis is an interesting addition to the array of genetically modified pigs available for xenotransplantation. Moreover, combining multiple modifications is possible today thanks to new, improved genomic editing tools. SUMMARY Genetic modification of large animals, pigs in particular, has come a long way during the last decade. These modifications can help minimize immunological and physiological incompatibilities between porcine and human cells, thus allowing for better tolerance and function of xenocells.
Collapse
Affiliation(s)
- Nizar I. Mourad
- Pôle de chirurgie expérimentale et transplantation, Université catholique de Louvain, SSS/IREC/CHEX, Avenue Hippocrate, 55 – Bte B1.55.04, 1200 Brussels, Belgium
| | - Pierre Gianello
- Pôle de chirurgie expérimentale et transplantation, Université catholique de Louvain, SSS/IREC/CHEX, Avenue Hippocrate, 55 – Bte B1.55.04, 1200 Brussels, Belgium
| |
Collapse
|
47
|
Liu Z, Hu W, He T, Dai Y, Hara H, Bottino R, Cooper DKC, Cai Z, Mou L. Pig-to-Primate Islet Xenotransplantation: Past, Present, and Future. Cell Transplant 2017; 26:925-947. [PMID: 28155815 PMCID: PMC5657750 DOI: 10.3727/096368917x694859] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 03/21/2017] [Indexed: 12/17/2022] Open
Abstract
Islet allotransplantation results in increasing success in treating type 1 diabetes, but the shortage of deceased human donor pancreata limits progress. Islet xenotransplantation, using pigs as a source of islets, is a promising approach to overcome this limitation. The greatest obstacle is the primate immune/inflammatory response to the porcine (pig) islets, which may take the form of rapid early graft rejection (the instant blood-mediated inflammatory reaction) or T-cell-mediated rejection. These problems are being resolved by the genetic engineering of the source pigs combined with improved immunosuppressive therapy. The results of pig-to-diabetic nonhuman primate islet xenotransplantation are steadily improving, with insulin independence being achieved for periods >1 year. An alternative approach is to isolate islets within a micro- or macroencapsulation device aimed at protecting them from the human recipient's immune response. Clinical trials using this approach are currently underway. This review focuses on the major aspects of pig-to-primate islet xenotransplantation and its potential for treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Zhengzhao Liu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Wenbao Hu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Tian He
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Hidetaka Hara
- Xenotransplantation Program/Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rita Bottino
- Institute for Cellular Therapeutics, Allegheny-Singer Research Institute, Pittsburgh, PA, USA
| | - David K. C. Cooper
- Xenotransplantation Program/Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhiming Cai
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| |
Collapse
|
48
|
Executive Summary of IPITA-TTS Opinion Leaders Report on the Future of β-Cell Replacement. Transplantation 2017; 100:e25-31. [PMID: 27082827 DOI: 10.1097/tp.0000000000001054] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The International Pancreas and Islet Transplant Association (IPITA), in conjunction with the Transplantation Society (TTS), convened a workshop to consider the future of pancreas and islet transplantation in the context of potential competing technologies that are under development, including the artificial pancreas, transplantation tolerance, xenotransplantation, encapsulation, stem cell derived beta cells, beta cell proliferation, and endogenous regeneration. Separate workgroups for each topic and then the collective group reviewed the state of the art, hurdles to application, and proposed research agenda for each therapy that would allow widespread application. Herein we present the executive summary of this workshop that focuses on obstacles to application and the research agenda to overcome them; the full length article with detailed background for each topic is published as an online supplement to Transplantation.
Collapse
|
49
|
Abstract
Type 1 diabetes is an autoimmune disorder in which the immune system attacks and destroys insulin-producing islet cells of the pancreas. Although islet transplantation has proved to be successful for some patients with type 1 diabetes, its widespread use is limited by islet donor shortage and the requirement for lifelong immunosuppression. An encapsulation strategy that can prevent the rejection of xenogeneic islets or of stem cell-derived allogeneic islets can potentially eliminate both of these barriers. Although encapsulation technology has met several challenges, the convergence of expertise in materials, nanotechnology, stem cell biology and immunology is allowing us to get closer to the goal of encapsulated islet cell therapy for humans.
Collapse
Affiliation(s)
- Tejal Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, Byers Hall Rm 203C, MC 2520, 1700 4th Street, San Francisco, California 94158-2330, USA
| | - Lonnie D Shea
- University of Michigan, Department of Biomedical Engineering, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109-2099, USA
| |
Collapse
|
50
|
Kang HJ, Lee H, Park EM, Kim JM, Min BH, Park CG. D-dimer level, in association with humoral responses, negatively correlates with survival of porcine islet grafts in non-human primates with immunosuppression. Xenotransplantation 2017; 24. [DOI: 10.1111/xen.12299] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/15/2017] [Accepted: 03/01/2017] [Indexed: 01/09/2023]
Affiliation(s)
- Hee Jung Kang
- Department of Laboratory Medicine; Hallym University College of Medicine; Anyang-si Korea
| | - Haneulnari Lee
- Department of Laboratory Medicine; Hallym University College of Medicine; Anyang-si Korea
| | - Eun Mi Park
- Department of Laboratory Medicine; Hallym University College of Medicine; Anyang-si Korea
| | - Jong-Min Kim
- Xenotransplantation Research Center; Seoul National University College of Medicine; Seoul Korea
| | - Byoung-Hoon Min
- Xenotransplantation Research Center; Seoul National University College of Medicine; Seoul Korea
| | - Chung-Gyu Park
- Xenotransplantation Research Center; Seoul National University College of Medicine; Seoul Korea
- Department of Microbiology and Immunology; Department of Biomedical Sciences; Cancer Research Institute; Institute of Endemic Diseases; Seoul National University College of Medicine; Seoul Korea
| |
Collapse
|